1
|
Xu J, Wang B, Zhao Z, Wu S, Zhang Z, Liu S, Huo N, Zheng W, Chen Y, Gao Z, Jia Z, Liu T, Zhu L, Hou L. Development of a novel adenovirus type 4 vector as a promising respiratory vaccine vehicle. Front Immunol 2025; 16:1572081. [PMID: 40276512 PMCID: PMC12018414 DOI: 10.3389/fimmu.2025.1572081] [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: 02/06/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Adenovirus (Ad) vectors are widely used for gene delivery, and some of them have been approved for vaccine development. In particular, the recombinant COVID-19 vaccine for inhalation, which was developed using adenovirus type 5 (Ad5), represents a milestone in respiratory immunization. Owing to the high pre-existing immunity (PEI) to Ad5, the development of an adenoviral vector with lower PEI and higher immunogenicity has been explored. However, the majority of the developed novel Ad vectors showed suboptimal immunogenicity compared to Ad5 in animal models. Method In this study, we constructed a novel replication-deficient viral vector based on human adenovirus type 4 (Ad4), which has long been used as a live virus vaccine with a favorable safety profile in the U.S. military. The mice were immunized intramuscularly or intranasally with an Ad4-vectored vaccine to verify immune responses and protective efficacy. Results Compared with Ad5, the novel Ad4 vector showed comparable viral growth kinetics and transgene expression in cells and similar exogenous protein expression and distribution in mice. Furthermore, the Ad4-vectored vaccine elicited superior humoral and cellular responses and protective effects when vaccinated intranasally than those triggered by the Ad5-vectored vaccine. Finally, the heterologous Ad5 prime and Ad4 boost immunization showed better immunogenicity and protective efficacy. Discussion This study broadens the research trajectory of adenovirus-vectored vaccines and offers a new option for the development of recombinant viral-vectored vaccines.
Collapse
Affiliation(s)
| | - Busen Wang
- *Correspondence: Lihua Hou, ; Li Zhu, ; Busen Wang,
| | | | | | | | | | | | | | | | | | | | | | - Li Zhu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology,
Beijing, China
| | - Lihua Hou
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology,
Beijing, China
| |
Collapse
|
2
|
Nieto-Ponce M, Reyna-Rosas E, Palencia-Reyes RA, Blancas-Ruíz C, Aguilar-Rafael G, Rubio-Diaz MPR, Ramírez-Martínez LA, Carranza C, Lozano-Dubernard B, Torres M, Zamudio-Meza H. The Validation of a Hemagglutination Inhibition Assay That Detects Antibodies Against a Newcastle Disease Virus-Based Vaccine Vector in Human Serum Samples. Vaccines (Basel) 2025; 13:342. [PMID: 40333216 PMCID: PMC12031337 DOI: 10.3390/vaccines13040342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND An NDV-based vector has been used as a veterinary vaccine and, recently, as a human COVID-19 vaccine. However, data for the potential immune response against the vector in humans are scarce; therefore, it is important to evaluate the levels of antibodies produced. The HI assay is the gold standard for assessing the humoral response against NDV in poultry serum. OBJECTIVE Here, the objective was to validate the HI assay against the NDV-vectored vaccine to analyze antibodies in human serum. METHODS First, we standardized the conditions in human sera before validation. RESULTS The results for analytical performance in terms of selectivity, sensitivity, specificity, and positive and negative predictive values, as well as positive and negative diagnostic reliability, indicate that the assay is highly selective, allowing clear discrimination between positive and negative samples. Regarding repeatability and intermediate precision, we demonstrated that the assay has the precision to obtain consistent results, guaranteeing their reliability and truthfulness. Finally, the results regarding accuracy, linearity, and robustness indicate that the assay is accurate across the evaluated concentration intervals, with a linear correlation between low and high levels, and demonstrate that it is robust and consistent when serum-antigen interaction times are changed. CONCLUSIONS We conclude that the suitability of the analytical method for its intended use is confirmed, guaranteeing the reliability of the results obtained under the established operating conditions.
Collapse
Affiliation(s)
- Milton Nieto-Ponce
- Laboratorio de Inmunobiología de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City CP 14080, Mexico; (M.N.-P.); (E.R.-R.); (R.A.P.-R.); (C.B.-R.); (C.C.)
| | - Edgar Reyna-Rosas
- Laboratorio de Inmunobiología de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City CP 14080, Mexico; (M.N.-P.); (E.R.-R.); (R.A.P.-R.); (C.B.-R.); (C.C.)
| | - Rosa Andrea Palencia-Reyes
- Laboratorio de Inmunobiología de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City CP 14080, Mexico; (M.N.-P.); (E.R.-R.); (R.A.P.-R.); (C.B.-R.); (C.C.)
| | - Carlos Blancas-Ruíz
- Laboratorio de Inmunobiología de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City CP 14080, Mexico; (M.N.-P.); (E.R.-R.); (R.A.P.-R.); (C.B.-R.); (C.C.)
| | - Guadalupe Aguilar-Rafael
- Diagnósticos Clínicos Veterinarios, S.A. de C.V. (DCV), Mexico City CP 09810, Mexico; (G.A.-R.); (M.P.R.R.-D.)
| | | | | | - Claudia Carranza
- Laboratorio de Inmunobiología de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City CP 14080, Mexico; (M.N.-P.); (E.R.-R.); (R.A.P.-R.); (C.B.-R.); (C.C.)
| | | | - Martha Torres
- Laboratorio de Inmunobiología de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City CP 14080, Mexico; (M.N.-P.); (E.R.-R.); (R.A.P.-R.); (C.B.-R.); (C.C.)
| | - Horacio Zamudio-Meza
- Laboratorio de Inmunobiología de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City CP 14080, Mexico; (M.N.-P.); (E.R.-R.); (R.A.P.-R.); (C.B.-R.); (C.C.)
| |
Collapse
|
3
|
Wallace R, Bliss CM, Parker AL. The Immune System-A Double-Edged Sword for Adenovirus-Based Therapies. Viruses 2024; 16:973. [PMID: 38932265 PMCID: PMC11209478 DOI: 10.3390/v16060973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Pathogenic adenovirus (Ad) infections are widespread but typically mild and transient, except in the immunocompromised. As vectors for gene therapy, vaccine, and oncology applications, Ad-based platforms offer advantages, including ease of genetic manipulation, scale of production, and well-established safety profiles, making them attractive tools for therapeutic development. However, the immune system often poses a significant challenge that must be overcome for adenovirus-based therapies to be truly efficacious. Both pre-existing anti-Ad immunity in the population as well as the rapid development of an immune response against engineered adenoviral vectors can have detrimental effects on the downstream impact of an adenovirus-based therapeutic. This review focuses on the different challenges posed, including pre-existing natural immunity and anti-vector immunity induced by a therapeutic, in the context of innate and adaptive immune responses. We summarise different approaches developed with the aim of tackling these problems, as well as their outcomes and potential future applications.
Collapse
Affiliation(s)
- Rebecca Wallace
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
| | - Carly M. Bliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Alan L. Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
4
|
Amosova IV, Timoshicheva TA, Kadyrova RA, Zabrodskaya YA, Vakin VS, Grudinin MP, Dzytseva VV, Khmelevsky MS, Lioznov DA. The investigation of the dynamics of changes in neutralizing antibody titers against type 5 adenovirus in the context of vaccination against a new coronavirus infection. Virology 2024; 594:110051. [PMID: 38489915 DOI: 10.1016/j.virol.2024.110051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/27/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
This research focuses on analyzing the dynamics of neutralizing antibody (nAbs) titers against type 5 adenovirus (Ad5) in the adult population of Russia following vaccination against the novel coronavirus infection with recombinant adenovirus type-5 COVID-19 vaccine (CanSino Biologics, China). The impact of the Ad5 vector on nAb titers was investigated using 302 blood serum samples from individuals who received a single dose of the Ad5-nCoV vector vaccine. The research revealed that 33.8% of adults in Russia had pre-existing anti-Ad5 nAbs before the pandemic. Notably, 40% of vaccinated individuals did not exhibit an increase in nAbs titers upon receiving the Ad5-based vaccine. However, in the group with no or low titers of anti-Ad5 nAbs (1:10-1:40), a significant 8-16-fold increase in nAb titers to Ad5 was observed.
Collapse
Affiliation(s)
- I V Amosova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg, 197376, Russia
| | - T A Timoshicheva
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg, 197376, Russia.
| | - R A Kadyrova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg, 197376, Russia
| | - Y A Zabrodskaya
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg, 197376, Russia; Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, St. Petersburg, 194064, Russia
| | - V S Vakin
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg, 197376, Russia
| | - M P Grudinin
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg, 197376, Russia
| | - V V Dzytseva
- NPO Petrovax Pharm LLC, 12 Presnenskaya Embankment, Moscow, 123112, Russia
| | - M S Khmelevsky
- NPO Petrovax Pharm LLC, 12 Presnenskaya Embankment, Moscow, 123112, Russia
| | - D A Lioznov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg, 197376, Russia; Pavlov First Saint Petersburg State Medical University, 197022, L'va Tolstogo St. 6-8, St. Petersburg, Russia
| |
Collapse
|
5
|
Li X, Le Y, Zhang Z, Nian X, Liu B, Yang X. Viral Vector-Based Gene Therapy. Int J Mol Sci 2023; 24:ijms24097736. [PMID: 37175441 PMCID: PMC10177981 DOI: 10.3390/ijms24097736] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Gene therapy is a technique involving the modification of an individual's genes for treating a particular disease. The key to effective gene therapy is an efficient carrier delivery system. Viral vectors that have been artificially modified to lose their pathogenicity are used widely as a delivery system, with the key advantages of their natural high transduction efficiency and stable expression. With decades of development, viral vector-based gene therapies have achieved promising clinical outcomes. Currently, the three key vector strategies are based on adeno-associated viruses, adenoviruses, and lentiviruses. However, certain challenges, such as immunotoxicity and "off-target", continue to exist. In the present review, the above three viral vectors are discussed along with their respective therapeutic applications. In addition, the major translational challenges encountered in viral vector-based gene therapies are summarized, and the possible strategies to address these challenges are also discussed.
Collapse
Affiliation(s)
- Xuedan Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Yang Le
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Zhegang Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Bo Liu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- China National Biotech Group Company Limited, Beijing 100029, China
| |
Collapse
|
6
|
Anti-Ad26 humoral immunity does not compromise SARS-COV-2 neutralizing antibody responses following Gam-COVID-Vac booster vaccination. NPJ Vaccines 2022; 7:145. [DOI: 10.1038/s41541-022-00566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
AbstractReplication-incompetent adenoviral vectors have been extensively used as a platform for vaccine design, with at least four anti-COVID-19 vaccines authorized to date. These vaccines elicit neutralizing antibody responses directed against SARS-CoV-2 Spike protein and confer significant level of protection against SARS-CoV-2 infection. Immunization with adenovirus-vectored vaccines is known to be accompanied by the production of anti-vector antibodies, which may translate into reduced efficacy of booster or repeated rounds of revaccination. Here, we used blood samples from patients who received an adenovirus-based Gam-COVID-Vac vaccine to address the question of whether anti-vector antibodies may influence the magnitude of SARS-CoV-2-specific humoral response after booster vaccination. We observed that rAd26-based prime vaccination with Gam-COVID-Vac induced the development of Ad26-neutralizing antibodies, which persisted in circulation for at least 9 months. Our analysis further indicates that high pre-boost Ad26 neutralizing antibody titers do not appear to affect the humoral immunogenicity of the Gam-COVID-Vac boost. The titers of anti-SARS-CoV-2 RBD IgGs and antibodies, which neutralized both the wild type and the circulating variants of concern of SARS-CoV-2 such as Delta and Omicron, were independent of the pre-boost levels of Ad26-neutralizing antibodies. Thus, our results support the development of repeated immunization schedule with adenovirus-based COVID-19 vaccines.
Collapse
|
7
|
Chang J. Adenovirus Vectors: Excellent Tools for Vaccine Development. Immune Netw 2021; 21:e6. [PMID: 33728099 PMCID: PMC7937504 DOI: 10.4110/in.2021.21.e6] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/17/2021] [Indexed: 12/16/2022] Open
Abstract
Adenovirus was originally used as a vector for gene therapy. In recent years, with the development of the next-generation vectors with increased safety and high immunogenicity to transgene products, its utility as a vaccine vector has continued to increase. Adenovirus-based vaccines are currently being tested not only to prevent various infectious diseases but also to be applied as cancer vaccines. In this review, I discuss the innate and adaptive aspects of the immunological characteristics of adenovirus vectors and further examine the current status of advanced adenovirus-based vaccine development. Various methods that can overcome the limitations of currently used adenoviruses as vaccine vehicles are also discussed. Through this study, I hope that vaccine development using adenovirus vectors will be expedited and more successful.
Collapse
Affiliation(s)
- Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| |
Collapse
|
8
|
Adamaki M, Zoumpourlis V. Immunotherapy as a Precision Medicine Tool for the Treatment of Prostate Cancer. Cancers (Basel) 2021; 13:E173. [PMID: 33419051 PMCID: PMC7825410 DOI: 10.3390/cancers13020173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed type of cancer among Caucasian males over the age of 60 and is characterized by remarkable heterogeneity and clinical behavior, ranging from decades of indolence to highly lethal disease. Despite the significant progress in PCa systemic therapy, therapeutic response is usually transient, and invasive disease is associated with high mortality rates. Immunotherapy has emerged as an efficacious and non-toxic treatment alternative that perfectly fits the rationale of precision medicine, as it aims to treat patients on the basis of patient-specific, immune-targeted molecular traits, so as to achieve the maximum clinical benefit. Antibodies acting as immune checkpoint inhibitors and vaccines entailing tumor-specific antigens seem to be the most promising immunotherapeutic strategies in offering a significant survival advantage. Even though patients with localized disease and favorable prognostic characteristics seem to be the ones that markedly benefit from such interventions, there is substantial evidence to suggest that the survival benefit may also be extended to patients with more advanced disease. The identification of biomarkers that can be immunologically targeted in patients with disease progression is potentially amenable in this process and in achieving significant advances in the decision for precision treatment of PCa.
Collapse
Affiliation(s)
- Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
| | | |
Collapse
|
9
|
Manish M, Verma S, Kandari D, Kulshreshtha P, Singh S, Bhatnagar R. Anthrax prevention through vaccine and post-exposure therapy. Expert Opin Biol Ther 2020; 20:1405-1425. [DOI: 10.1080/14712598.2020.1801626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Manish Manish
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Shashikala Verma
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Divya Kandari
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Parul Kulshreshtha
- Department of Zoology, Shivaji College, University of Delhi, Delhi, India
| | - Samer Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- Department of Microbial Biotechnology, Panjab University, Chandigarh, India
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
10
|
Neukirch L, Fougeroux C, Andersson AMC, Holst PJ. The potential of adenoviral vaccine vectors with altered antigen presentation capabilities. Expert Rev Vaccines 2020; 19:25-41. [PMID: 31889453 DOI: 10.1080/14760584.2020.1711054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Despite their appeal as vaccine vectors, adenoviral vectors are yet unable to induce protective immune responses against some weakly immunogenic antigens. Additionally, the maximum doses of adenovirus-based vaccines are limited by vector-induced toxicity, causing vector elimination and diminished immune responses against the target antigen. In order to increase immune responses to the transgene, while maintaining a moderate vector dose, new technologies for improved transgene presentation have been developed for adenoviral vaccine vectors.Areas covered: This review provides an overview of different genetic-fusion adjuvants that aim to improve antigen presentation in the context of adenoviral vector-based vaccines. The influence on both T cell and B cell responses are discussed, with a main focus on two technologies: MHC class II-associated invariant chain and virus-like-vaccines.Expert opinion: Different strategies have been tested to improve adenovirus-based vaccinations with varying degrees of success. The reviewed genetic adjuvants were designed to increase antigen processing and MHC presentation, or promote humoral immune responses with an improved conformational antigen display. While none of the introduced technologies is universally applicable, this review shall give an overview to identify potential improvements for future vaccination approaches.
Collapse
Affiliation(s)
- Lasse Neukirch
- Clinical Cooperation Unit "Applied Tumor Immunity", National Center for Tumor Diseases and German Cancer Research Center, Heidelberg, Germany.,Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Cyrielle Fougeroux
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Marie Carola Andersson
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,InProTher ApS, Copenhagen, Denmark
| | - Peter Johannes Holst
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,InProTher ApS, Copenhagen, Denmark
| |
Collapse
|
11
|
Ababneh M, Alrwashdeh M, Khalifeh M. Recombinant adenoviral vaccine encoding the spike 1 subunit of the Middle East Respiratory Syndrome Coronavirus elicits strong humoral and cellular immune responses in mice. Vet World 2019; 12:1554-1562. [PMID: 31849416 PMCID: PMC6868266 DOI: 10.14202/vetworld.2019.1554-1562] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 08/26/2019] [Indexed: 01/05/2023] Open
Abstract
Background and Aim Middle East respiratory syndrome coronavirus (MERS-CoV) has rapidly spread throughout the Middle East since its discovery in 2012. The virus poses a significant global public health threat with potentially devastating effects. In this study, a recombinant adenoviral-based vaccine encoding the spike 1 (S1) subunit of the MERS-CoV genome was constructed, and its humoral, and cellular immune responses were evaluated in mice. Materials and Methods Mice were immunized initially by intramuscular injection and boosted 3 weeks later by intranasal application. Expression of the S1 protein in the lungs and kidneys was detected using conventional polymerase chain reaction (PCR) and immunohistochemistry (IHC) targeting specific regions within the S1 subunit at weeks 3, 4, 5, and 6 after the first vaccination. Antigen-specific humoral and cellular immune responses were evaluated in serum and in cell culture following in vitro stimulation with a specific 9-mer epitope within the S1 protein (CYSSLILDY). Results S1 protein expression was only detected by IHC in the kidneys of the Ad-MERS-S1 group at week 6 from first immunization, and in both lungs and kidneys of Ad-MERS-S1 group by conventional PCR at weeks 3 and 5 post-prime. The vaccine elicited a specific S1-immunoglobulin G antibody response, which was detected in the sera of the vaccinated mice at weeks 4 and 6 from the onset of the first immunization. There was a significant increase in the amount of Th1-related cytokines (interferon-γ and interleukin [IL] 12), and a significant decrease in the Th2-related cytokine IL-4 in splenocyte cell culture of the vaccinated group compared with the control groups. Conclusion The results of this study suggest that this recombinant adenovirus vaccine encoding the S1 subunit of MERS-CoV elicits potentially protective antigen-specific humoral and cellular immune responses in mice. This study demonstrates a promising vaccine for the control and/or prevention of MERS-CoV infection in humans.
Collapse
Affiliation(s)
- Mustafa Ababneh
- Department of Basic Medical Veterinary Sciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Mu'men Alrwashdeh
- Department of Basic Medical Veterinary Sciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Mohammad Khalifeh
- Department of Basic Medical Veterinary Sciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| |
Collapse
|
12
|
Wilmschen S, Schmitz JE, Kimpel J. Viral Vectors for the Induction of Broadly Neutralizing Antibodies against HIV. Vaccines (Basel) 2019; 7:vaccines7030119. [PMID: 31546894 PMCID: PMC6789710 DOI: 10.3390/vaccines7030119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 01/10/2023] Open
Abstract
Extensive research on generating an efficient HIV vaccine is ongoing. A major aim of HIV vaccines is the induction of long-lasting, broadly neutralizing antibodies (bnAbs) that can confer sterile immunity for a prolonged period of time. Several strategies have been explored to reach this goal, i.e. protein immunization, DNA, or viral vectors, or a combination thereof. In this review, we give an overview of approaches using viral vectors for the induction of HIV-specific bnAbs. Many pre-clinical studies were performed using various replication-competent and -incompetent vectors. Amongst them, poxviral and adenoviral vectors were the most prevalent ones. In many studies, viral vectors were combined with a DNA prime or a protein boost. However, neutralizing antibodies were mainly induced against the homologous HIV-1 vaccine strain or tier 1 viruses, and in rare cases, against tier 2 viruses, indicating the need for improved antigens and vaccination strategies. Furthermore, we also review next generation Env antigens that are currently being used in protein vaccination approaches and point out how they could be utilized in viral vectors.
Collapse
Affiliation(s)
- Sarah Wilmschen
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
13
|
Rahman MA, Robert-Guroff M. Accelerating HIV vaccine development using non-human primate models. Expert Rev Vaccines 2018; 18:61-73. [PMID: 30526159 DOI: 10.1080/14760584.2019.1557521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The search for a preventative HIV vaccine is ongoing after three decades of research. Contributions of non-human primate (NHP) models to this research are irrefutable, however interpreting data obtained for translation to humans has been problematic. As knowledge concerning NHP models has accumulated, their utility and value in assessing immunogenicity and efficacy of novel vaccines have become apparent. NHP models have become a critical component of vaccine design. AREAS COVERED Beginning with early vaccine studies, we trace the development and evolution of NHP models concurrent with changes in HIV vaccine concepts and in response to their ability to predict clinical trial efficacy. The value of NHP studies in guiding vaccine design is highlighted along with their importance in opening new areas of investigation and facilitating movement of promising approaches into the clinic. EXPERT COMMENTARY Due to their close relatedness to humans, NHPs are an excellent choice for immunogenicity studies. The ability of NHP models to predict clinical efficacy has improved with the introduction of low-dose challenge viruses and recognition of confounding variables in study outcomes. Use of NHP models has opened new research areas with outstanding potential for generating vaccine efficacy against HIV and other infectious agents.
Collapse
Affiliation(s)
- Mohammad Arif Rahman
- a Vaccine Branch, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Marjorie Robert-Guroff
- a Vaccine Branch, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
14
|
Jung SY, Kang KW, Lee EY, Seo DW, Kim HL, Kim H, Kwon T, Park HL, Kim H, Lee SM, Nam JH. Heterologous prime-boost vaccination with adenoviral vector and protein nanoparticles induces both Th1 and Th2 responses against Middle East respiratory syndrome coronavirus. Vaccine 2018; 36:3468-3476. [PMID: 29739720 PMCID: PMC7115429 DOI: 10.1016/j.vaccine.2018.04.082] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 12/20/2022]
Abstract
Immunization with MERS spike protein nanoparticles induced only Th2-biased response. Heterologous prime-boost immunization induced both Th1 and Th2-biased responses. Our vaccination strategy showed the protective effect against MERS-CoV.
The Middle East respiratory syndrome coronavirus (MERS-CoV) is a highly pathogenic and zoonotic virus with a fatality rate in humans of over 35%. Although several vaccine candidates have been developed, there is still no clinically available vaccine for MERS-CoV. In this study, we developed two types of MERS-CoV vaccines: a recombinant adenovirus serotype 5 encoding the MERS-CoV spike gene (Ad5/MERS) and spike protein nanoparticles formulated with aluminum (alum) adjuvant. Next, we tested a heterologous prime–boost vaccine strategy, which compared priming with Ad5/MERS and boosting with spike protein nanoparticles and vice versa, with homologous prime–boost vaccination comprising priming and boosting with either spike protein nanoparticles or Ad5/MERS. Although both types of vaccine could induce specific immunoglobulin G against MERS-CoV, neutralizing antibodies against MERS-CoV were induced only by heterologous prime–boost immunization and homologous immunization with spike protein nanoparticles. Interestingly, Th1 cell activation was induced by immunization schedules including Ad5/MERS, but not by those including only spike protein nanoparticles. Heterologous prime–boost vaccination regimens including Ad5/MERS elicited simultaneous Th1 and Th2 responses, but homologous prime–boost regimens did not. Thus, heterologous prime–boost may induce longer-lasting immune responses against MERS-CoV because of an appropriate balance of Th1/Th2 responses. However, both heterologous prime–boost and homologous spike protein nanoparticles vaccinations could provide protection from MERS-CoV challenge in mice. Our results demonstrate that heterologous immunization by priming with Ad5/MERS and boosting with spike protein nanoparticles could be an efficient prophylactic strategy against MERS-CoV infection.
Collapse
Affiliation(s)
- Seo-Yeon Jung
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Kyung Won Kang
- Division of Biotechnology, Chonbuk National University, Iksan 570-752, Republic of Korea
| | - Eun-Young Lee
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Dong-Won Seo
- Division of Biotechnology, Chonbuk National University, Iksan 570-752, Republic of Korea
| | - Hong-Lim Kim
- Seoul St. Mary's Hospital, School of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hak Kim
- Division of VAX R&D, Life Science Research Institute, SK Chemical, Seongnam 12771, Republic of Korea
| | - TaeWoo Kwon
- Division of VAX R&D, Life Science Research Institute, SK Chemical, Seongnam 12771, Republic of Korea
| | - Hye-Lim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hun Kim
- Division of VAX R&D, Life Science Research Institute, SK Chemical, Seongnam 12771, Republic of Korea
| | - Sang-Myeong Lee
- Division of Biotechnology, Chonbuk National University, Iksan 570-752, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea.
| |
Collapse
|
15
|
Intranasal immunisation with recombinant adenovirus vaccines protects against a lethal challenge with pneumonia virus of mice. Vaccine 2015; 33:6641-9. [PMID: 26529077 PMCID: PMC7125973 DOI: 10.1016/j.vaccine.2015.10.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 10/13/2015] [Accepted: 10/22/2015] [Indexed: 01/16/2023]
Abstract
Pneumonia virus of mice (PVM) infection of BALB/c mice induces bronchiolitis leading to a fatal pneumonia in a dose-dependent manner, closely paralleling the development of severe disease during human respiratory syncytial virus infection in man, and is thus a recognised model in which to study the pathogenesis of pneumoviruses. This model system was used to investigate delivery of the internal structural proteins of PVM as a potential vaccination strategy to protect against pneumovirus disease. Replication-deficient recombinant human adenovirus serotype 5 (rAd5) vectors were constructed that expressed the M or N gene of PVM pathogenic strain J3666. Intranasal delivery of these rAd5 vectors gave protection against a lethal challenge dose of PVM in three different mouse strains, and protection lasted for at least 20 weeks post-immunisation. Whilst the PVM-specific antibody response in such animals was weak and inconsistent, rAd5N primed a strong PVM-specific CD8+ T cell response and, to a lesser extent, a CD4+ T cell response. These findings suggest that T-cell responses may be more important than serum IgG in the observed protection induced by rAd5N.
Collapse
|
16
|
Ye L, Yang C. Development of vaccines for prevention of Ebola virus infection. Microbes Infect 2015; 17:98-108. [DOI: 10.1016/j.micinf.2014.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 12/07/2014] [Accepted: 12/08/2014] [Indexed: 01/25/2023]
|
17
|
Bovine adenovirus-3 as a vaccine delivery vehicle. Vaccine 2014; 33:493-9. [PMID: 25498212 PMCID: PMC7115382 DOI: 10.1016/j.vaccine.2014.11.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/21/2014] [Accepted: 11/28/2014] [Indexed: 12/15/2022]
Abstract
The use of vaccines is an effective and relatively inexpensive means of controlling infectious diseases, which cause heavy economic losses to the livestock industry through animal loss, decreased productivity, treatment expenses and decreased carcass quality. However, some vaccines produced by conventional means are imperfect in many respects including virulence, safety and efficacy. Moreover, there are no vaccines for some animal diseases. Although genetic engineering has provided new ways of producing effective vaccines, the cost of production for veterinary use is a critical criterion for selecting the method of production and delivery of vaccines. The cost effective production and intrinsic ability to enter cells has made adenovirus vectors a highly efficient tool for delivery of vaccine antigens. Moreover, adenoviruses induce both humoral and cellular immune responses to expressed vaccine antigens. Since nonhuman adenoviruses are species specific, the development of animal specific adenoviruses as vaccine delivery vectors is being evaluated. This review summarizes the work related to the development of bovine adenovirus-3 as a vaccine delivery vehicle in animals, particularly cattle.
Collapse
|
18
|
Bogers WM, Oostermeijer H, Mooij P, Koopman G, Verschoor EJ, Davis D, Ulmer JB, Brito LA, Cu Y, Banerjee K, Otten GR, Burke B, Dey A, Heeney JL, Shen X, Tomaras GD, Labranche C, Montefiori DC, Liao HX, Haynes B, Geall AJ, Barnett SW. Potent immune responses in rhesus macaques induced by nonviral delivery of a self-amplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsion. J Infect Dis 2014; 211:947-55. [PMID: 25234719 DOI: 10.1093/infdis/jiu522] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Self-amplifying messenger RNA (mRNA) of positive-strand RNA viruses are effective vectors for in situ expression of vaccine antigens and have potential as a new vaccine technology platform well suited for global health applications. The SAM vaccine platform is based on a synthetic, self-amplifying mRNA delivered by a nonviral delivery system. The safety and immunogenicity of an HIV SAM vaccine encoding a clade C envelope glycoprotein formulated with a cationic nanoemulsion (CNE) delivery system was evaluated in rhesus macaques. The HIV SAM vaccine induced potent cellular immune responses that were greater in magnitude than those induced by self-amplifying mRNA packaged in a viral replicon particle (VRP) or by a recombinant HIV envelope protein formulated with MF59 adjuvant, anti-envelope binding (including anti-V1V2), and neutralizing antibody responses that exceeded those induced by the VRP vaccine. These studies provide the first evidence in nonhuman primates that HIV vaccination with a relatively low dose (50 µg) of formulated self-amplifying mRNA is safe and immunogenic.
Collapse
Affiliation(s)
- Willy M Bogers
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | | | - Petra Mooij
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Gerrit Koopman
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | | | - David Davis
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | | | | | - Yen Cu
- Novartis Vacccines, Cambridge, Massachusetts
| | | | | | - Brian Burke
- Novartis Vacccines, Cambridge, Massachusetts
| | - Antu Dey
- Novartis Vacccines, Cambridge, Massachusetts
| | - Jonathan L Heeney
- Department of Veterinary Medicine, University of Cambridge, United Kingdom
| | | | - Georgia D Tomaras
- Duke Human Vaccine Institute Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Celia Labranche
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - David C Montefiori
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Hua-Xin Liao
- Duke Human Vaccine Institute Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Barton Haynes
- Duke Human Vaccine Institute Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | | |
Collapse
|
19
|
Gray GE, Moodie Z, Metch B, Gilbert PB, Bekker LG, Churchyard G, Nchabeleng M, Mlisana K, Laher F, Roux S, Mngadi K, Innes C, Mathebula M, Allen M, McElrath MJ, Robertson M, Kublin J, Corey L. Recombinant adenovirus type 5 HIV gag/pol/nef vaccine in South Africa: unblinded, long-term follow-up of the phase 2b HVTN 503/Phambili study. THE LANCET. INFECTIOUS DISEASES 2014; 14:388-96. [PMID: 24560541 DOI: 10.1016/s1473-3099(14)70020-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The HVTN 503/Phambili study, which assessed the efficacy of the Merck Ad5 gag/pol/nef subtype B HIV-1 preventive vaccine in South Africa, was stopped when futility criteria in the Step study (assessing the same vaccine in the Americas, Caribbean, and Australia) were met. Here we report long-term follow-up data. METHODS HVTN 503/Phambili was a double-blind, placebo-controlled, randomised trial that recruited HIV-1 uninfected, sexually active adults aged 18-35 years from five sites in South Africa. Eligible participants were randomly assigned (1:1) by computer-generated random numbers to either vaccine or placebo, stratified by site and sex. Cox proportional hazards models were used to estimate HIV-1 infection in the modified intention-to-treat cohort, all of whom were unmasked early in follow-up. The trial is registered with ClinicalTrials.gov, number NCT00413725 and the South African National Health Research Database, number DOH-27-0207-1539. FINDINGS Between Jan 24, 2007, and Sept 19, 2007, 801 participants (26·7%) of a planned 3000 were randomly assigned (400 to vaccine, 401 to placebo); 216 (27%) received only one injection, 529 (66%) received only two injections, and 56 (7%) received three injections. At a median follow-up of 42 months (IQR 31-42), 63 vaccine recipients (16%) had HIV-1 infection compared with 37 placebo recipients (9%; adjusted HR 1·70, 95% CI 1·13-2·55; p=0·01). Risk for HIV-1 infection did not differ according to the number of vaccinations received, sex, circumcision, or adenovirus type 5 (Ad5) serostatus. Differences in risk behaviour at baseline or during the study, or annualised dropout rate (7·7% [95% CI 6·2-9·5] for vaccine recipients vs 8·8% [7·1-10·7] for placebo recipients; p=0·40) are unlikely explanations for the increased rate of HIV-1 infections seen in vaccine recipients. INTERPRETATION The increased risk of HIV-1 acquisition in vaccine recipients, irrespective of number of doses received, warrants further investigation to understand the biological mechanism. We caution against further use of the Ad5 vector for HIV vaccines. FUNDING National Institute of Allergy and Infectious Diseases, Merck, and South African Medical Research Council.
Collapse
Affiliation(s)
- Glenda E Gray
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa; South African Medical Research Council, Cape Town, South Africa.
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Barbara Metch
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Linda-Gail Bekker
- Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | | | | | - Koleka Mlisana
- Centre for AIDS Programme for Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - Fatima Laher
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Surita Roux
- Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | - Kathryn Mngadi
- Centre for AIDS Programme for Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - Craig Innes
- Aurum Institute for Health Research, Johannesburg, South Africa
| | - Matsontso Mathebula
- MEDUNSA HIV Clinical Research Unit, University of Limpopo, Mankweng-E, South Africa
| | - Mary Allen
- Vaccine Research Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M Julie McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michael Robertson
- Infectious Diseases and Vaccines Clinical Research, Merck and Company, North Wales, PA, USA
| | - James Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | |
Collapse
|
20
|
Abstract
Immune-based therapy (IBT) interventions have found a window of opportunity within some limitations of the otherwise successful combined antiretroviral therapy (cART). Two major paradigms drove immunotherapeutic research to combat human immunodeficiency virus (HIV) infection. First, IBTs were proposed either to help restore CD4(+) T-cell counts in cases of therapeutic failures with cytokines, interleukin-2 (IL-2) or IL-7, or to better control HIV and disease progression during treatment interruptions with anti-HIV therapeutic candidate vaccines. The most widely used candidates were HIV-recombinant live vector-based alone or combined with other vaccine compounds and dendritic cell (DC) therapies. A more recent and current paradigm aims at achieving HIV cure by combining IBT with cART using either cytokines to reactivate virus production in latently infected cells and/or therapeutic immunization to boost HIV-specific immunity in a 'shock and kill' strategy. This review summarizes the rationale, hopes, and mechanisms of successes and failures of these cytokine-based and vaccine-based immune interventions. Results from these first series of IBTs have been so far somewhat disappointing in terms of clinical relevance, but have provided lessons that are discussed in light of the future combined strategies to be developed toward an HIV cure.
Collapse
Affiliation(s)
- Guislaine Carcelain
- UPMC Univ Paris 06, UMR-S945, Laboratory of Immunity and Infection, Paris, France
| | | |
Collapse
|
21
|
de Cassan SC, Draper SJ. Recent advances in antibody-inducing poxviral and adenoviral vectored vaccine delivery platforms for difficult disease targets. Expert Rev Vaccines 2014; 12:365-78. [DOI: 10.1586/erv.13.11] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
Abstract
Vaccines to prevent HIV remain desperately needed, but a number of challenges, including retroviral integration, establishment of anatomic reservoir sites, high sequence diversity, and heavy envelope glycosylation. have precluded development of a highly effective vaccine. DNA vaccines have been utilized as candidate HIV vaccines because of their ability to generate cellular and humoral immune responses, the lack of anti-vector response allowing for repeat administration, and their ability to prime the response to viral-vectored vaccines. Because the HIV epidemic has disproportionately affected the developing world, the favorable thermostability profile and relative ease and low cost of manufacture of DNA vaccines offer additional advantages. In vivo electroporation (EP) has been utilized to improve immune responses to DNA vaccines as candidate HIV-1 vaccines in standalone or prime-boost regimens with both proteins and viral-vectored vaccines in several animal models and, more recently, in human clinical trials. This chapter describes the preclinical and clinical development of candidate DNA vaccines for HIV-1 delivered by EP, including challenges to bringing this technology to the developing world.
Collapse
Affiliation(s)
- Sandhya Vasan
- Department of Retrovirology, US Army Medical Component, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| |
Collapse
|
23
|
Capone S, D'Alise AM, Ammendola V, Colloca S, Cortese R, Nicosia A, Folgori A. Development of chimpanzee adenoviruses as vaccine vectors: challenges and successes emerging from clinical trials. Expert Rev Vaccines 2013; 12:379-93. [PMID: 23560919 DOI: 10.1586/erv.13.15] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Replication-defective chimpanzee adenovirus vectors are emerging as a promising new class of genetic vaccine carriers. Chimpanzee adenovirus vectors have now reached the clinical stage and appear to be endowed with all the properties needed for human vaccine development, including high quality and magnitude of the immune response induced against the encoded antigens, good safety and ease of manufacturing on a large-scale basis. Here the authors review the recent findings of this novel class of adenovirus vectors and compare their properties to other clinical stage vaccine vectors derived from poxvirus, alphavirus and human adenovirus.
Collapse
|
24
|
Baldwin SL, Ching LK, Pine SO, Moutaftsi M, Lucas E, Vallur A, Orr MT, Bertholet S, Reed SG, Coler RN. Protection against tuberculosis with homologous or heterologous protein/vector vaccine approaches is not dependent on CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:2514-2525. [PMID: 23904160 DOI: 10.4049/jimmunol.1301161] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Considerable effort has been directed to develop Mycobacterium tuberculosis vaccines to boost bacille Calmette-Guérin or for those who cannot be immunized with bacille Calmette-Guérin. We hypothesized that CD4(+) and CD8(+) T cell responses with a heterologous prime/boost vaccine approach could induce long-lived vaccine efficacy against M. tuberculosis in C57BL/6 mice. We produced an adenovirus vector expressing ID93 (Ad5-ID93) for induction of CD8 T cells to use with our candidate tuberculosis vaccine, ID93/glucopyranosyl lipid adjuvant (GLA)-stable emulsion (SE), which induces potent Th1 CD4 T cells. Ad5-ID93 generates ID93-specific CD8(+) T cell responses and induces protection against M. tuberculosis. When Ad5-ID93 is administered in a prime-boost strategy with ID93/GLA-SE, both CD4(+) and CD8(+) T cells are generated and provide protection against M. tuberculosis. In a MHC class I-deficient mouse model, all groups including the Ad5-ID93 group elicited an Ag-specific CD4(+) T cell response and significantly fewer Ag-specific CD8(+) T cells, but were still protected against M. tuberculosis, suggesting that CD4(+) Th1 T cells could compensate for the loss of CD8(+) T cells. Lastly, the order of the heterologous immunizations was critical. Long-lived vaccine protection was observed only when Ad5-ID93 was given as the boost following an ID93/GLA-SE prime. The homologous ID93/GLA-SE prime/boost regimen also induced long-lived protection. One of the correlates of protection between these two approaches was an increase in the total number of ID93-specific IFN-γ-producing CD4(+) T cells 6 mo following the last immunization. Our findings provide insight into the development of vaccines not only for tuberculosis, but other diseases requiring T cell immunity.
Collapse
Affiliation(s)
- Susan L Baldwin
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Lance K Ching
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Samuel O Pine
- Allergan, Inc. 2525 Dupont Dr., Irvine, CA USA 92612
| | - Magdalini Moutaftsi
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Elyse Lucas
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Aarthy Vallur
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Mark T Orr
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | | | - Steven G Reed
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102.,Department of Global Health, University of Washington, Seattle, WA, USA 98195.,Immune Design Corp., 1124 Columbia Street, Suite 700, Seattle, WA, USA 98104
| | - Rhea N Coler
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102.,Department of Global Health, University of Washington, Seattle, WA, USA 98195
| |
Collapse
|
25
|
Kron MW, Engler T, Schmidt E, Schirmbeck R, Kochanek S, Kreppel F. High-capacity adenoviral vectors circumvent the limitations of ΔE1 and ΔE1/ΔE3 adenovirus vectors to induce multispecific transgene product-directed CD8 T-cell responses. J Gene Med 2013; 13:648-57. [PMID: 22095925 DOI: 10.1002/jgm.1629] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The ability to induce cytotoxic T lymphocyte (CTL) responses that are multispecific is considered to comprise an essential feature for an efficacious genetic vaccine against many pathogens including HIV and hepatitis C virus. ΔE1Ad vectors are promising vectored vaccines but have been shown to induce antigen-specific CTLs with only limited multispecificity. In the present study, we investigated the applicability of gene-deleted high-capacity adenovirus (HC-Ad) vectors and focused on the induction of multispecific CTL responses. METHODS We generated Δ E1 and HC-Ad vectors expressing hepatitis B virus small surface antigen (HBsAg). We comparatively analyzed the CTL profiles against various transgene product- and vector-derived epitopes in several mouse strains and HBsAg- and vector-directed antibody responses. RESULTS HC-Ad vectors efficiently induced multispecific HBsAg-directed CTLs. By contrast, ΔE1Ad vectors mainly primed CTLs against one immunodominant epitope of HBsAg. This absence of multispecific CTL responses correlated with the induction of CTLs against viral epitopes generated by de novo expression of Ad genes from the ΔE1Ad vector. However, Ad-specific CTLs induced in trans did not impair HC-AdS-induced multispecific CTL responses against HBsAg. Finally, HC-Ad vectors also induced higher HBsAg antibody titers compared to ΔE1Ad vectors. CONCLUSIONS De novo expression of viral genes from ΔE1Ad vector genomes restricts the multispecificity of transgene product-specific CTLs by immunodominance effects. HC-Ad vectors devoid of Ad genes are favorable for the induction of both multispecific CD8 T-cell responses and high antibody responses. Our results suggest the deletion of Ad genes as an important means for developing potent Ad-based vectored vaccines.
Collapse
|
26
|
Vitelli A, Quirion MR, Lo CY, Misplon JA, Grabowska AK, Pierantoni A, Ammendola V, Price GE, Soboleski MR, Cortese R, Colloca S, Nicosia A, Epstein SL. Vaccination to conserved influenza antigens in mice using a novel Simian adenovirus vector, PanAd3, derived from the bonobo Pan paniscus. PLoS One 2013; 8:e55435. [PMID: 23536756 PMCID: PMC3594242 DOI: 10.1371/journal.pone.0055435] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 12/23/2012] [Indexed: 12/11/2022] Open
Abstract
Among approximately 1000 adenoviruses from chimpanzees and bonobos studied recently, the Pan Adenovirus type 3 (PanAd3, isolated from a bonobo, Pan paniscus) has one of the best profiles for a vaccine vector, combining potent transgene immunogenicity with minimal pre-existing immunity in the human population. In this study, we inserted into a replication defective PanAd3 a transgene expressing a fusion protein of conserved influenza antigens nucleoprotein (NP) and matrix 1 (M1). We then studied antibody and T cell responses as well as protection from challenge infection in a mouse model. A single intranasal administration of PanAd3-NPM1 vaccine induced strong antibody and T cell responses, and protected against high dose lethal influenza virus challenge. Thus PanAd3 is a promising candidate vector for vaccines, including universal influenza vaccines.
Collapse
MESH Headings
- Adenoviruses, Human/immunology
- Adenoviruses, Simian/genetics
- Adenoviruses, Simian/immunology
- Amino Acid Sequence
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Cross Reactions/immunology
- Female
- Gene Expression
- Genetic Vectors/administration & dosage
- Genetic Vectors/genetics
- Genetic Vectors/immunology
- Humans
- Immunity, Mucosal
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Mice
- Molecular Sequence Data
- Nucleocapsid Proteins
- Nucleophosmin
- Orthomyxoviridae Infections/prevention & control
- Pan paniscus
- RNA-Binding Proteins/chemistry
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/immunology
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes/immunology
- Viral Core Proteins/chemistry
- Viral Core Proteins/genetics
- Viral Core Proteins/immunology
- Viral Matrix Proteins/chemistry
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
Collapse
Affiliation(s)
| | - Mary R. Quirion
- Gene Therapy and Immunogenicity Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Chia-Yun Lo
- Gene Therapy and Immunogenicity Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Julia A. Misplon
- Gene Therapy and Immunogenicity Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | | | | | | | - Graeme E. Price
- Gene Therapy and Immunogenicity Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Mark R. Soboleski
- Gene Therapy and Immunogenicity Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | | | | | - Alfredo Nicosia
- Okairòs, Rome, Italy
- Centro di Ingegneria Genetica e Biotecnologia Avanzate (CEINGE), Naples, Italy
- Department of Biochemistry and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Suzanne L. Epstein
- Gene Therapy and Immunogenicity Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
27
|
Sullivan NJ, Fausther-Bovendo H, Nabel GJ. Ebola vaccine. Vaccines (Basel) 2013. [DOI: 10.1016/b978-1-4557-0090-5.00049-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
28
|
Vasconcelos JR, Dominguez MR, Araújo AF, Ersching J, Tararam CA, Bruna-Romero O, Rodrigues MM. Relevance of long-lived CD8(+) T effector memory cells for protective immunity elicited by heterologous prime-boost vaccination. Front Immunol 2012; 3:358. [PMID: 23264773 PMCID: PMC3525016 DOI: 10.3389/fimmu.2012.00358] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/10/2012] [Indexed: 11/13/2022] Open
Abstract
Owing to the importance of major histocompatibility complex class Ia-restricted CD8(+) T cells for host survival following viral, bacterial, fungal, or parasitic infection, it has become largely accepted that these cells should be considered in the design of a new generation of vaccines. For the past 20 years, solid evidence has been provided that the heterologous prime-boost regimen achieves the best results in terms of induction of long-lived protective CD8(+) T cells against a variety of experimental infections. Although this regimen has often been used experimentally, as is the case for many vaccines, the mechanism behind the efficacy of this vaccination regimen is still largely unknown. The main purpose of this review is to examine the characteristics of the protective CD8(+) T cells generated by this vaccination regimen. Part of its efficacy certainly relies on the generation and maintenance of large numbers of specific lymphocytes. Other specific characteristics may also be important, and studies on this direction have only recently been initiated. So far, the characterization of these protective, long-lived T cell populations suggests that there is a high frequency of polyfunctional T cells; these cells cover a large breadth and display a T effector memory (TEM) phenotype. These TEM cells are capable of proliferating after an infectious challenge and are highly refractory to apoptosis due to a control of the expression of pro-apoptotic receptors such as CD95. Also, they do not undergo significant long-term immunological erosion. Understanding the mechanisms that control the generation and maintenance of the protective activity of these long-lived TEM cells will certainly provide important insights into the physiology of CD8(+) T cells and pave the way for the design of new or improved vaccines.
Collapse
Affiliation(s)
- José R Vasconcelos
- Centro de Terapia Celular e Molecular, Universidade Federal de São Paulo - Escola Paulista de Medicina São Paulo, São Paulo, Brazil ; Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo - Escola Paulista de Medicina São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
29
|
An adenovirus-vectored nasal vaccine confers rapid and sustained protection against anthrax in a single-dose regimen. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 20:1-8. [PMID: 23100479 DOI: 10.1128/cvi.00280-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Bacillus anthracis is the causative agent of anthrax, and its spores have been developed into lethal bioweapons. To mitigate an onslaught from airborne anthrax spores that are maliciously disseminated, it is of paramount importance to develop a rapid-response anthrax vaccine that can be mass administered by nonmedical personnel during a crisis. We report here that intranasal instillation of a nonreplicating adenovirus vector encoding B. anthracis protective antigen could confer rapid and sustained protection against inhalation anthrax in mice in a single-dose regimen in the presence of preexisting adenovirus immunity. The potency of the vaccine was greatly enhanced when codons of the antigen gene were optimized to match the tRNA pool found in human cells. In addition, an adenovirus vector encoding lethal factor can confer partial protection against inhalation anthrax and might be coadministered with a protective antigen-based vaccine.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The major target groups for an HIV vaccine include breastfeeding infants and adolescents. Differential immune maturity in these age groups may significantly impact vaccine efficacy, and should be taken into account when developing vaccines. Here we review these differences, with an emphasis on the immune response to vaccines for HIV and other pathogens. Recommendations for potential adaptation of current HIV vaccines are also made. RECENT FINDINGS An effective neonatal vaccine needs to be immunogenic in the presence of maternal antibody, and must induce cytotoxic T-lymphocyte responses, neutralizing antibody responses, both systemic and mucosal. There is renewed hope in the possibility of stimulating neutralizing antibodies with HIV vaccination. DNA vaccines are promising for neonates, but will need appropriate boosting. Certain adjuvants and vector delivery systems are more suitable for neonates. Adolescents may have stronger immune responses to HIV vaccines than adults, and will also require induction of mucosal neutralizing humoral and cellular immunity. SUMMARY Some current HIV vaccine strategies may need adaptation for neonates and suitable product development should be accelerated. Vaccines could induce better responses in adolescents and therefore should not be discarded prematurely. Development of vaccines that have potential for these age groups is an urgent global priority.
Collapse
|
31
|
Lifson JD, Haigwood NL. Lessons in nonhuman primate models for AIDS vaccine research: from minefields to milestones. Cold Spring Harb Perspect Med 2012; 2:a007310. [PMID: 22675663 PMCID: PMC3367532 DOI: 10.1101/cshperspect.a007310] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nonhuman primate (NHP) disease models for AIDS have made important contributions to the search for effective vaccines for AIDS. Viral diversity, persistence, capacity for immune evasion, and safety considerations have limited development of conventional approaches using killed or attenuated vaccines, necessitating the development of novel approaches. Here we highlight the knowledge gained and lessons learned in testing vaccine concepts in different virus/NHP host combinations.
Collapse
Affiliation(s)
- Jeffrey D Lifson
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., National Cancer Institute, Frederick, Maryland, USA
| | | |
Collapse
|
32
|
Santra S, Muldoon M, Watson S, Buzby A, Balachandran H, Carlson KR, Mach L, Kong WP, McKee K, Yang ZY, Rao SS, Mascola JR, Nabel GJ, Korber BT, Letvin NL. Breadth of cellular and humoral immune responses elicited in rhesus monkeys by multi-valent mosaic and consensus immunogens. Virology 2012; 428:121-7. [PMID: 22521913 DOI: 10.1016/j.virol.2012.03.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/12/2012] [Accepted: 03/22/2012] [Indexed: 12/01/2022]
Abstract
To create an HIV-1 vaccine that generates sufficient breadth of immune recognition to protect against the genetically diverse forms of the circulating virus, we have been exploring vaccines based on consensus and mosaic protein designs. Increasing the valency of a mosaic immunogen cocktail increases epitope coverage but with diminishing returns, as increasingly rare epitopes are incorporated into the mosaic proteins. In this study we compared the immunogenicity of 2-valent and 3-valent HIV-1 envelope mosaic immunogens in rhesus monkeys. Immunizations with the 3-valent mosaic immunogens resulted in a modest increase in the breadth of vaccine-elicited T lymphocyte responses compared to the 2-valent mosaic immunogens. However, the 3-valent mosaic immunogens elicited significantly higher neutralizing responses to Tier 1 viruses than the 2-valent mosaic immunogens. These findings underscore the potential utility of polyvalent mosaic immunogens for eliciting both cellular and humoral immune responses to HIV-1.
Collapse
Affiliation(s)
- Sampa Santra
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Next-generation dengue vaccines: novel strategies currently under development. Viruses 2011; 3:1800-14. [PMID: 22069516 PMCID: PMC3205382 DOI: 10.3390/v3101800] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/20/2011] [Accepted: 09/20/2011] [Indexed: 11/17/2022] Open
Abstract
Dengue has become the most important arboviral infection worldwide with more than 30 million cases of dengue fever estimated to occur each year. The need for a dengue vaccine is great and several live attenuated dengue candidate vaccines are proceeding through clinical evaluation. The need to induce a balanced immune response against all four DENV serotypes with a single vaccine has been a challenge for dengue vaccine developers. A live attenuated DENV chimeric vaccine produced by Sanofi Pasteur has recently entered Phase III evaluation in numerous dengue-endemic regions of the world. Viral interference between serotypes contained in live vaccines has required up to three doses of the vaccine be given over a 12-month period of time. For this reason, novel DENV candidate vaccines are being developed with the goal of achieving a protective immune response with an immunization schedule that can be given over the course of a few months. These next-generation candidates include DNA vaccines, recombinant adenovirus vectored vaccines, alphavirus replicons, and sub-unit protein vaccines. Several of these novel candidates will be discussed.
Collapse
|
35
|
Grant-Klein RJ, Altamura LA, Schmaljohn CS. Progress in recombinant DNA-derived vaccines for Lassa virus and filoviruses. Virus Res 2011; 162:148-61. [PMID: 21925552 DOI: 10.1016/j.virusres.2011.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/01/2011] [Accepted: 09/04/2011] [Indexed: 11/19/2022]
Abstract
Developing vaccines for highly pathogenic viruses such as those causing Lassa, Ebola, and Marburg hemorrhagic fevers is a daunting task due to both scientific and logistical constraints. Scientific hurdles to overcome include poorly defined relationships between pathogenicity and protective immune responses, genetic diversity of viruses, and safety in a target population that includes a large number of individuals with compromised immune systems. Logistical obstacles include the requirement for biosafety level-4 containment to study the authentic viruses, the poor public health infrastructure of the endemic disease areas, and the cost of developing these vaccines for use in non-lucrative markets. Recombinant DNA-based vaccine approaches offer promise of overcoming some of these issues. In this review, we consider the status of various recombinant DNA candidate vaccines against Lassa virus and filoviruses which have been tested in animals.
Collapse
Affiliation(s)
- Rebecca J Grant-Klein
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | | |
Collapse
|
36
|
Chen Y, Xie Y, Chan T, Sami A, Ahmed S, Liu Q, Xiang J. Adjuvant effect of HER-2/neu-specific adenoviral vector stimulating CD8⁺ T and natural killer cell responses on anti-HER-2/neu antibody therapy for well-established breast tumors in HER-2/neu transgenic mice. Cancer Gene Ther 2011; 18:489-99. [PMID: 21566669 PMCID: PMC7091910 DOI: 10.1038/cgt.2011.18] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 12/22/2010] [Accepted: 02/17/2011] [Indexed: 12/21/2022]
Abstract
Approximately one third of patients with advanced human epidermal growth factor receptor 2 (HER-2)/neu-positive breast cancer respond to trastuzumab monotherapy, a humanized anti-HER-2/neu antibody. However, de novo and acquired antibody resistance is one of the major limitations of trastuzumab therapy warranting the search for other therapeutic strategies. One of the most remarkable features of adenovirus (AdV)-based vaccine is its ability to induce exceptionally high and sustained frequencies of transgene product-specific CD8(+) T-cell responses. In this study, we constructed two recombinant AdVs (AdV(OVA) and AdV(HER-2)) expressing ovalbumin (OVA) and HER-2/neu, and assessed AdV-induced antigen-specific cellular immune responses and preventive/therapeutic antitumor immunity. We demonstrate that AdV(OVA) stimulates efficient OVA-specific CD8(+) cytotoxic T lymphocyte (CTL) and natural killer responses, leading to preventive long-term immunity against OVA-expressing BL6-10ova melanoma in wild-type C56BL/6 mice. We further demonstrate that AdV(HER-2) stimulates HER-2/neu-specific CD8(+) CTL responses, leading to a significant reduction in breast carcinogenesis in transgenic FVBneuN mice (P<0.05), but has little therapeutic effect on pre-existing Tg1-1 tumor even at early stage (15 mm(3)). In contrast, the anti-HER-2/neu antibody therapy is capable of completely inhibiting Tg1-1 tumor growth at early stage, but fails to eradicate well-established Tg1-1 breast tumor (100 mm(3)). Interestingly, a combinatorial immunotherapy of anti-HER-2/neu antibody with AdV(HER-2) vaccine was capable of curing 4 of 10 studied mice bearing well-established Tg1-1 breast tumors and significantly delaying in death of the remaining six tumor-bearing mice (P<0.05). Taken together, our results suggest an adjuvant effect of AdV(HER-2) on anti-HER-2/neu antibody therapy for well-established breast tumor in transgenic FVBneuN mice, and this combinatorial immunotherapy of trastuzumab with AdV(HER-2) vaccine may be used as a new therapeutic strategy for treatment of advanced HER-2/neu-positive breast cancer.
Collapse
Affiliation(s)
- Y Chen
- Research Division, Cancer Research Unit, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan Canada
| | - Y Xie
- Research Division, Cancer Research Unit, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan Canada
| | - T Chan
- Research Division, Cancer Research Unit, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan Canada
| | - A Sami
- Department of Oncology, Saskatoon Cancer Center, University of Saskatchewan, Saskatoon, Saskatchewan Canada
| | - S Ahmed
- Department of Oncology, Saskatoon Cancer Center, University of Saskatchewan, Saskatoon, Saskatchewan Canada
| | - Q Liu
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan Canada
| | - J Xiang
- Research Division, Cancer Research Unit, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan Canada
- Department of Oncology, Saskatoon Cancer Center, University of Saskatchewan, Saskatoon, Saskatchewan Canada
| |
Collapse
|
37
|
Pillai VKB, Kannanganat S, Penaloza-Macmaster P, Chennareddi L, Robinson HL, Blackwell J, Amara RR. Different patterns of expansion, contraction and memory differentiation of HIV-1 Gag-specific CD8 T cells elicited by adenovirus type 5 and modified vaccinia Ankara vaccines. Vaccine 2011; 29:5399-406. [PMID: 21651938 DOI: 10.1016/j.vaccine.2011.05.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 05/18/2011] [Accepted: 05/23/2011] [Indexed: 12/29/2022]
Abstract
The magnitude and functional quality of antiviral CD8 T cell responses are critical for the efficacy of T cell based vaccines. Here, we investigate the influence of two popular viral vectors, adenovirus type 5 (Ad5) and modified vaccinia Ankara (MVA), on expansion, contraction and memory differentiation of HIV-1 Gag insert-specific CD8 T cell responses following immunization and show different patterns for the two recombinant viral vectors. The Ad5 vector primed 6-fold higher levels of insert-specific CD8 effector T cells than the MVA vector. The Ad5-primed effector cells also underwent less contraction (<2-fold) than the MVA-primed cells (>5-fold). The Ad5-primed memory cells were predominantly CD62L negative (effector memory) whereas the MVA-primed memory cells were predominantly CD62L positive (central memory). Consistent with their memory phenotype, MVA-primed CD8 T cells underwent higher fold expansion than Ad5-primed CD8 T cells following a homologous or heterologous boost. Impressively, the Ad5 boost changed the quality of MVA-primed memory response such that they undergo less contraction with effector memory phenotype. However, the MVA boost did not influence the contraction and memory phenotype of Ad5-primed response. In conclusion, our results demonstrate that vaccine vector strongly influences the expansion, contraction and the functional quality of insert-specific CD8 T cell responses and have implications for vaccine development against infectious diseases.
Collapse
Affiliation(s)
- Vinod Kumar Bhaskara Pillai
- Vaccine Research Center, Department of Microbiology and Immunology, Yerkes National Primate Research Center and Emory University School of Medicine, Emory University, 954 Gatewood Road NE, Atlanta, GA 30329, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Vasan S, Hurley A, Schlesinger SJ, Hannaman D, Gardiner DF, Dugin DP, Boente-Carrera M, Vittorino R, Caskey M, Andersen J, Huang Y, Cox JH, Tarragona-Fiol T, Gill DK, Cheeseman H, Clark L, Dally L, Smith C, Schmidt C, Park HH, Kopycinski JT, Gilmour J, Fast P, Bernard R, Ho DD. In vivo electroporation enhances the immunogenicity of an HIV-1 DNA vaccine candidate in healthy volunteers. PLoS One 2011; 6:e19252. [PMID: 21603651 PMCID: PMC3095594 DOI: 10.1371/journal.pone.0019252] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 03/23/2011] [Indexed: 11/29/2022] Open
Abstract
Background DNA-based vaccines have been safe but weakly immunogenic in humans to
date. Methods and Findings We sought to determine the safety, tolerability, and immunogenicity of ADVAX,
a multigenic HIV-1 DNA vaccine candidate, injected intramuscularly by
in vivo electroporation (EP) in a Phase-1,
double-blind, randomized placebo-controlled trial in healthy volunteers.
Eight volunteers each received 0.2 mg, 1 mg, or 4 mg ADVAX or saline placebo
via EP, or 4 mg ADVAX via standard intramuscular injection at weeks 0 and 8.
A third vaccination was administered to eleven volunteers at week 36. EP was
safe, well-tolerated and considered acceptable for a prophylactic vaccine.
EP delivery of ADVAX increased the magnitude of HIV-1-specific cell mediated
immunity by up to 70-fold over IM injection, as measured by gamma interferon
ELISpot. The number of antigens to which the response was detected improved
with EP and increasing dosage. Intracellular cytokine staining analysis of
ELISpot responders revealed both CD4+ and CD8+ T cell responses,
with co-secretion of multiple cytokines. Conclusions This is the first demonstration in healthy volunteers that EP is safe,
tolerable, and effective in improving the magnitude, breadth and durability
of cellular immune responses to a DNA vaccine candidate. Trial Registration ClinicalTrials.gov NCT00545987
Collapse
Affiliation(s)
- Sandhya Vasan
- Aaron Diamond AIDS Research Center, New York, New York, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dhesi Z, Stebbing J. The HVTN 503/Phambili study: efficacy is always the issue. THE LANCET. INFECTIOUS DISEASES 2011; 11:490-1. [PMID: 21570356 DOI: 10.1016/s1473-3099(11)70103-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Zaneeta Dhesi
- Section of Infectious Diseases and Immunity, Imperial College, London, UK
| | | |
Collapse
|
40
|
Nicholson O, DiCandilo F, Kublin J, Sun X, Quirk E, Miller M, Gray G, Pape J, Robertson MN, Mehrotra DV, Self S, Turner K, Sanchez J, Pitisuttithum P, Duerr A, Dubey S, Kierstead L, Casimiro D, Hammer For The Merck V/Hiv Vaccine Trials Network Study Team SM. Safety and Immunogenicity of the MRKAd5 gag HIV Type 1 Vaccine in a Worldwide Phase 1 Study of Healthy Adults. AIDS Res Hum Retroviruses 2011; 27:557-567. [PMID: 20854108 PMCID: PMC3422055 DOI: 10.1089/aid.2010.0151] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The safety and immunogenicity of the MRK adenovirus type 5 (Ad5) HIV-1 clade B gag vaccine was assessed in an international Phase I trial. Three-hundred and sixty healthy HIV-uninfected adults were enrolled on five continents. Subjects received placebo or 1 × 109 or 1 × 1010 viral particles (vp) per dose of the MRKAd5 HIV-1 gag vaccine at day 1, week 4, and week 26. Immunogenicity was evaluated using an IFN-γ ELISPOT gag 15-mer assay with positive responses defined as ≥55 SFC/106 PBMCs and ≥4-fold over mock control. The vaccine was well tolerated. The most common adverse events were injection site reactions, headache, pyrexia, diarrhea, fatigue, and myalgia. At week 30, geometric mean ELISPOT responses were 24, 114, and 226 SFC/106 PBMCs in the placebo, 1 × 109 vp/dose, and 1 × 1010 vp/dose groups, respectively. Overall, responses to 1 × 1010 vp were 85% and 68% in subjects with low (≤200) and high (>200) baseline Ad5 titers, respectively. The MRKAd5 HIV-1 gag vaccine was immunogenic in diverse geographic regions. Gag ELISPOT responses were greater in the 1 × 1010 vp/dose groups than in the 1 × 109 vp/dose groups. Data from this first international study indicate that adenovirus-vectored vaccines are well tolerated and may be immunogenic in subjects from regions with high prevalence of preexisting Ad5 immunity.
Collapse
Affiliation(s)
- Ouzama Nicholson
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Fay DiCandilo
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - James Kublin
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Xiao Sun
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Erin Quirk
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Michelle Miller
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Glenda Gray
- University of Witwatersrand, Johannesburg, South Africa
| | - Jean Pape
- Weill Cornell Medical College, New York, New York
| | - Michael N Robertson
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Devan V Mehrotra
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Steven Self
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Jorge Sanchez
- Asociacion Civil Impacta Salud y Educacion, Lima, Peru and Investigaciones Medicas en Salu, Immensa, Lima, Peru
| | | | - Ann Duerr
- Fred Hutchinson Cancer Research Center, Seattle, Washington
- University of Washington, Seattle, Washington
| | - Sheri Dubey
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Lisa Kierstead
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Danilo Casimiro
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | | |
Collapse
|
41
|
Hulot SL, Cale EM, Korber BT, Letvin NL. Vaccine-Induced CD8+T Lymphocytes of Rhesus Monkeys Recognize Variant Forms of an HIV Epitope but Do Not Mediate Optimal Functional Activity. THE JOURNAL OF IMMUNOLOGY 2011; 186:5663-74. [DOI: 10.4049/jimmunol.1100287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Makarova N, Zhao C, Zhang Y, Bhosle S, Suppiah S, Rhea JM, Kozyr N, Arnold RS, Ly H, Molinaro RJ, Parslow TG, Hunter E, Liotta D, Petros J, Blackwell JL. Antibody responses against xenotropic murine leukemia virus-related virus envelope in a murine model. PLoS One 2011; 6:e18272. [PMID: 21494670 PMCID: PMC3071813 DOI: 10.1371/journal.pone.0018272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 02/24/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Xenotropic murine leukemia virus-related virus (XMRV) was recently discovered to be the first human gammaretrovirus that is associated with chronic fatigue syndrome and prostate cancer (PC). Although a mechanism for XMRV carcinogenesis is yet to be established, this virus belongs to the family of gammaretroviruses well known for their ability to induce cancer in the infected hosts. Since its original identification XMRV has been detected in several independent investigations; however, at this time significant controversy remains regarding reports of XMRV detection/prevalence in other cohorts and cell type/tissue distribution. The potential risk of human infection, coupled with the lack of knowledge about the basic biology of XMRV, warrants further research, including investigation of adaptive immune responses. To study immunogenicity in vivo, we vaccinated mice with a combination of recombinant vectors expressing codon-optimized sequences of XMRV gag and env genes and virus-like particles (VLP) that had the size and morphology of live infectious XMRV. RESULTS Immunization elicited Env-specific binding and neutralizing antibodies (NAb) against XMRV in mice. The peak titers for ELISA-binding antibodies and NAb were 1:1024 and 1:464, respectively; however, high ELISA-binding and NAb titers were not sustained and persisted for less than three weeks after immunizations. CONCLUSIONS Vaccine-induced XMRV Env antibody titers were transiently high, but their duration was short. The relatively rapid diminution in antibody levels may in part explain the differing prevalences reported for XMRV in various prostate cancer and chronic fatigue syndrome cohorts. The low level of immunogenicity observed in the present study may be characteristic of a natural XMRV infection in humans.
Collapse
Affiliation(s)
- Natalia Makarova
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Chunxia Zhao
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Yuanyuan Zhang
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Sushma Bhosle
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Suganthi Suppiah
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Jeanne M. Rhea
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Natalia Kozyr
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Rebecca S. Arnold
- Department of Urology, Emory University, Atlanta, Georgia, United States of America
| | - Hinh Ly
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Ross J. Molinaro
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Core Laboratories Emory University Hospital Midtown, Emory University, Atlanta, Georgia, United States of America
| | - Tristram G. Parslow
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Eric Hunter
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Dennis Liotta
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
| | - John Petros
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Department of Urology, Emory University, Atlanta, Georgia, United States of America
- Department of Hematology-Oncology, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Jerry L. Blackwell
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Division of Infectious Diseases, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
43
|
Recombinant adenovirus serotype 26 (Ad26) and Ad35 vaccine vectors bypass immunity to Ad5 and protect nonhuman primates against ebolavirus challenge. J Virol 2011; 85:4222-33. [PMID: 21325402 DOI: 10.1128/jvi.02407-10] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The use of adenoviruses (Ad) as vaccine vectors against a variety of pathogens has demonstrated their capacity to elicit strong antibody and cell-mediated immune responses. Adenovirus serotype C vectors, such as Ad serotype 5 (Ad5), expressing Ebolavirus (EBOV) glycoprotein (GP), protect completely after a single inoculation at a dose of 10(10) viral particles. However, the clinical application of a vaccine based on Ad5 vectors may be hampered, since impairment of Ad5 vaccine efficacy has been demonstrated for humans and nonhuman primates with high levels of preexisting immunity to the vector. Ad26 and Ad35 segregate genetically from Ad5 and exhibit lower seroprevalence in humans, making them attractive vaccine vector alternatives. In the series of studies presented, we show that Ad26 and Ad35 vectors generate robust antigen-specific cell-mediated and humoral immune responses against EBOV GP and that Ad5 immune status does not affect the generation of GP-specific immune responses by these vaccines. We demonstrate partial protection against EBOV by a single-shot Ad26 vaccine and complete protection when this vaccine is boosted by Ad35 1 month later. Increases in efficacy are paralleled by substantial increases in T- and B-cell responses to EBOV GP. These results suggest that Ad26 and Ad35 vectors warrant further development as candidate vaccines for EBOV.
Collapse
|
44
|
Falzarano D, Geisbert TW, Feldmann H. Progress in filovirus vaccine development: evaluating the potential for clinical use. Expert Rev Vaccines 2011; 10:63-77. [PMID: 21162622 PMCID: PMC3398800 DOI: 10.1586/erv.10.152] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Marburg and Ebola viruses cause severe hemorrhagic fever in humans and nonhuman primates. Currently, there are no effective treatments and no licensed vaccines; although a number of vaccine platforms have proven successful in animal models. The ideal filovirus vaccine candidate should be able to provide rapid protection following a single immunization, have the potential to work postexposure and be cross-reactive or multivalent against all Marburg virus strains and all relevant Ebola virus species and strains. Currently, there are multiple platforms that have provided prophylactic protection in nonhuman primates, including DNA, recombinant adenovirus serotype 5, recombinant human parainfluenza virus 3 and virus-like particles. In addition, a single platform, recombinant vesicular stomatitis virus, has demonstrated both prophylactic and postexposure protection in nonhuman primates. These results demonstrate that achieving a vaccine that is protective against filoviruses is possible; the challenge now is to prove its safety and efficacy in order to obtain a vaccine that is ready for human use.
Collapse
Affiliation(s)
- Darryl Falzarano
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA
| | - Thomas W Geisbert
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
45
|
Gamble LJ, Matthews QL. Current progress in the development of a prophylactic vaccine for HIV-1. DRUG DESIGN DEVELOPMENT AND THERAPY 2010; 5:9-26. [PMID: 21267356 PMCID: PMC3023272 DOI: 10.2147/dddt.s6959] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since its discovery and characterization in the early 1980s as a virus that attacks the immune system, there has been some success for the treatment of human immunodeficiency virus-1 (HIV-1) infection. However, due to the overwhelming public health impact of this virus, a vaccine is needed urgently. Despite the tireless efforts of scientist and clinicians, there is still no safe and effective vaccine that provides sterilizing immunity. A vaccine that provides sterilizing immunity against HIV infection remains elusive in part due to the following reasons: 1) degree of diversity of the virus, 2) ability of the virus to evade the hosts' immunity, and 3) lack of appropriate animal models in which to test vaccine candidates. There have been several attempts to stimulate the immune system to provide protection against HIV-infection. Here, we will discuss attempts that have been made to induce sterilizing immunity, including traditional vaccination attempts, induction of broadly neutralizing antibody production, DNA vaccines, and use of viral vectors. Some of these attempts show promise pending continued research efforts.
Collapse
Affiliation(s)
- Lena J Gamble
- Department of Medicine, The Gene Therapy Center, University of Alabama at Birmingham, 35294, USA
| | | |
Collapse
|
46
|
Baize S. Towards broad protection against Ebolaviruses. Future Microbiol 2010; 5:1469-73. [PMID: 21073307 DOI: 10.2217/fmb.10.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The Ebola and Marburg viruses (from the filovirus family) induce deadly hemorrhagic fevers for which there is currently no licensed vaccine or treatment. Frequent outbreaks have occurred in sub-Saharan Africa, in humans and nonhuman primates over the last 15 years or so and constitute a major public health problem. Of particular concern, a new species of Ebolavirus recently emerged in Uganda, highlighting the high potential of these viruses to evolve and the need to develop 'broad-spectrum' vaccines against filoviruses. Hensley et al. used their well studied vaccine platform based on DNA vectors and recombinant, replication-defective, adenoviruses producing Ebolavirus glycoproteins to protect cynomolgus monkeys against a heterologous challenge with the new species. Further developments are required before this experimental approach could be adapted for field use in humans, but this study nevertheless constitutes a proof-of-concept for broad protection against Ebolaviruses.
Collapse
Affiliation(s)
- Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, IFR128 Biosciences Gerland, Lyon Sud, 21 av. Tony Garnier, 69365 Lyon cedex 7, France.
| |
Collapse
|
47
|
Wilks AB, Christian EC, Seaman MS, Sircar P, Carville A, Gomez CE, Esteban M, Pantaleo G, Barouch DH, Letvin NL, Permar SR. Robust vaccine-elicited cellular immune responses in breast milk following systemic simian immunodeficiency virus DNA prime and live virus vector boost vaccination of lactating rhesus monkeys. THE JOURNAL OF IMMUNOLOGY 2010; 185:7097-106. [PMID: 21041730 DOI: 10.4049/jimmunol.1002751] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Breast milk transmission of HIV remains an important mode of infant HIV acquisition. Enhancement of mucosal HIV-specific immune responses in milk of HIV-infected mothers through vaccination may reduce milk virus load or protect against virus transmission in the infant gastrointestinal tract. However, the ability of HIV/SIV strategies to induce virus-specific immune responses in milk has not been studied. In this study, five uninfected, hormone-induced lactating, Mamu A*01(+) female rhesus monkey were systemically primed and boosted with rDNA and the attenuated poxvirus vector, NYVAC, containing the SIVmac239 gag-pol and envelope genes. The monkeys were boosted a second time with a recombinant Adenovirus serotype 5 vector containing matching immunogens. The vaccine-elicited immunodominant epitope-specific CD8(+) T lymphocyte response in milk was of similar or greater magnitude than that in blood and the vaginal tract but higher than that in the colon. Furthermore, the vaccine-elicited SIV Gag-specific CD4(+) and CD8(+) T lymphocyte polyfunctional cytokine responses were more robust in milk than in blood after each virus vector boost. Finally, SIV envelope-specific IgG responses were detected in milk of all monkeys after vaccination, whereas an SIV envelope-specific IgA response was only detected in one vaccinated monkey. Importantly, only limited and transient increases in the proportion of activated or CCR5-expressing CD4(+) T lymphocytes in milk occurred after vaccination. Therefore, systemic DNA prime and virus vector boost of lactating rhesus monkeys elicits potent virus-specific cellular and humoral immune responses in milk and may warrant further investigation as a strategy to impede breast milk transmission of HIV.
Collapse
Affiliation(s)
- Andrew B Wilks
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
HIV-1 Gag p17 presented as virus-like particles on the E2 scaffold from Geobacillus stearothermophilus induces sustained humoral and cellular immune responses in the absence of IFNγ production by CD4+ T cells. Virology 2010; 407:296-305. [PMID: 20850858 DOI: 10.1016/j.virol.2010.08.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/13/2010] [Accepted: 08/22/2010] [Indexed: 02/07/2023]
Abstract
We have constructed stable virus-like particles displaying the HIV-1 Gag(p17) protein as an N-terminal fusion with an engineered protein domain from the Geobacillus stearothermophilus pyruvate dehydrogenase subunit E2. Mice immunized with the Gag(p17)-E2 60-mer scaffold particles mounted a strong and sustained antibody response. Antibodies directed to Gag(p17) were boosted significantly with additional immunizations, while anti-E2 responses reached a plateau. The isotype of the induced antibodies was biased towards IgG1, and the E2-primed CD4+ T cells did not secrete IFNγ. Using transgenic mouse model systems, we demonstrated that CD8+ T cells primed with E2 particles were able to exert lytic activity and produce IFNγ. These results show that the E2 scaffold represents a powerful vaccine delivery system for whole antigenic proteins or polyepitope engineered proteins, evoking antibody production and antigen specific CTL activity even in the absence of IFNγ-producing CD4+ T cells.
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Recombinant adenovirus (rAd) vectors have emerged as promising vaccine platform technologies due to their capacity to elicit potent humoral and cellular immune responses to encoded antigens. These vectors are being explored as potential vaccine candidates for a variety of pathogens. This review summarizes current efforts to develop rAd vector-based vaccines for HIV-1. RECENT FINDINGS In the phase 2b Step study, rAd5 vectors expressing clade B HIV-1 Gag, Pol, and Nef antigens failed to afford protection and may have resulted in increased HIV-1 acquisition in certain subgroups. Recent studies have explored the potential reasons for this failure and the utility of novel rAd vectors derived from non-Ad5 serotypes. SUMMARY Current areas of active investigation include the development of alternative serotype rAd vectors, the incorporation of rAd vectors into heterologous vector prime-boost regimens, and the use of rAd vectors to express novel HIV-1 antigens. These HIV-1 vaccine candidates will be evaluated in clinical trials over the next several years.
Collapse
Affiliation(s)
- Dan H Barouch
- Division of Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
| |
Collapse
|
50
|
Induction of both cellular and humoral immunity following a rational prime-boost immunization regimen that incorporates recombinant ovine atadenovirus and fowlpox virus. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1679-86. [PMID: 20810681 DOI: 10.1128/cvi.00291-10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recombinant fowlpox viruses (rFPV) and ovine atadenoviruses (rOAdV) are being developed as safe, nonpathogenic, prophylactic and therapeutic vaccine vectors. There is scope, however, to improve the limited immune responses elicited by each of these vaccine vectors. Using previously determined and optimized routes of administration and viral doses, we characterized the primary adaptive immune responses elicited by recombinant variants of each virus. We demonstrate the contrasting nature of the response elicited by each recombinant virus. Whereas rFPV generates predominately cell-mediated immunity to our nominal target antigen, ovalbumin (OVA), rOAdV drives strong humoral responses. By defining the time taken to achieve maximal cytotoxic T cell responses and by studying the different patterns and kinetics of major histocompatibility complex class I-restricted OVA antigen expression postimmunization, we proposed a heterologous prime-boost regimen of immunization with rOAdV followed by rFPV. The subsequent experimental results showed that this approach produced robust cell-mediated and humoral immune responses against OVA that, importantly, were accompanied by weak anti-viral vector antibody responses. These results, therefore, represent a novel and potentially clinically applicable way to achieve broadly based and effective immunity to the antigens encoded by vectored vaccines.
Collapse
|