1
|
Parish LA, Rele S, Hofmeyer KA, Luck BB, Wolfe DN. Strategic and Technical Considerations in Manufacturing Viral Vector Vaccines for the Biomedical Advanced Research and Development Authority Threats. Vaccines (Basel) 2025; 13:73. [PMID: 39852852 PMCID: PMC11769106 DOI: 10.3390/vaccines13010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/23/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Over the past few decades, the world has seen a considerable uptick in the number of new and emerging infectious disease outbreaks. The development of new vaccines, vaccine technologies, and platforms are critical to enhance our preparedness for biological threats and prevent future pandemics. Viral vectors can be an important tool in the repertoire of technologies available to develop effective vaccines against new and emerging infectious diseases. In many instances, vaccines may be needed in a reactive scenario, requiring technologies than can elicit rapid and robust immune responses with a single dose. Here, we discuss how viral vector vaccines are utilized in a vaccine portfolio for priority biological threats, some of the challenges in manufacturing viral vector vaccines, the need to strengthen live virus manufacturing capabilities, and future opportunities to capitalize on the use of viral vectors to improve the sustainability of the Biomedical Advanced Research and Development Authority's vaccine portfolio.
Collapse
Affiliation(s)
- Lindsay A. Parish
- CBRN Vaccines, Biomedical Advanced Research & Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC 20201, USA
| | - Shyam Rele
- Division of Research, Innovation, and Ventures (DRIVe), Biomedical Advanced Research & Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC 20201, USA
| | - Kimberly A. Hofmeyer
- CBRN Vaccines, Biomedical Advanced Research & Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC 20201, USA
| | - Brooke B. Luck
- Pharmaceutical Countermeasures Infrastructure (PCI) Division, Biomedical Advanced Research & Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC 20201, USA
| | - Daniel N. Wolfe
- CBRN, Biomedical Advanced Research & Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC 20201, USA
| |
Collapse
|
2
|
Abdelmageed AA, Dewhurst S, Ferran MC. Employing the Oncolytic Vesicular Stomatitis Virus in Cancer Virotherapy: Resistance and Clinical Considerations. Viruses 2024; 17:16. [PMID: 39861805 PMCID: PMC11768927 DOI: 10.3390/v17010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Vesicular Stomatitis Virus (VSV) has emerged as a promising candidate for various clinical applications, including vaccine development, virus pseudotyping, and gene delivery. Its broad host range, ease of propagation, and lack of pre-existing immunity in humans make it ideal for therapeutic use. VSV's potential as an oncolytic virus has garnered attention; however, resistance to VSV-mediated oncolysis has been observed in some cell lines and tumor types, limiting its effectiveness. This review provides a detailed analysis of recent advances in VSV-based oncolysis, focusing on resistance mechanisms such as sustained type-I IFN signaling, upregulation of ISGs, immune cell activation, the tumor microenvironment (TME), and tumor-intrinsic factors. Strategies to overcome resistance include enhancing viral oncoselectivity, inhibiting IFN responses, modulating the TME, and combining VSV with chemotherapies, radiation, and immune checkpoint inhibitors. Several VSV-based phase I/II clinical trials show promise; however, addressing resistance and developing novel strategies to enhance therapeutic efficacy are essential for realizing the full potential of VSV oncolytic virotherapy. Future research should focus on patient-specific approaches, as tumor heterogeneity implies varying resistance mechanisms. Personalized treatments tailored to tumor molecular profiles, along with identifying biomarkers predictive of resistance to VSV oncolysis, will enhance patient selection and enable more effective, individualized VSV-based therapies.
Collapse
Affiliation(s)
- Alaa A. Abdelmageed
- Biomedical Genetics and Genomics Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; (A.A.A.); (S.D.)
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Stephen Dewhurst
- Biomedical Genetics and Genomics Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; (A.A.A.); (S.D.)
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Maureen C. Ferran
- Thomas H. Gosnell School for Life Sciences, Rochester Institute of Technology, Rochester, NY 14623, USA
| |
Collapse
|
3
|
Gholap AD, Gupta J, Kamandar P, Bhowmik DD, Rojekar S, Faiyazuddin M, Hatvate NT, Mohanto S, Ahmed MG, Subramaniyan V, Kumarasamy V. Harnessing Nanovaccines for Effective Immunization─A Special Concern on COVID-19: Facts, Fidelity, and Future Prospective. ACS Biomater Sci Eng 2024; 10:271-297. [PMID: 38096426 DOI: 10.1021/acsbiomaterials.3c01247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Nanotechnology has emerged as a transformative pathway in vaccine research and delivery. Nanovaccines, encompassing lipid and nonlipid formulations, exhibit considerable advantages over traditional vaccine techniques, including enhanced antigen stability, heightened immunogenicity, targeted distribution, and the potential for codelivery with adjuvants or immune modulators. This review provides a comprehensive overview of the latest advancements and applications of lipid and non-lipid-based nanovaccines in current vaccination strategies for immunization. The review commences by outlining the fundamental concepts underlying lipid and nonlipid nanovaccine design before delving into the diverse components and production processes employed in their development. Subsequently, a comparative analysis of various nanocarriers is presented, elucidating their distinct physicochemical characteristics and impact on the immune response, along with preclinical and clinical studies. The discussion also highlights how nanotechnology enables the possibility of personalized and combined vaccination techniques, facilitating the creation of tailored nanovaccines to meet the individual patient needs. The ethical aspects concerning the use of nanovaccines, as well as potential safety concerns and public perception, are also addressed. The study underscores the gaps and challenges that must be overcome before adopting nanovaccines in clinical practice. This comprehensive analysis offers vital new insights into lipid and nonlipid nanovaccine status. It emphasizes the significance of continuous research, collaboration among interdisciplinary experts, and regulatory measures to fully unlock the potential of nanotechnology in enhancing immunization and ensuring a healthier, more resilient society.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Juhi Gupta
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Pallavi Kamandar
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Deblina D Bhowmik
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Md Faiyazuddin
- Department of Pharmaceutics, School of Pharmacy, Al-Karim University, Katihar 854106, Bihar, India
| | - Navnath T Hatvate
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Beitari S, Agbayani G, Hewitt M, Duque D, Bavananthasivam J, Sandhu JK, Akache B, Hadžisejdić I, Tran A. Effectiveness of VSV vectored SARS-CoV-2 spike when administered through intranasal, intramuscular or a combination of both. Sci Rep 2023; 13:21390. [PMID: 38049498 PMCID: PMC10695950 DOI: 10.1038/s41598-023-48397-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/26/2023] [Indexed: 12/06/2023] Open
Abstract
A critical feature of the VSV vector platform is the ability to pseudotype the virus with different glycoproteins from other viruses, thus altering cellular tropism of the recombinant virus. The route of administration is critical in triggering local and systemic immune response and protection. Most of the vaccine platforms used at the forefront are administered by intramuscular injection. However, it is not known at what level ACE2 is expressed on the surface of skeletal muscle cells, which will have a significant impact on the efficiency of a VSV-SARS-CoV-2 spike vaccine to mount a protective immune response when administered intramuscularly. In this study, we investigate the immunogenicity and efficacy of a prime-boost immunization regimen administered intranasally (IN), intramuscularly (IM), or combinations of the two. We determined that the prime-boost combinations of IM followed by IN immunization (IM + IN) or IN followed by IN immunization (IN + IN) exhibited strong spike-specific IgG, IgA and T cell response in vaccinated K18 knock-in mice. Hamsters vaccinated with two doses of VSV expressing SARS-CoV-2 spike, both delivered by IN or IM + IN, showed strong protection against SARS-CoV-2 variants of concern Alpha and Delta. This protection was also observed in aged hamsters. Our study underscores the highly crucial role immunization routes have with the VSV vector platform to elicit a strong and protective immune response.
Collapse
Affiliation(s)
- Saina Beitari
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Gerard Agbayani
- Immunomodulation, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Melissa Hewitt
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Diana Duque
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jegarubee Bavananthasivam
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jagdeep K Sandhu
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Bassel Akache
- Immunomodulation, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Ita Hadžisejdić
- Clinical Department of Pathology and Cytology Clinical Hospital Center Rijeka, University of Rijeka, Rijeka, Croatia
| | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada.
| |
Collapse
|
5
|
Bakhshizadeh Gashti A, Chahal PS, Gaillet B, Garnier A. Purification of recombinant vesicular stomatitis virus-based HIV vaccine candidate. Vaccine 2023; 41:2198-2207. [PMID: 36842887 DOI: 10.1016/j.vaccine.2023.02.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/10/2023] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
In this work, laboratory- and large-scale methods were tested for purification of a human immunodeficiency virus (HIV) vaccine candidate, based on recombinant vesicular stomatitis virus (rVSV). First step of the purification, the clarification of the rVSVs produced in serum-free cell culture medium, was tested by centrifugation and filtration using different filtration media and pore sizes (0.45 to 30 µm). To reduce the supernatant volume and process time, the clarified sample was concentrated by ultrafiltration either using tangential flow filtration or centrifugal-based filtration units, depending on the process scale. The final purification step at laboratory-scale, was carried out by density gradient ultracentrifugation, the recovery of which was compared with chromatographic purification at large-scale. The virus preparations were analyzed using dynamic light scattering to verify the virus size and transmission electron microscopy for purity and virus morphology. Density gradient ultracentrifugation allowed the recovery of ≥ 80% infectious particles and reduced the contaminant DNA and host cell proteins relatively to standard ultracentrifugation pelleting using a sucrose cushion. At large-scale, weak and strong anion-exchangers were tested and compared. The best columns allowed infectious virus recoveries as high as 77% and eliminated 92% of host cell proteins.
Collapse
Affiliation(s)
- Anahita Bakhshizadeh Gashti
- Department of Chemical Engineering, Faculty of Sciences and Engineering, Université Laval, Quebec, QC, Canada; Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Parminder S Chahal
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Bruno Gaillet
- Department of Chemical Engineering, Faculty of Sciences and Engineering, Université Laval, Quebec, QC, Canada
| | - Alain Garnier
- Department of Chemical Engineering, Faculty of Sciences and Engineering, Université Laval, Quebec, QC, Canada.
| |
Collapse
|
6
|
Nonclinical pharmacokinetics and biodistribution of VSV-GP using methods to decouple input drug disposition and viral replication. Mol Ther Methods Clin Dev 2022; 28:190-207. [PMID: 36700123 PMCID: PMC9843450 DOI: 10.1016/j.omtm.2022.12.013] [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: 07/19/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Viral replication places oncolytic viruses (OVs) in a unique niche in the field of drug pharmacokinetics (PK) as their self-amplification obscures exposure-response relationships. Moreover, standard bioanalytical techniques are unable to distinguish the input from replicated drug products. Here, we combine two novel approaches to characterize PK and biodistribution (BD) after systemic administration of vesicular stomatitis virus pseudotyped with lymphocytic choriomeningitis virus glycoprotein (VSV-GP) in healthy mice. First: to decouple input drug PK/BD versus replication PK/BD, we developed and fully characterized a replication-incompetent tool virus that retained all other critical attributes of the drug. We used this approach to quantify replication in blood and tissues and to determine its impact on PK and BD. Second: to discriminate the genomic and antigenomic viral RNA strands contributing to replication dynamics in tissues, we developed an in situ hybridization method using strand-specific probes and assessed their spatiotemporal distribution in tissues. This latter approach demonstrated that distribution, transcription, and replication localized to tissue-resident macrophages, indicating their role in PK and BD. Ultimately, our study results in a refined PK/BD profile for a replicating OV, new proposed PK parameters, and deeper understanding of OV PK/BD using unique approaches that could be applied to other replicating vectors.
Collapse
|
7
|
Zhang R, Zhang XF, Chi Y, Xu Y, Chen H, Guo Z, Wei T. Nucleoprotein of a Rice Rhabdovirus Serves as the Effector to Attenuate Hemolymph Melanization and Facilitate Viral Persistent Propagation in its Leafhopper Vector. Front Immunol 2022; 13:904244. [PMID: 35655780 PMCID: PMC9152149 DOI: 10.3389/fimmu.2022.904244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Melanization in the hemolymph of arthropods is a conserved defense strategy against infection by invading pathogens. Numerous plant viruses are persistently transmitted by insect vectors, and must overcome hemolymph melanization. Here, we determine that the plant rhabdovirus rice stripe mosaic virus (RSMV) has evolved to evade the antiviral melanization response in the hemolymph in leafhopepr vectors. After virions enter vector hemolymph cells, viral nucleoprotein N is initially synthesized and directly interacts with prophenoloxidase (PPO), a core component of the melanization pathway and this process strongly activates the expression of PPO. Furthermore, such interaction could effectively inhibit the proteolytic cleavage of the zymogen PPO to active phenoloxidase (PO), finally suppressing hemolymph melanization. The knockdown of PPO expression or treatment with the PO inhibitor also suppresses hemolymph melanization and causes viral excessive accumulation, finally causing a high insect mortality rate. Consistent with this function, microinjection of N into leafhopper vectors attenuates melanization and promotes viral infection. These findings demonstrate that RSMV N serves as the effector to attenuate hemolymph melanization and facilitate viral persistent propagation in its insect vector. Our findings provide the insights in the understanding of ongoing arms race of insect immunity defense and viral counter-defense.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Taiyun Wei
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
8
|
Lundstrom K. Self-replicating vehicles based on negative strand RNA viruses. Cancer Gene Ther 2022:10.1038/s41417-022-00436-7. [PMID: 35169298 PMCID: PMC8853047 DOI: 10.1038/s41417-022-00436-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 11/10/2022]
Abstract
Self-replicating RNA viruses have been engineered as efficient expression vectors for vaccine development for infectious diseases and cancers. Moreover, self-replicating RNA viral vectors, particularly oncolytic viruses, have been applied for cancer therapy and immunotherapy. Among negative strand RNA viruses, measles viruses and rhabdoviruses have been frequently applied for vaccine development against viruses such as Chikungunya virus, Lassa virus, Ebola virus, influenza virus, HIV, Zika virus, and coronaviruses. Immunization of rodents and primates has elicited strong neutralizing antibody responses and provided protection against lethal challenges with pathogenic viruses. Several clinical trials have been conducted. Ervebo, a vaccine based on a vesicular stomatitis virus (VSV) vector has been approved for immunization of humans against Ebola virus. Different types of cancers such as brain, breast, cervical, lung, leukemia/lymphoma, ovarian, prostate, pancreatic, and melanoma, have been the targets for cancer vaccine development, cancer gene therapy, and cancer immunotherapy. Administration of measles virus and VSV vectors have demonstrated immune responses, tumor regression, and tumor eradication in various animal models. A limited number of clinical trials have shown well-tolerated treatment, good safety profiles, and dose-dependent activity in cancer patients.
Collapse
|
9
|
Mangion M, Gélinas JF, Bakhshi Zadeh Gashti A, Azizi H, Kiesslich S, Nassoury N, Chahal PS, Kobinger G, Gilbert R, Garnier A, Gaillet B, Kamen A. Evaluation of novel HIV vaccine candidates using recombinant vesicular stomatitis virus vector produced in serum-free Vero cell cultures. Vaccine 2020; 38:7949-7955. [PMID: 33139138 DOI: 10.1016/j.vaccine.2020.10.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/09/2020] [Accepted: 10/18/2020] [Indexed: 12/20/2022]
Abstract
Acquired Immune Deficiency Syndrome (AIDS) in humans is a result of the destruction of the immune system caused by Human Immunodeficiency Virus (HIV) infection. This serious epidemic is still progressing world-wide. Despite advances in treatment, a safe and effective preventive HIV vaccine is desired to combat this disease, and to save millions of lives. However, such a vaccine is not available yet although extensive amounts of resources in research and development have been invested over three decades. In light of the recently approved Ebola virus disease vaccine based on a recombinant vesicular stomatitis virus (rVSV-ZEBOV), we present the results of our work on three novel VSV-vectored HIV vaccine candidates. We describe the design, rescue, production and purification method and evaluate their immunogenicity in mice prior to preclinical studies that will be performed in non-human primates. The production of each of the three candidate vaccines (rVSV-B6-NL4.3Env/SIVtm, rVSV-B6-NL4.3Env/Ebtm and rVSV-B6-A74Env(PN6)/SIVtm) was evaluated in small scale in Vero cells and it was found that production kinetics on Vero cells vary depending on the HIV gp surface protein used. Purified virus preparations complied with the WHO restrictions for the residual DNA and host cell protein contents. Finally, when administered to mice, all three rVSV-HIV vaccine candidates induced an HIV gp140-specific antibody response.
Collapse
Affiliation(s)
- Mathias Mangion
- Département de génie chimique, Université Laval, Québec, QC, Canada
| | | | | | - Hiva Azizi
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Quebec, QC, Canada
| | - Sascha Kiesslich
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Nasha Nassoury
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Parminder S Chahal
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Gary Kobinger
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Quebec, QC, Canada
| | - Rénald Gilbert
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Alain Garnier
- Département de génie chimique, Université Laval, Québec, QC, Canada
| | - Bruno Gaillet
- Département de génie chimique, Université Laval, Québec, QC, Canada
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, QC, Canada.
| |
Collapse
|
10
|
Munis AM, Bentley EM, Takeuchi Y. A tool with many applications: vesicular stomatitis virus in research and medicine. Expert Opin Biol Ther 2020; 20:1187-1201. [PMID: 32602788 DOI: 10.1080/14712598.2020.1787981] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Vesicular stomatitis virus (VSV) has long been a useful research tool in virology and recently become an essential part of medicinal products. Vesiculovirus research is growing quickly following its adaptation to clinical gene and cell therapy and oncolytic virotherapy. AREAS COVERED This article reviews the versatility of VSV as a research tool and biological reagent, its use as a viral and vaccine vector delivering therapeutic and immunogenic transgenes and an oncolytic virus aiding cancer treatment. Challenges such as the immune response against such advanced therapeutic medicinal products and manufacturing constraints are also discussed. EXPERT OPINION The field of in vivo gene and cell therapy is advancing rapidly with VSV used in many ways. Comparison of VSV's use as a versatile therapeutic reagent unveils further prospects and problems for each application. Overcoming immunological challenges to aid repeated administration of viral vectors and minimizing harmful host-vector interactions remains one of the major challenges. In the future, exploitation of reverse genetic tools may assist the creation of recombinant viral variants that have improved onco-selectivity and more efficient vaccine vector activity. This will add to the preferential features of VSV as an excellent advanced therapy medicinal product (ATMP) platform.
Collapse
Affiliation(s)
- Altar M Munis
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford , Oxford, UK.,Division of Advanced Therapies, National Institute for Biological Standards and Control , South Mimms, UK
| | - Emma M Bentley
- Division of Virology, National Institute for Biological Standards and Control , South Mimms, UK
| | - Yasuhiro Takeuchi
- Division of Advanced Therapies, National Institute for Biological Standards and Control , South Mimms, UK.,Division of Infection and Immunity, University College London , London, UK
| |
Collapse
|
11
|
Gélinas JF, Azizi H, Kiesslich S, Lanthier S, Perdersen J, Chahal PS, Ansorge S, Kobinger G, Gilbert R, Kamen AA. Production of rVSV-ZEBOV in serum-free suspension culture of HEK 293SF cells. Vaccine 2019; 37:6624-6632. [DOI: 10.1016/j.vaccine.2019.09.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022]
|
12
|
Affiliation(s)
| | - Jerome H. Kim
- International Vaccine Institute, Seoul, Republic of Korea
| |
Collapse
|
13
|
Heterologous prime-boost immunization with vesiculovirus-based vectors expressing HBV Core antigen induces CD8 + T cell responses in naïve and persistently infected mice and protects from challenge. Antiviral Res 2019; 168:156-167. [PMID: 31153968 DOI: 10.1016/j.antiviral.2019.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 01/05/2023]
Abstract
Chronic hepatitis B virus (HBV) infections cause more than 800,000 deaths per year and currently approved treatments do not cure the disease. Because a hallmark of acute infection resolution is the presence of functional CD8+ T cells to the virus, activation of the immune system with therapeutic vaccines represents a potential approach for treating chronic hepatitis B. In this study, we evaluated the immunogenicity and efficacy of two attenuated vesiculovirus-based platforms expressing HBV Core antigen, the highly attenuated vesicular stomatitis virus (VSV) N4CT1 and a unique vaccine platform [virus-like vesicles (VLV)] that is based on a Semliki Forest virus replicon expressing the VSV glycoprotein. We found that heterologous prime-boost immunization with VLV and N4CT1 induced Core-specific CD8+ T cell responses in naïve mice. When immunized mice were later challenged with AAV-HBV, functional Core-specific CD8+ T cells were present in the liver, and mice were protected from establishment of persistent infection. In contrast, when mice with pre-established persistent HBV replication received prime-boost immunization, functional Core-specific CD8+ T cells were found in the spleen but not in the liver. These results highlight the importance of investigating the therapeutic value of different HBV antigens alone and in combination using preclinical animal models, and understanding the correlation between anti-HBV efficacy in these models with human infection.
Collapse
|
14
|
A Highly Attenuated Vesicular Stomatitis Virus-Based Vaccine Platform Controls Hepatitis B Virus Replication in Mouse Models of Hepatitis B. J Virol 2019; 93:JVI.01586-18. [PMID: 30541859 DOI: 10.1128/jvi.01586-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
Therapeutic vaccines may be an important component of a treatment regimen for curing chronic hepatitis B virus (HBV) infection. We previously demonstrated that recombinant wild-type vesicular stomatitis virus (VSV) expressing the HBV middle surface glycoprotein (MHBs) elicits functional immune responses in mouse models of HBV replication. However, VSV has some undesirable pathogenic properties, and the use of this platform in humans requires further viral attenuation. We therefore generated a highly attenuated VSV that expresses MHBs and contains two attenuating mutations. This vector was evaluated for immunogenicity, pathogenesis, and anti-HBV function in mice. Compared to wild-type VSV, the highly attenuated virus displayed markedly reduced pathogenesis but induced similar MHBs-specific CD8+ T cell and antibody responses. The CD8+ T cell responses elicited by this vector in naive mice prevented HBV replication in animals that were later challenged by hydrodynamic injection or transduction with adeno-associated virus encoding the HBV genome (AAV-HBV). In mice in which persistent HBV replication was first established by AAV-HBV transduction, subsequent immunization with the attenuated VSV induced MHBs-specific CD8+ T cell responses that corresponded with reductions in serum and liver HBV antigens and nucleic acids. HBV control was associated with an increase in the frequency of intrahepatic HBV-specific CD8+ T cells and a transient elevation in serum alanine aminotransferase activity. The ability of VSV to induce a robust multispecific T cell response that controls HBV replication combined with the improved safety profile of the highly attenuated vector suggests that this platform offers a new approach for HBV therapeutic vaccination.IMPORTANCE A curative treatment for chronic hepatitis B must eliminate the virus from the liver, but current antiviral therapies typically fail to do so. Immune-mediated resolution of infection occurs in a small fraction of chronic HBV patients, which suggests the potential efficacy of therapeutic strategies that boost the patient's own immune response to the virus. We modified a safe form of VSV to express an immunogenic HBV protein and evaluated the efficacy of this vector in the prevention and treatment of HBV infection in mouse models. Our results show that this vector elicits HBV-specific immune responses that prevent the establishment of HBV infection and reduce viral proteins in the serum and viral DNA/RNA in the liver of mice with persistent HBV replication. These findings suggest that highly attenuated and safe virus-based vaccine platforms have the potential to be utilized for the development of an effective therapeutic vaccine against chronic HBV infection.
Collapse
|
15
|
Elizaga ML, Li SS, Kochar NK, Wilson GJ, Allen MA, Tieu HVN, Frank I, Sobieszczyk ME, Cohen KW, Sanchez B, Latham TE, Clarke DK, Egan MA, Eldridge JH, Hannaman D, Xu R, Ota-Setlik A, McElrath MJ, Hay CM, on behalf of the NIAID HIV Vaccine Trials Network (HVTN) 087 Study Team. Safety and tolerability of HIV-1 multiantigen pDNA vaccine given with IL-12 plasmid DNA via electroporation, boosted with a recombinant vesicular stomatitis virus HIV Gag vaccine in healthy volunteers in a randomized, controlled clinical trial. PLoS One 2018; 13:e0202753. [PMID: 30235286 PMCID: PMC6147413 DOI: 10.1371/journal.pone.0202753] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The addition of plasmid cytokine adjuvants, electroporation, and live attenuated viral vectors may further optimize immune responses to DNA vaccines in heterologous prime-boost combinations. The objective of this study was to test the safety and tolerability of a novel prime-boost vaccine regimen incorporating these strategies with different doses of IL-12 plasmid DNA adjuvant. METHODS In a phase 1 study, 88 participants received an HIV-1 multiantigen (gag/pol, env, nef/tat/vif) DNA vaccine (HIV-MAG, 3000 μg) co-administered with IL-12 plasmid DNA adjuvant at 0, 250, 1000, or 1500 μg (N = 22/group) given intramuscularly with electroporation (Ichor TriGrid™ Delivery System device) at 0, 1 and 3 months; followed by attenuated recombinant vesicular stomatitis virus, serotype Indiana, expressing HIV-1 Gag (VSV-Gag), 3.4 ⊆ 107 plaque-forming units (PFU), at 6 months; 12 others received placebo. Injections were in both deltoids at each timepoint. Participants were monitored for safety and tolerability for 15 months. RESULTS The dose of IL-12 pDNA did not increase pain scores, reactogenicity, or adverse events with the co-administered DNA vaccine, or following the VSV-Gag boost. Injection site pain and reactogenicity were common with intramuscular injections with electroporation, but acceptable to most participants. VSV-Gag vaccine often caused systemic reactogenicity symptoms, including a viral syndrome (in 41%) of fever, chills, malaise/fatigue, myalgia, and headache; and decreased lymphocyte counts 1 day after vaccination. CONCLUSIONS HIV-MAG DNA vaccine given by intramuscular injection with electroporation was safe at all doses of IL-12 pDNA. The VSV-Gag vaccine at this dose was associated with fever and viral symptoms in some participants, but the vaccine regimens were safe and generally well-tolerated. TRIAL REGISTRATION Clinical Trials.gov NCT01578889.
Collapse
Affiliation(s)
- Marnie L. Elizaga
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Shuying S. Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Nidhi K. Kochar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Gregory J. Wilson
- Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Mary A. Allen
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hong Van N. Tieu
- Laboratory of Infectious Disease Prevention, New York Blood Center, New York, New York, United States of America
| | - Ian Frank
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Magdalena E. Sobieszczyk
- Division of Infectious Diseases, Columbia University Medical Center, New York, New York, United States of America
| | - Kristen W. Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Brittany Sanchez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Theresa E. Latham
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - David K. Clarke
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Michael A. Egan
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - John H. Eldridge
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Drew Hannaman
- Ichor Medical Systems, Incorporated, San Diego, California, United States of America
| | - Rong Xu
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Ayuko Ota-Setlik
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Christine Mhorag Hay
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, New York, United States of America
| | | |
Collapse
|
16
|
Zhang X, Mao G, van den Pol AN. Chikungunya-vesicular stomatitis chimeric virus targets and eliminates brain tumors. Virology 2018; 522:244-259. [PMID: 30055515 DOI: 10.1016/j.virol.2018.06.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/26/2018] [Accepted: 06/28/2018] [Indexed: 01/17/2023]
Abstract
Vesicular stomatitis virus (VSV) shows potential for targeting and killing cancer cells, but can be dangerous in the brain due to its neurotropic glycoprotein. Here we test a chimeric virus in which the VSV glycoprotein is replaced with the Chikungunya polyprotein E3-E2-6K-E1 (VSVΔG-CHIKV). Control mice with brain tumors survived a mean of 40 days after tumor implant. VSVΔG-CHIKV selectively infected and eliminated the tumor, and extended survival substantially in all tumor-bearing mice to over 100 days. VSVΔG-CHIKV also targeted intracranial primary patient derived melanoma xenografts. Virus injected into one melanoma spread to other melanomas within the same brain with little detectable infection of normal cells. Intravenous VSVΔG-CHIKV infected tumor cells but not normal tissue. In immunocompetent mice, VSVΔG-CHIKV selectively infected mouse melanoma cells within the brain. These data suggest VSVΔG-CHIKV can target and destroy brain tumors in multiple animal models without the neurotropism associated with the wild type VSV glycoprotein.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06520, United States
| | - Guochao Mao
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06520, United States
| | - Anthony N van den Pol
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06520, United States.
| |
Collapse
|
17
|
Lundstrom K. Viral Vectors in Gene Therapy. Diseases 2018; 6:diseases6020042. [PMID: 29883422 PMCID: PMC6023384 DOI: 10.3390/diseases6020042] [Citation(s) in RCA: 310] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 01/02/2023] Open
Abstract
Applications of viral vectors have found an encouraging new beginning in gene therapy in recent years. Significant improvements in vector engineering, delivery, and safety have placed viral vector-based therapy at the forefront of modern medicine. Viral vectors have been employed for the treatment of various diseases such as metabolic, cardiovascular, muscular, hematologic, ophthalmologic, and infectious diseases and different types of cancer. Recent development in the area of immunotherapy has provided both preventive and therapeutic approaches. Furthermore, gene silencing generating a reversible effect has become an interesting alternative, and is well-suited for delivery by viral vectors. A number of preclinical studies have demonstrated therapeutic and prophylactic efficacy in animal models and furthermore in clinical trials. Several viral vector-based drugs have also been globally approved.
Collapse
|
18
|
Chen Z. Parainfluenza virus 5-vectored vaccines against human and animal infectious diseases. Rev Med Virol 2018; 28. [PMID: 29316047 PMCID: PMC7169218 DOI: 10.1002/rmv.1965] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/25/2022]
Abstract
Parainfluenza virus 5 (PIV5), known as canine parainfluenza virus in the veterinary field, is a negative‐sense, nonsegmented, single‐stranded RNA virus belonging to the Paramyxoviridae family. Parainfluenza virus 5 is an excellent viral vector and has been used as a live vaccine for kennel cough for many years in dogs without any safety concern. It can grow to high titers in many cell types, and its genome is stable even in the presence of foreign gene insertions. So far, PIV5 has been used to develop vaccines against influenza virus, respiratory syncytial virus, rabies virus, and Mycobacterium tuberculosis, demonstrating its ability to elicit robust and protective immune responses in preclinical animal models. Parainfluenza virus 5–based vaccines can be administered intranasally, intramuscularly, or orally. Interestingly, prior exposure of PIV5 does not prevent a PIV5‐vectored vaccine from generating robust immunity, indicating that the vector can be used more than once. Here, these encouraging results are reviewed together along with discussion of the desirable advantages of the PIV5 vaccine vector to aid future vaccine design and to accelerate progression of PIV5‐based vaccines into clinical trials.
Collapse
Affiliation(s)
- Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, JS, China
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW The purpose is to review recent novel approaches in HIV vaccine research and development being undertaken in the preclinical and early clinical space, as well as related and novel nonvaccine approaches such as genetic delivery of broadly neutralizing antibodies for protection from HIV infection and AIDS. RECENT FINDINGS We review novel HIV envelope immunogen design, including native trimer and germline targeting approaches as well as genetic delivery of broadly neutralizing antibodies and replicating vector vaccinesSUMMARY: Despite 30+ years of research and development, and billions of dollars spent, a well tolerated and effective HIV vaccine remains a public health priority for any chance of ending the AIDS pandemic. It has become very clear that significant investments in novel technologies, innovation, and multidisciplinary science will be necessary to accelerate progress.
Collapse
|
20
|
Chikungunya, Influenza, Nipah, and Semliki Forest Chimeric Viruses with Vesicular Stomatitis Virus: Actions in the Brain. J Virol 2017; 91:JVI.02154-16. [PMID: 28077641 DOI: 10.1128/jvi.02154-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/02/2017] [Indexed: 02/02/2023] Open
Abstract
Recombinant vesicular stomatitis virus (VSV)-based chimeric viruses that include genes from other viruses show promise as vaccines and oncolytic viruses. However, the critical safety concern is the neurotropic nature conveyed by the VSV glycoprotein. VSVs that include the VSV glycoprotein (G) gene, even in most recombinant attenuated strains, can still show substantial adverse or lethal actions in the brain. Here, we test 4 chimeric viruses in the brain, including those in which glycoprotein genes from Nipah, chikungunya (CHIKV), and influenza H5N1 viruses were substituted for the VSV glycoprotein gene. We also test a virus-like vesicle (VLV) in which the VSV glycoprotein gene is expressed from a replicon encoding the nonstructural proteins of Semliki Forest virus. VSVΔG-CHIKV, VSVΔG-H5N1, and VLV were all safe in the adult mouse brain, as were VSVΔG viruses expressing either the Nipah F or G glycoprotein. In contrast, a complementing pair of VSVΔG viruses expressing Nipah G and F glycoproteins were lethal within the brain within a surprisingly short time frame of 2 days. Intranasal inoculation in postnatal day 14 mice with VSVΔG-CHIKV or VLV evoked no adverse response, whereas VSVΔG-H5N1 by this route was lethal in most mice. A key immune mechanism underlying the safety of VSVΔG-CHIKV, VSVΔG-H5N1, and VLV in the adult brain was the type I interferon response; all three viruses were lethal in the brains of adult mice lacking the interferon receptor, suggesting that the viruses can infect and replicate and spread in brain cells if not blocked by interferon-stimulated genes within the brain.IMPORTANCE Vesicular stomatitis virus (VSV) shows considerable promise both as a vaccine vector and as an oncolytic virus. The greatest limitation of VSV is that it is highly neurotropic and can be lethal within the brain. The neurotropism can be mostly attributed to the VSV G glycoprotein. Here, we test 4 chimeric viruses of VSV with glycoprotein genes from Nipah, chikungunya, and influenza viruses and nonstructural genes from Semliki Forest virus. Two of the four, VSVΔG-CHIKV and VLV, show substantially attenuated neurotropism and were safe in the healthy adult mouse brain. VSVΔG-H5N1 was safe in the adult brain but lethal in the younger brain. VSVΔG Nipah F+G was even more neurotropic than wild-type VSV, evoking a rapid lethal response in the adult brain. These results suggest that while chimeric VSVs show promise, each must be tested with both intranasal and intracranial administration to ensure the absence of lethal neurotropism.
Collapse
|
21
|
Clarke DK, Hendry RM, Singh V, Rose JK, Seligman SJ, Klug B, Kochhar S, Mac LM, Carbery B, Chen RT. Live virus vaccines based on a vesicular stomatitis virus (VSV) backbone: Standardized template with key considerations for a risk/benefit assessment. Vaccine 2016; 34:6597-6609. [PMID: 27395563 PMCID: PMC5220644 DOI: 10.1016/j.vaccine.2016.06.071] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 12/30/2022]
Abstract
The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety of live, recombinant viral vaccines incorporating genes from heterologous viral and other microbial pathogens in their genome (so-called "chimeric virus vaccines"). Many such viral vector vaccines are now at various stages of clinical evaluation. Here, we introduce an attenuated form of recombinant vesicular stomatitis virus (rVSV) as a potential chimeric virus vaccine for HIV-1, with implications for use as a vaccine vector for other pathogens. The rVSV/HIV-1 vaccine vector was attenuated by combining two major genome modifications. These modifications acted synergistically to greatly enhance vector attenuation and the resulting rVSV vector demonstrated safety in sensitive mouse and non-human primate neurovirulence models. This vector expressing HIV-1 gag protein has completed evaluation in two Phase I clinical trials. In one trial the rVSV/HIV-1 vector was administered in a homologous two-dose regimen, and in a second trial with pDNA in a heterologous prime boost regimen. No serious adverse events were reported nor was vector detected in blood, urine or saliva post vaccination in either trial. Gag specific immune responses were induced in both trials with highest frequency T cell responses detected in the prime boost regimen. The rVSV/HIV-1 vector also demonstrated safety in an ongoing Phase I trial in HIV-1 positive participants. Additionally, clinical trial material has been produced with the rVSV vector expressing HIV-1 env, and Phase I clinical evaluation will initiate in the beginning of 2016. In this paper, we use a standardized template describing key characteristics of the novel rVSV vaccine vectors, in comparison to wild type VSV. The template facilitates scientific discourse among key stakeholders by increasing transparency and comparability of information. The Brighton Collaboration V3SWG template may also be useful as a guide to the evaluation of other recombinant viral vector vaccines.
Collapse
MESH Headings
- AIDS Vaccines/adverse effects
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- Clinical Trials, Phase I as Topic
- Drug Carriers
- Drug Evaluation, Preclinical
- Drug-Related Side Effects and Adverse Reactions/epidemiology
- Drug-Related Side Effects and Adverse Reactions/pathology
- Genetic Vectors
- Humans
- Primates
- Risk Assessment
- T-Lymphocytes/immunology
- Vaccines, Attenuated/adverse effects
- Vaccines, Attenuated/genetics
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vesiculovirus/genetics
- env Gene Products, Human Immunodeficiency Virus/genetics
- env Gene Products, Human Immunodeficiency Virus/immunology
- gag Gene Products, Human Immunodeficiency Virus/genetics
- gag Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
| | - R Michael Hendry
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention (NCHHSTP), Centers for Disease Control and Prevention (CDC), Atlanta, GA 30333, USA
| | - Vidisha Singh
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention (NCHHSTP), Centers for Disease Control and Prevention (CDC), Atlanta, GA 30333, USA.
| | - John K Rose
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stephen J Seligman
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | | | | | - Lisa Marie Mac
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention (NCHHSTP), Centers for Disease Control and Prevention (CDC), Atlanta, GA 30333, USA
| | - Baevin Carbery
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention (NCHHSTP), Centers for Disease Control and Prevention (CDC), Atlanta, GA 30333, USA
| | - Robert T Chen
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention (NCHHSTP), Centers for Disease Control and Prevention (CDC), Atlanta, GA 30333, USA
| |
Collapse
|
22
|
Fuchs JD, Frank I, Elizaga ML, Allen M, Frahm N, Kochar N, Li S, Edupuganti S, Kalams SA, Tomaras GD, Sheets R, Pensiero M, Tremblay MA, Higgins TJ, Latham T, Egan MA, Clarke DK, Eldridge JH. First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Vesicular Stomatitis Virus Human Immunodeficiency Virus-1 gag Vaccine (HVTN 090). Open Forum Infect Dis 2015; 2:ofv082. [PMID: 26199949 PMCID: PMC4504730 DOI: 10.1093/ofid/ofv082] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/01/2015] [Indexed: 01/17/2023] Open
Abstract
Background. We report the first-in-human safety and immunogenicity evaluation of a highly attenuated, replication-competent recombinant vesicular stomatitis virus (rVSV) human immunodeficiency virus (HIV)-1 vaccine. Methods. Sixty healthy, HIV-1-uninfected adults were enrolled in a randomized, double-blinded, placebo-controlled dose-escalation study. Groups of 12 participants received rVSV HIV-1 gag vaccine at 5 dose levels (4.6 × 10(3) to 3.4 × 10(7) particle forming units) (N = 10/group) or placebo (N = 2/group), delivered intramuscularly as bilateral injections at 0 and 2 months. Safety monitoring included VSV cultures from blood, urine, saliva, and swabs of oral lesions. Vesicular stomatitis virus-neutralizing antibodies, T-cell immunogenicity, and HIV-1 specific binding antibodies were assessed. Results. Local and systemic reactogenicity symptoms were mild to moderate and increased with dose. No severe reactogenicity or product-related serious adverse events were reported, and all rVSV cultures were negative. All vaccine recipients became seropositive for VSV after 2 vaccinations. gag-specific T-cell responses were detected in 63% of participants by interferon-γ enzyme-linked immunospot at the highest dose post boost. Conclusions. An attenuated replication-competent rVSV gag vaccine has an acceptable safety profile in healthy adults. This rVSV vector is a promising new vaccine platform for the development of vaccines to combat HIV-1 and other serious human diseases.
Collapse
Affiliation(s)
- Jonathan D. Fuchs
- San Francisco Department of Public Health, California
- University of California, San Francisco
| | - Ian Frank
- University of Pennsylvania, Philadelphia
| | - Marnie L. Elizaga
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Mary Allen
- Division of AIDS, National Institutes of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Nidhi Kochar
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sue Li
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| | - Rebecca Sheets
- Division of AIDS, National Institutes of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Michael Pensiero
- Division of AIDS, National Institutes of Allergy and Infectious Diseases, Bethesda, Maryland
| | | | | | | | | | | | | |
Collapse
|
23
|
STAT3 inhibition reduces toxicity of oncolytic VSV and provides a potentially synergistic combination therapy for hepatocellular carcinoma. Cancer Gene Ther 2015; 22:317-25. [PMID: 25930184 DOI: 10.1038/cgt.2015.23] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 03/27/2015] [Accepted: 03/28/2015] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a refractory malignancy with a high mortality and increasing worldwide incidence rates, including the United States and central Europe. In this study, we demonstrate that a specific inhibitor of signal transducer and activator of transcription 3 (STAT3), NSC74859, efficiently reduces HCC cell proliferation and can be successfully combined with oncolytic virotherapy using vesicular stomatitis virus (VSV). The potential benefits of this combination treatment are strengthened by the ability of NSC74859 to protect primary hepatocytes and nervous system cells against virus-induced cytotoxicity, with an elevation of the VSV maximum tolerated dose in mice. Hereby we propose a strategy for improving the current regimen for HCC treatment and seek to further explore the molecular mechanisms underlying selective oncolytic specificity of VSV.
Collapse
|
24
|
Lorenz IC, Nguyen HT, Kemelman M, Lindsay RW, Yuan M, Wright KJ, Arendt H, Back JW, DeStefano J, Hoffenberg S, Morrow G, Jurgens CK, Phogat SK, Zamb TJ, Parks CL. The stem of vesicular stomatitis virus G can be replaced with the HIV-1 Env membrane-proximal external region without loss of G function or membrane-proximal external region antigenic properties. AIDS Res Hum Retroviruses 2014; 30:1130-44. [PMID: 24597516 DOI: 10.1089/aid.2013.0206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The structure of the HIV-1 envelope membrane-proximal external region (MPER) is influenced by its association with the lipid bilayer on the surface of virus particles and infected cells. To develop a replicating vaccine vector displaying MPER sequences in association with membrane, Env epitopes recognized by the broadly neutralizing antibodies 2F5, 4E10, or both were grafted into the membrane-proximal stem region of the vesicular stomatitis virus (VSV) glycoprotein (G). VSV encoding functional G-MPER chimeras based on G from the Indiana or New Jersey serotype propagated efficiently, although grafting of both epitopes (G-2F5-4E10) modestly reduced replication and resulted in the acquisition of one to two adaptive mutations in the grafted MPER sequence. Monoclonal antibodies 2F5 and 4E10 efficiently neutralized VSV G-MPER vectors and bound to virus particles in solution, indicating that the epitopes were accessible in the preattachment form of the G-MPER chimeras. Overall, our results showed that the HIV Env MPER could functionally substitute for the VSV G-stem region implying that both perform similar functions even though they are from unrelated viruses. Furthermore, we found that the MPER sequence grafts induced low but detectable MPER-specific antibody responses in rabbits vaccinated with live VSV, although additional vector and immunogen modifications or use of a heterologous prime-boost vaccination regimen will be required to increase the magnitude of the immune response.
Collapse
Affiliation(s)
- Ivo C. Lorenz
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Hanh T. Nguyen
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
- Program in Molecular and Cellular Biology, School of Graduate Studies, The State University of New York Downstate Medical Center, Brooklyn, New York
| | - Marina Kemelman
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Ross W. Lindsay
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Maoli Yuan
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Kevin J. Wright
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Heather Arendt
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | | | - Joanne DeStefano
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Simon Hoffenberg
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Gavin Morrow
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Christy K. Jurgens
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Sanjay K. Phogat
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Timothy J. Zamb
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| | - Christopher L. Parks
- The International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York
| |
Collapse
|
25
|
Rabinovich S, Powell RLR, Lindsay RWB, Yuan M, Carpov A, Wilson A, Lopez M, Coleman JW, Wagner D, Sharma P, Kemelman M, Wright KJ, Seabrook JP, Arendt H, Martinez J, DeStefano J, Chiuchiolo MJ, Parks CL. A novel, live-attenuated vesicular stomatitis virus vector displaying conformationally intact, functional HIV-1 envelope trimers that elicits potent cellular and humoral responses in mice. PLoS One 2014; 9:e106597. [PMID: 25215861 PMCID: PMC4162551 DOI: 10.1371/journal.pone.0106597] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 08/03/2014] [Indexed: 01/09/2023] Open
Abstract
Though vaccination with live-attenuated SIV provides the greatest protection from progressive disease caused by SIV challenge in rhesus macaques, attenuated HIV presents safety concerns as a vaccine; therefore, live viral vectors carrying HIV immunogens must be considered. We have designed a replication-competent vesicular stomatitis virus (VSV) displaying immunogenic HIV-1 Env trimers and attenuating quantities of the native surface glycoprotein (G). The clade B Env immunogen is an Env-VSV G hybrid (EnvG) in which the transmembrane and cytoplasmic tail regions are derived from G. Relocation of the G gene to the 5'terminus of the genome and insertion of EnvG into the natural G position induced a ∼1 log reduction in surface G, significant growth attenuation compared to wild-type, and incorporation of abundant EnvG. Western blot analysis indicated that ∼75% of incorporated EnvG was a mature proteolytically processed form. Flow cytometry showed that surface EnvG bound various conformationally- and trimer-specific antibodies (Abs), and in-vitro growth assays on CD4+CCR5+ cells demonstrated EnvG functionality. Neither intranasal (IN) or intramuscular (IM) administration in mice induced any observable pathology and all regimens tested generated potent Env-specific ELISA titers of 10(4)-10(5), with an IM VSV prime/IN VSV boost regimen eliciting the highest binding and neutralizing Ab titers. Significant quantities of Env-specific CD4+ T cells were also detected, which were augmented as much as 70-fold by priming with IM electroporated plasmids encoding EnvG and IL-12. These data suggest that our novel vector can achieve balanced safety and immunogenicity and should be considered as an HIV vaccine platform.
Collapse
Affiliation(s)
- Svetlana Rabinovich
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
- Molecular and Cellular Biology Program, The School of Graduate Studies, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Rebecca L. R. Powell
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Ross W. B. Lindsay
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Maoli Yuan
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Alexei Carpov
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Aaron Wilson
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Mary Lopez
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - John W. Coleman
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Denise Wagner
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Palka Sharma
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Marina Kemelman
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Kevin J. Wright
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - John P. Seabrook
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Heather Arendt
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Jennifer Martinez
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Joanne DeStefano
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Maria J. Chiuchiolo
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
- Molecular and Cellular Biology Program, The School of Graduate Studies, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Christopher L. Parks
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
- Molecular and Cellular Biology Program, The School of Graduate Studies, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| |
Collapse
|
26
|
Ghobadloo SM, Balcerzak AK, Gargaun A, Muharemagic D, Mironov GG, Capicciotti CJ, Briard JG, Ben RN, Berezovski MV. Carbohydrate-based ice recrystallization inhibitors increase infectivity and thermostability of viral vectors. Sci Rep 2014; 4:5903. [PMID: 25078058 PMCID: PMC4116624 DOI: 10.1038/srep05903] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 07/16/2014] [Indexed: 12/28/2022] Open
Abstract
The inability of vaccines to retain sufficient thermostability has been an obstacle to global vaccination programs. To address this major limitation, we utilized carbohydrate-based ice recrystallization inhibitors (IRIs) to eliminate the cold chain and stabilize the potency of Vaccinia virus (VV), Vesicular Stomatitis virus (VSV) and Herpes virus-1 (HSV-1). The impact of these IRIs was tested on the potency of the viral vectors using a plaque forming unit assay following room temperature storage, cryopreservation with successive freeze-thaw cycles and lyophilization. Viral potency after storage with all three conditions demonstrated that N-octyl-gluconamide (NOGlc) recovered the infectivity of shelf stored VV, 5.6 Log10 PFU mL−1 during 40 days, and HSV-1, 2.7 Log10 PFU mL−1 during 9 days. Carbon-linked antifreeze glycoprotein analogue ornithine-glycine-glycine-galactose (OGG-Gal) increases the recovery of VV and VSV more than 1 Log10 PFU mL−1 after 10 freeze-thaw cycles. In VSV, cryostorage with OGG-Gal maintains high infectivity and reduces temperature-induced aggregation of viral particles by 2 times that of the control. In total, OGG-Gal and NOGlc preserve virus potency during cryostorage. Remarkably, NOGlc has potential to eliminate the cold chain and permit room temperature storage of viral vectors.
Collapse
Affiliation(s)
| | - Anna K Balcerzak
- Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Ana Gargaun
- Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Darija Muharemagic
- Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Gleb G Mironov
- Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | | | - Jennie G Briard
- Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Robert N Ben
- Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Maxim V Berezovski
- Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
27
|
Tober R, Banki Z, Egerer L, Muik A, Behmüller S, Kreppel F, Greczmiel U, Oxenius A, von Laer D, Kimpel J. VSV-GP: a potent viral vaccine vector that boosts the immune response upon repeated applications. J Virol 2014; 88:4897-907. [PMID: 24554655 PMCID: PMC3993835 DOI: 10.1128/jvi.03276-13] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 02/03/2014] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Antivector immunity limits the response to homologous boosting for viral vector vaccines. Here, we describe a new, potent vaccine vector based on replication-competent vesicular stomatitis virus pseudotyped with the glycoprotein of the lymphocytic choriomeningitis virus (VSV-GP), which we previously showed to be safe in mice. In mice, VSV and VSV-GP encoding ovalbumin (OVA) as a model antigen (VSV-OVA and VSV-GP-OVA) induced equal levels of OVA-specific humoral and cellular immune responses upon a single immunization. However, boosting with the same vector was possible only for VSV-GP-OVA as neutralizing antibodies to VSV limited the immunogenicity of the VSV-OVA boost. OVA-specific cytotoxic T-lymphocyte (CTL) responses induced by VSV-GP-OVA were at least as potent as those induced by an adenoviral state-of-the-art vaccine vector and completely protected mice in a Listeria monocytogenes challenge model. VSV-GP is so far the only replication-competent vaccine vector that does not lose efficacy upon repeated application. IMPORTANCE Although there has been great progress in treatment and prevention of infectious diseases in the past several years, effective vaccines against some of the most serious infections, e.g., AIDS, malaria, hepatitis C, or tuberculosis, are urgently needed. Here, several approaches based on viral vector vaccines are under development. However, for all viral vaccine vectors currently in clinical testing, repeated application is limited by neutralizing antibodies to the vector itself. Here, we have exploited the potential of vesicular stomatitis virus pseudotyped with the glycoprotein of the lymphocytic choriomeningitis virus (VSV-GP) as a vaccine platform. VSV-GP is the first replication-competent viral vector vaccine that does not induce vector-specific humoral immunity, i.e., neutralizing antibodies, and therefore can boost immune responses against a foreign antigen by repeated applications. The vector allows introduction of various antigens and therefore can serve as a platform technology for the development of novel vaccines against a broad spectrum of diseases.
Collapse
Affiliation(s)
- Reinhard Tober
- Division of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Zoltan Banki
- Division of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Lisa Egerer
- Division of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Alexander Muik
- Applied Virology and Gene Therapy Unit, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | | | | | - Ute Greczmiel
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | | | - Dorothee von Laer
- Division of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Janine Kimpel
- Division of Virology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
28
|
Neurovirulence and immunogenicity of attenuated recombinant vesicular stomatitis viruses in nonhuman primates. J Virol 2014; 88:6690-701. [PMID: 24696472 DOI: 10.1128/jvi.03441-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED In previous work, a prototypic recombinant vesicular stomatitis virus Indiana serotype (rVSIV) vector expressing simian immunodeficiency virus (SIV) gag and human immunodeficiency virus type 1 (HIV-1) env antigens protected nonhuman primates (NHPs) from disease following challenge with an HIV-1/SIV recombinant (SHIV). However, when tested in a stringent NHP neurovirulence (NV) model, this vector was not adequately attenuated for clinical evaluation. For the work described here, the prototypic rVSIV vector was attenuated by combining specific G protein truncations with either N gene translocations or mutations (M33A and M51A) that ablate expression of subgenic M polypeptides, by incorporation of temperature-sensitive mutations in the N and L genes, and by deletion of the VSIV G gene to generate a replicon that is dependent on trans expression of G protein for in vitro propagation. When evaluated in a series of NHP NV studies, these attenuated rVSIV variants caused no clinical disease and demonstrated a very significant reduction in neuropathology compared to wild-type VSIV and the prototypic rVSIV vaccine vector. In spite of greatly increased in vivo attenuation, some of the rVSIV vectors elicited cell-mediated immune responses that were similar in magnitude to those induced by the much more virulent prototypic vector. These data demonstrate novel approaches to the rational attenuation of VSIV NV while retaining vector immunogenicity and have led to identification of an rVSIV N4CT1gag1 vaccine vector that has now successfully completed phase I clinical evaluation. IMPORTANCE The work described in this article demonstrates a rational approach to the attenuation of vesicular stomatitis virus neurovirulence. The major attenuation strategy described here will be most likely applicable to other members of the Rhabdoviridae and possibly other families of nonsegmented negative-strand RNA viruses. These studies have also enabled the identification of an attenuated, replication-competent rVSIV vector that has successfully undergone its first clinical evaluation in humans. Therefore, these studies represent a major milestone in the development of attenuated rVSIV, and likely other vesiculoviruses, as a new vaccine platform(s) for use in humans.
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW To briefly describe some of the replication-competent vectors being investigated for development of candidate HIV vaccines focusing primarily on technologies that have advanced to testing in macaques or have entered clinical trials. RECENT FINDINGS Replication-competent viral vectors have advanced to the stage at which decisions can be made regarding the future development of HIV vaccines. The viruses being used as replication-competent vector platforms vary considerably, and their unique attributes make it possible to test multiple vaccine design concepts and also mimic various aspects of an HIV infection. Replication-competent viral vectors encoding simian immunodeficiency virus or HIV proteins can be used to safely immunize macaques, and in some cases, there is evidence of significant vaccine efficacy in challenge protection studies. Several live HIV vaccine vectors are in clinical trials to evaluate immunogenicity, safety, the effect of mucosal delivery, and potential effects of preexisting immunity. SUMMARY A variety of DNA and RNA viruses are being used to develop replication-competent viral vectors for HIV vaccine delivery. Multiple viral vector platforms have proven to be well tolerated and immunogenic with evidence of efficacy in macaques. Some of the more advanced HIV vaccine prototypes based on vesicular stomatitis virus, vaccinia virus, measles virus, and Sendai virus are in clinical trials.
Collapse
|
30
|
Kurisetty VVS, Heiber J, Myers R, Pereira GS, Goodwin JW, Federspiel MJ, Russell SJ, Peng KW, Barber G, Merchan JR. Preclinical safety and activity of recombinant VSV-IFN-β in an immunocompetent model of squamous cell carcinoma of the head and neck. Head Neck 2014; 36:1619-27. [PMID: 24115092 DOI: 10.1002/hed.23502] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 07/12/2013] [Accepted: 09/10/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Recombinant vesicular stomatitis virus expressing interferon-β (VSV-IFN-β) has demonstrated antitumor activity in vitro and in vivo. In preparation for clinical testing in human squamous cell carcinoma (SCC) of the head and neck, we conducted preclinical studies of VSV-IFN-β in syngeneic SCC models. METHODS In vitro, VSV-IFN-β (expressing rat or mouse interferon [IFN]-β)-induced cytotoxicity and propagated in rat (FAT-7) or mouse (SCC-VII) SCC cells during normoxia and hypoxia. In vivo, intratumoral administration of VSV-rat-IFN-β or VSV-human-IFN-β in FAT-7 bearing or non-tumor bearing immunocompetent rats did not result in acute organ toxicity or death. RESULTS VSV-r-IFN-β replicated predominantly in tumors and a dose dependent anti-VSV antibody response was observed. Intratumoral or intravenous administration of VSV-IFN-β resulted in growth delay and improved survival compared with controls. CONCLUSION The above data confirm safety and feasibility of VSV-IFN-β administration in immunocompetent animals and support its clinical evaluation in advanced human head and neck cancer.
Collapse
Affiliation(s)
- Vittal V S Kurisetty
- Division of Hematology and Oncology, University of Miami/Miller School of Medicine, Miami, Florida
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Overcoming cancer cell resistance to VSV oncolysis with JAK1/2 inhibitors. Cancer Gene Ther 2013; 20:582-9. [PMID: 24030211 DOI: 10.1038/cgt.2013.55] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/10/2013] [Indexed: 02/01/2023]
Abstract
Oncolytic vesicular stomatitis virus (VSV) has potent antitumor activity but some cancer cells are resistant to VSV killing, either constitutively or due to type I interferon (IFN) inducing an antiviral state in the cells. Here, we evaluated VSV oncolysis of a panel of human head and neck cancer cells and showed that VSV resistance in SCC25 and SCC15 cells could be reversed with Janus kinase (JAK) 1/2 inhibitors (JAK inhibitor I and ruxolitinib). Pre-treatment of cells with JAK1/2 inhibitors before or in conjunction with VSV enhanced viral infection, spread and progeny yield (100- to 1000-fold increase). In contrast, inhibitors of histone deacetylase (LBH589), phosphatidylinositol 3-kinase (GDC-0941, LY294002), mammalian target of rapamycin (rapamycin) or signal transducer and activator of transcription 3 (STAT3 inhibitor VII) were ineffective. Compared with VSV-sensitive SW579 cells, IFNα/β responsive antiviral genes (IRF-9, IRF-7, OAS1 but not MxA) are constitutively expressed in SCC25 cells. Pretreatment with JAK inhibitors reduced mRNA levels of these genes, increasing VSV expression in the cells. Interestingly, 1 h of drug exposure was sufficient to reverse SCC25 resistance to VSV and was still effective if virus was added 24 h later. Overall, we show here that JAK inhibitor I and ruxolitinib (Jakafi) can reverse resistance to VSV, supporting the rationale to incorporate JAK1/2 inhibitors in future VSV virotherapy trials.
Collapse
|
32
|
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
|
33
|
Mooney AJ, Tompkins SM. Experimental vaccines against potentially pandemic and highly pathogenic avian influenza viruses. Future Virol 2013; 8:25-41. [PMID: 23440999 PMCID: PMC3579652 DOI: 10.2217/fvl.12.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Influenza A viruses continue to emerge and re-emerge, causing outbreaks, epidemics and occasionally pandemics. While the influenza vaccines licensed for public use are generally effective against seasonal influenza, issues arise with production, immunogenicity, and efficacy in the case of vaccines against pandemic and emerging influenza viruses, and highly pathogenic avian influenza virus in particular. Thus, there is need of improved influenza vaccines and vaccination strategies. This review discusses advances in alternative influenza vaccines, touching briefly on licensed vaccines and vaccine antigens; then reviewing recombinant subunit vaccines, virus-like particle vaccines and DNA vaccines, with the main focus on virus-vectored vaccine approaches.
Collapse
Affiliation(s)
- Alaina J Mooney
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| |
Collapse
|
34
|
Non-propagating, recombinant vesicular stomatitis virus vectors encoding respiratory syncytial virus proteins generate potent humoral and cellular immunity against RSV and are protective in mice. Immunol Lett 2012; 150:134-44. [PMID: 23261719 DOI: 10.1016/j.imlet.2012.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 11/21/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract illness in infants, the elderly, and other high-risk individuals. Despite years of research in this field, there is no effective licensed vaccine to prevent RSV infection. We have generated candidate RSV vaccines using a recombinant vesicular stomatitis virus (rVSV) replicon in which the attachment and fusion domains of the VSV glycoprotein (G) have been deleted (rVSV-Gstem), rendering the virus propagation-defective except in the presence of complementing VSV G provided in trans. A form of this vector encoding the RSV fusion protein (F) gene expressed high levels of F in vitro and elicited durable neutralizing antibody responses as well as complete protection against RSV challenge in vivo. Mice vaccinated with rVSV-Gstem-RSV-F replicons also developed robust cellular responses characterized by both primary and memory Th1-biased CD8+ and CD4+ T cells. Furthermore, a single high dose of the Gstem-RSV-F replicon was effective against challenge with both RSV A and B subgroup viruses. Finally, addition of an RSV glycoprotein (G)-expressing Gstem vector significantly improved the incomplete protection achieved with a single low dose of Gstem-RSV-F vector alone.
Collapse
|
35
|
An HY, Kim GN, Wu K, Kang CY. Genetically modified VSV(NJ) vector is capable of accommodating a large foreign gene insert and allows high level gene expression. Virus Res 2012. [PMID: 23207069 DOI: 10.1016/j.virusres.2012.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It is desirable to develop a RNA virus vector capable of accommodating large foreign genes for high level gene expression. Vesicular stomatitis virus (VSV) has been used as a gene expression vector, especially Indiana serotype (VSV(Ind)), but less with New Jersey serotype (VSV(NJ)). Here, we report constructions of genetically modified rVSV(NJ) vector carrying various lengths of human hepatitis C virus (HCV) non-structural (NS) protein genes, level of inserted gene expression and characterization of rVSV(NJ). We modified the M gene of VSV(NJ) by changing methionine to arginine at positions 48 and 51 (rVSV(NJ)-M) (Kim and Kang, 2007) for construction of rVSV(NJ) with various lengths of HCV non-structural genes. The NS polyprotein genes of HCV were inserted between the G and L genes of the rVSV(NJ)-M vector, and recombinant VSV(NJ)-M viruses with HCV gene inserts were recovered by the reverse genetics. The recombinant VSV(NJ)-M vector with the HCV NS genes express high levels of all different forms of the NS proteins. The electron microscopic examination showed that lengths of recombinant VSV(NJ)-M without gene of interests, VSV(NJ)-M with a gene of HCV NS3 and NS4A (VSV(NJ)-M-NS3/4A), VSV(NJ)-M with a gene of HCV NS4AB plus NS5AB (VSV(NJ)-M-NS4AB/5AB), and VSV(NJ)-M carrying a gene of HCV NS3, NS4AB, and NS5AB (VSV(NJ)-M-NS3/4AB/5AB) were 172±10.5 nm, 201±12.5 nm, 226±12.9 nm, and 247±18.2 nm, respectively. The lengths of recombinant VSVs increased approximately 10nm by insertion of 1kb of foreign genes. The diameter of these recombinant viruses also increased slightly by longer HCV gene inserts. Our results showed that the recombinant VSV(NJ)-M vector can accommodate as much as 6000 bases of the foreign gene. We compared the magnitude of the IFN induction in mouse fibroblast L(Y) cells infected with rVSV(NJ) wild type and rVSV(NJ) M mutant viruses and show that the rVSV(NJ) M mutant virus infection induced a higher level of the IFN-β compare to the wild type virus. In addition, we showed that the NS protein expression level in IFN-incompetent cells (Mouse-L) infected with rVSV(NJ)-M viruses was higher than in IFN-competent L(Y) cells. In addition, we confirmed that HCV NS protein genes were expressed and properly processed. We also confirmed that NS3 protein expressed from the rVSV(NJ)-M cleaves NS polyprotein at junctions and that NS4A plays an important role as a co-factor for NS3 protease to cleave at the NS4B/5A site and at the NS5A/5B site.
Collapse
Affiliation(s)
- Hwa-Yong An
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Siebens-Drake Research Institute, Western University, London, ON N6G 2V4, Canada
| | | | | | | |
Collapse
|
36
|
Highly attenuated recombinant vesicular stomatitis virus VSV-12'GFP displays immunogenic and oncolytic activity. J Virol 2012; 87:1019-34. [PMID: 23135719 DOI: 10.1128/jvi.01106-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vesicular stomatitis virus (VSV) has shown considerable promise both as an immunization vector and as an oncolytic virus. In both applications, an important concern is the safety profile of the virus. To generate a highly attenuated virus, we added two reporter genes to the 3' end of the VSV genome, thereby shifting the NPMGL genes from positions 1 to 5 to positions 3 to 7. The resulting virus (VSV-12'GFP) was highly attenuated, generating smaller plaques than four other attenuated VSVs. In one-step growth curves, VSV-12'GFP displayed the slowest growth kinetics. The mechanism of attenuation appears to be due to reduced expression of VSV genes downstream of the reporter genes, as suggested by a 10.4-fold reduction in L-protein RNA transcript. Although attenuated, VSV-12'GFP was highly effective at generating an immune response, indicated by a high-titer antibody response against the green fluorescent protein (GFP) expressed by the virus. Although VSV-12'GFP was more attenuated than other VSVs on both normal and cancer cells, it nonetheless showed a greater level of infection of human cancer cells (glioma and melanoma) than of normal cells, and this effect was magnified in glioma by interferon application, indicating selective oncolysis. Intravenous VSV-12'GFP selectively infected human gliomas implanted into SCID mice subcutaneously or intracranially. All postnatal day 16 mice given intranasal VSV-12'GFP survived, whereas only 10% of those given VSV-G/GFP survived, indicating reduced neurotoxicity. Intratumoral injection of tumors with VSV-12'GFP dramatically suppressed tumor growth and enhanced survival. Together these data suggest this recombinant virus merits further study for its oncolytic and vaccine potential.
Collapse
|
37
|
Azizi A, Mironov GG, Muharemagic D, Wehbe M, Bell JC, Berezovski MV. Viral quantitative capillary electrophoresis for counting and quality control of RNA viruses. Anal Chem 2012; 84:9585-91. [PMID: 23046075 DOI: 10.1021/ac302525y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The world of health care has witnessed an explosive boost to its capacity within the past few decades due to the introduction of viral therapeutics to its medicinal arsenal. As a result, a need for new methods of viral quantification has arisen to accommodate this rapid advancement in virology and associated requirements for efficiency, speed, and quality control. In this work, we apply viral quantitative capillary electrophoresis (viral qCE) to determine (i) the number of intact virus particles (ivp) in viral samples, (ii) the amount of DNA contamination, and (iii) the degree of viral degradation after sonication, vortexing, and freeze-thaw cycles. This quantification method is demonstrated on an RNA-based vesicular stomatitis virus (VSV) with oncolytic properties. A virus sample contains intact VSV particles as well as residual DNA from host cells, which is regulated by WHO guidelines, and may include some carried-over RNA. We use capillary zone electrophoresis with laser-induced fluorescent detection to separate intact virus particles from DNA and RNA impurities. YOYO-1 dye is used to stain all DNA and RNA in the sample. After soft lysis of VSV with proteinase K digestion of viral capsid and ribonucleoproteins, viral RNA is released. Therefore, the initial concentration of intact virus is calculated based on the gain of a nucleic acid peak and an RNA calibration curve. After additional NaOH treatment of the virus sample, RNA is hydrolyzed leaving residual DNA only, which is also calculated by a DNA calibration curve made by the same CE instrument. Viral qCE works in a wide dynamic range of virus concentrations from 10(8) to 10(13) ivp/mL. It can be completed in a few hours and requires minimum optimization of CE separation.
Collapse
Affiliation(s)
- Afnan Azizi
- Department of Chemistry, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
38
|
Ayala-Breton C, Barber GN, Russell SJ, Peng KW. Retargeting vesicular stomatitis virus using measles virus envelope glycoproteins. Hum Gene Ther 2012; 23:484-91. [PMID: 22171635 DOI: 10.1089/hum.2011.146] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Oncolytic vesicular stomatitis virus (VSV) has potent antitumor activity, but infects a broad range of cell types. Here, we used the measles virus (MV) hemagglutinin (H) and fusion (F) envelope glycoproteins to redirect VSV entry and infection specifically to tumor-associated receptors. Replication-defective VSV, deleted of its glycoprotein gene (VSVΔG), was pseudotyped with MV-F and MV-H displaying single-chain antibodies (scFv) specific for epidermal growth factor receptor (EGFR), folate receptor (FR), or prostate membrane-specific antigen (PSMA). Viral titers were ∼10(5) PFU/ml, but could be concentrated to 10(7) PFU/ml. Immunoblotting confirmed incorporation of the MV-H-scFv and MV-F into functional VSV virions. Although VSV-G was able to infect all tumor cell lines tested, the retargeted VSV infected only cells that expressed the targeted receptor. In vivo specificities of the EGFR-, FR-, and PSMA-retargeted VSV were assessed by intratumoral injection into human tumor xenografts. Analysis of green fluorescent protein reporter gene expression indicated that VSV infection was restricted to receptor-positive tumors. In summary, we have demonstrated for the first time that VSV can be efficiently retargeted to different cellular receptors using the measles display technology, yielding retargeted VSV vectors that are highly specific for tumors that express the relevant receptor.
Collapse
|
39
|
|
40
|
Sato H, Yoneda M, Honda T, Kai C. Recombinant vaccines against the mononegaviruses--what we have learned from animal disease controls. Virus Res 2011; 162:63-71. [PMID: 21982973 PMCID: PMC7114506 DOI: 10.1016/j.virusres.2011.09.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 09/28/2011] [Indexed: 11/30/2022]
Abstract
The mononegaviruses include a number of highly contagious and severe disease-causing viruses of both animals and humans. For the control of these viral diseases, development of vaccines, either with classical methods or with recombinant DNA virus vectors, has been attempted over the years. Recently reverse genetics of mononegaviruses has been developed and used to generate infectious viruses possessing genomes derived from cloned cDNA in order to study the consequent effects of viral gene manipulations on phenotype. This technology allows us to develop novel candidate vaccines. In particular, a variety of different attenuation strategies to produce a range of attenuated mononegavirus vaccines have been studied. In addition, because of their ideal nature as live vaccines, recombinant mononegaviruses expressing foreign proteins have also been produced with the aim of developing multivalent vaccines against more than one pathogen. These recombinant mononegaviruses are currently under evaluation as new viral vectors for vaccination. Reverse genetics could have great potential for the preparation of vaccines against many mononegaviruses.
Collapse
Affiliation(s)
- Hiroki Sato
- Laboratory Animal Research Center/International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | | | | | | |
Collapse
|
41
|
Excler JL, Parks CL, Ackland J, Rees H, Gust ID, Koff WC. Replicating viral vectors as HIV vaccines: summary report from the IAVI-sponsored satellite symposium at the AIDS vaccine 2009 conference. Biologicals 2011; 38:511-21. [PMID: 20537552 DOI: 10.1016/j.biologicals.2010.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 03/29/2010] [Indexed: 01/30/2023] Open
Abstract
In October 2009, The International AIDS Vaccine Initiative (IAVI) convened a satellite symposium entitled 'Replicating Viral Vectors for use in AIDS Vaccines' at the AIDS Vaccine 2009 Conference in Paris. The purpose of the symposium was to gather together researchers, representatives from regulatory agencies, and vaccine developers to discuss issues related to advancement of replication-competent viral vector- based HIV vaccines into clinical trials. The meeting introduced the rationale for accelerating the development of replicating viral vectors for use as AIDS vaccines. It noted that the EMEA recently published draft guidelines that are an important first step in providing guidance for advancing live viral vectors into clinical development. Presentations included case studies and development challenges for viral vector-based vaccine candidates. These product development challenges included cell substrates used for vaccine manufacturing, the testing needed to assess vaccine safety, conducting clinical trials with live vectors, and assessment of vaccination risk versus benefit. More in depth discussion of risk and benefit highlighted the fact that AIDS vaccine efficacy trials must be conducted in the developing world where HIV incidence is greatest and how inequities in global health dramatically influence the political and social environment in developing countries.
Collapse
Affiliation(s)
- J L Excler
- International AIDS Vaccine Initiative, 110 William Street, 27th Floor, New York, NY 10038-3901, USA
| | | | | | | | | | | |
Collapse
|
42
|
Cafaro A, Macchia I, Maggiorella MT, Titti F, Ensoli B. Innovative approaches to develop prophylactic and therapeutic vaccines against HIV/AIDS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 655:189-242. [PMID: 20047043 DOI: 10.1007/978-1-4419-1132-2_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The acquired immunodeficiency syndrome (AIDS) emerged in the human population in the summer of 1981. According to the latest United Nations estimates, worldwide over 33 million people are infected with human immunodeficiency virus (HIV) and the prevalence rates continue to rise globally. To control the alarming spread of HIV, an urgent need exists for developing a safe and effective vaccine that prevents individuals from becoming infected or progressing to disease. To be effective, an HIV/AIDS vaccine should induce broad and long-lasting humoral and cellular immune responses, at both mucosal and systemic level. However, the nature of protective immune responses remains largely elusive and this represents one of the major roadblocks preventing the development of an effective vaccine. Here we summarize our present understanding of the factors responsible for resistance to infection or control of progression to disease in human and monkey that may be relevant to vaccine development and briefly review recent approaches which are currently being tested in clinical trials. Finally, the rationale and the current status of novel strategies based on nonstructural HIV-1 proteins, such as Tat, Nef and Rev, used alone or in combination with modified structural HIV-1 Env proteins are discussed.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, V.le Regina Elena 299, 00161, Rome, Italy
| | | | | | | | | |
Collapse
|
43
|
Jenks N, Myers R, Greiner SM, Thompson J, Mader EK, Greenslade A, Griesmann GE, Federspiel MJ, Rakela J, Borad MJ, Vile RG, Barber GN, Meier TR, Blanco MC, Carlson SK, Russell SJ, Peng KW. Safety studies on intrahepatic or intratumoral injection of oncolytic vesicular stomatitis virus expressing interferon-beta in rodents and nonhuman primates. Hum Gene Ther 2010; 21:451-62. [PMID: 19911974 DOI: 10.1089/hum.2009.111] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Toxicology studies were performed in rats and rhesus macaques to establish a safe starting dose for intratumoral injection of an oncolytic vesicular stomatitis virus expressing human interferon-beta (VSV-hIFNbeta) in patients with hepatocellular carcinoma (HCC). No adverse events were observed after administration of 7.59 x 10(9) TCID(50) (50% tissue culture infective dose) of VSV-hIFNbeta into the left lateral hepatic lobe of Harlan Sprague Dawley rats. Plasma alanine aminotransferase and alkaline phosphatase levels increased and platelet counts decreased in the virus-treated animals on days 1 and 2 but returned to pretreatment levels by day 4. VSV-hIFNbeta was also injected into normal livers or an intrahepatic McA-RH7777 HCC xenograft established in Buffalo rats. Buffalo rats were more sensitive to neurotoxic effects of VSV; the no observable adverse event level (NOAEL) of VSV-hIFNbeta in Buffalo rats was 10(7) TCID(50). Higher doses were associated with fatal neurotoxicity and infectious virus was recovered from tumor and brain. Compared with VSV-hIFNbeta, toxicity of VSV-rIFNbeta (recombinant VSV expressing rat IFN-beta) was greatly diminished in Buffalo rats (NOAEL, >10(10) TCID(50)). Two groups of two adult male rhesus macaques received 10(9) or 10(10) TCID(50) of VSV-hIFNbeta injected directly into the left hepatic lobe under computed tomographic guidance. No neurological signs were observed at any time point. No abnormalities (hematology, clinical chemistry, body weights, behavior) were seen and all macaques developed neutralizing anti-VSV antibodies. Plasma interleukin-6, tumor necrosis factor-alpha, and hIFN-beta remained below detection levels by ELISA. On the basis of these studies, we will be proposing a cautious approach to dose escalation in a phase I clinical trial among patients with HCC.
Collapse
Affiliation(s)
- Nathan Jenks
- Toxicology and Pharmacology Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
A vesicular stomatitis virus-based hepatitis B virus vaccine vector provides protection against challenge in a single dose. J Virol 2010; 84:7513-22. [PMID: 20504927 DOI: 10.1128/jvi.00200-10] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
As one of the world's most common infectious diseases, hepatitis B virus (HBV) is a serious worldwide public health problem, with HBV-associated liver disease accounting for more than half a million deaths each year. Although there is an effective prophylactic vaccine currently available to prevent infection, it has a number of characteristics that are suboptimal: multiple doses are needed to induce long-lasting immunity, immunity declines over time, it does not elicit protection in some individuals, and it is not effective therapeutically. We produced a recombinant vesicular stomatitis virus (VSV)-based vaccine vector expressing the HBV middle envelope surface protein (MS) and found that this vector was able to efficiently generate a strong HBs-specific antibody response following a single immunization in mice. A single immunization with the VSV-MS vector also induced robust CD8 T-cell activation. The CD8 T-cell response was greater in magnitude and broader in specificity than the response generated by a vaccinia virus-based vaccine vector or by recombinant protein immunization. Furthermore, a single VSV-MS immunization provided protection against virus challenge in mice. Given the similar antibody titers and superior T-cell responses elicited from a single immunization, a VSV-based HBV vaccine may have advantages over the current recombinant protein vaccine.
Collapse
|
45
|
Progress towards development of an HIV vaccine: report of the AIDS Vaccine 2009 Conference. THE LANCET. INFECTIOUS DISEASES 2010; 10:305-16. [PMID: 20417413 DOI: 10.1016/s1473-3099(10)70069-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The search for an HIV/AIDS vaccine is steadily moving ahead, generating and validating new concepts in terms of novel vectors for antigen delivery and presentation, new vaccine and adjuvant strategies, alternative approaches to design HIV-1 antigens for eliciting protective cross-neutralising antibodies, and identification of key mechanisms in HIV infection and modulation of the immune system. All these different perspectives are contributing to the unprecedented challenge of developing a protective HIV-1 vaccine. The high scientific value of this massive effort is its great impact on vaccinology as a whole, providing invaluable scientific information for the current and future development of new preventive vaccine as well as therapeutic knowledge-based infectious-disease and cancer vaccines.
Collapse
|
46
|
Immunogenicity of viral vector, prime-boost SIV vaccine regimens in infant rhesus macaques: attenuated vesicular stomatitis virus (VSV) and modified vaccinia Ankara (MVA) recombinant SIV vaccines compared to live-attenuated SIV. Vaccine 2009; 28:1481-92. [PMID: 19995539 DOI: 10.1016/j.vaccine.2009.11.061] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 11/09/2009] [Accepted: 11/22/2009] [Indexed: 11/21/2022]
Abstract
In a previously developed infant macaque model mimicking HIV infection by breast-feeding, we demonstrated that intramuscular immunization with recombinant poxvirus vaccines expressing simian immunodeficiency virus (SIV) structural proteins provided partial protection against infection following oral inoculation with virulent SIV. In an attempt to further increase systemic but also local antiviral immune responses at the site of viral entry, we tested the immunogenicity of different orally administered, replicating vaccines. One group of newborn macaques received an oral prime immunization with a recombinant vesicular stomatitis virus expressing SIVmac239 gag, pol and env (VSV-SIVgpe), followed 2 weeks later by an intramuscular boost immunization with MVA-SIV. Another group received two immunizations with live-attenuated SIVmac1A11, administered each time both orally and intravenously. Control animals received mock immunizations or non-SIV VSV and MVA control vectors. Analysis of SIV-specific immune responses in blood and lymphoid tissues at 4 weeks of age demonstrated that both vaccine regimens induced systemic antibody responses and both systemic and local cell-mediated immune responses. The safety and immunogenicity of the VSV-SIVgpe+MVA-SIV immunization regimen described in this report provide the scientific incentive to explore the efficacy of this vaccine regimen against virulent SIV exposure in the infant macaque model.
Collapse
|
47
|
Witko SE, Johnson JE, Kalyan NK, Felber BK, Pavlakis GN, Sidhu MK, Hendry RM, Udem SA, Parks CL. Refined methods for propagating vesicular stomatitis virus vectors that are defective for G protein expression. J Virol Methods 2009; 164:43-50. [PMID: 19941901 DOI: 10.1016/j.jviromet.2009.11.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 11/09/2009] [Accepted: 11/16/2009] [Indexed: 11/16/2022]
Abstract
Propagation-defective vesicular stomatitis virus (VSV) vectors that encode a truncated G protein (VSV-Gstem) or lack the G gene entirely (VSV-DeltaG) are attractive vaccine vectors because they are immunogenic, cannot replicate and spread after vaccination, and do not express many of the epitopes that elicit neutralizing anti-VSV immunity. To consider advancing non-propagating VSV vectors towards clinical assessment, scalable technology that is compliant with human vaccine manufacturing must be developed to produce clinical trial material. Accordingly, two propagation methods were developed for VSV-Gstem and VSV-DeltaG vectors encoding HIV gag that have the potential to support large-scale production. One method is based on transient expression of G protein after electroporating plasmid DNA into Vero cells and the second is based on a stable Vero cell line that contains a G gene controlled by a heat shock-inducible transcription unit. Both methods reproducibly supported production of 1 x 10(7) to 1 x 10(8) infectious units (I.U.s) of vaccine vector per milliliter. Results from these studies also showed that optimization of the G gene is necessary for abundant G protein expression from electroporated plasmid DNA or from DNA integrated in the genome of a stable cell line, and that the titers of VSV-Gstem vectors generally exceeded VSV-DeltaG.
Collapse
Affiliation(s)
- Susan E Witko
- Pfizer Vaccine Research, 401 North Middletown Road, Pearl River, NY 10965, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Prime-boost vaccination with recombinant mumps virus and recombinant vesicular stomatitis virus vectors elicits an enhanced human immunodeficiency virus type 1 Gag-specific cellular immune response in rhesus macaques. J Virol 2009; 83:9813-23. [PMID: 19625392 DOI: 10.1128/jvi.00550-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Intramuscular inoculation of rhesus macaques with one or more doses of recombinant vesicular stomatitis virus (rVSV) expressing human immunodeficiency virus type 1 (HIV-1) Gag (rVSVgag) typically elicits peak cellular immune responses of 500 to 1,000 gamma interferon (IFN-gamma) enzyme-linked immunospots (ELISPOTS)/10(6) peripheral blood lymphocytes (PBL). Here, we describe the generation of a novel recombinant mumps virus (rMuV) expressing HIV-1 Gag (rMuVgag) and measure the Gag-specific cellular immune responses detected in rhesus macaques following vaccination with a highly attenuated form of rVSV expressing HIV-1 Gag (rVSVN4CT1gag1) and rMuVgag in various prime-boost combinations. Notably, peak Gag-specific cellular immune responses of 3,000 to 3,500 ELISPOTS/10(6) PBL were detected in macaques that were primed with rMuVgag and boosted with rVSVN4CT1gag1. Lower peak cellular immune responses were detected in macaques that were primed with rVSVN4CT1gag1 and boosted with rMuVgag, although longer-term gag-specific responses appeared to remain higher in this group of macaques. These findings indicate that rMuVgag may significantly enhance Gag-specific cellular immune responses when administered with rVSVN4CT1gag1 in heterologous prime-boost regimens.
Collapse
|
49
|
Abstract
The antiquated system used to manufacture the currently licensed inactivated influenza virus vaccines would not be adequate during an influenza virus pandemic. There is currently a search for vaccines that can be developed faster and provide superior, long-lasting immunity to influenza virus as well as other highly pathogenic viruses and bacteria. Recombinant vectors provide a safe and effective method to elicit a strong immune response to a foreign protein or epitope. This review explores the advantages and limitations of several different vectors that are currently being tested, and highlights some of the newer viruses being used as recombinant vectors.
Collapse
|
50
|
Johnson JE, Coleman JW, Kalyan NK, Calderon P, Wright KJ, Obregon J, Ogin-Wilson E, Natuk RJ, Clarke DK, Udem SA, Cooper D, Hendry RM. In vivo biodistribution of a highly attenuated recombinant vesicular stomatitis virus expressing HIV-1 Gag following intramuscular, intranasal, or intravenous inoculation. Vaccine 2009; 27:2930-9. [PMID: 19428903 PMCID: PMC2747378 DOI: 10.1016/j.vaccine.2009.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 03/03/2009] [Accepted: 03/04/2009] [Indexed: 12/19/2022]
Abstract
Recombinant vesicular stomatitis viruses (rVSVs) are being developed as potential HIV-1 vaccine candidates. To characterize the in vivo replication and dissemination of rVSV vectors in mice, high doses of a highly attenuated vector expressing HIV-1 Gag, rVSVIN-N4CT9-Gag1, and a prototypic reference virus, rVSVIN-HIVGag5, were delivered intramuscularly (IM), intranasally (IN), or intravenously (IV). We used quantitative, real-time RT-PCR (Q-PCR) and standard plaque assays to measure the temporal dissemination of these viruses to various tissues. Following IM inoculation, both viruses were detected primarily at the injection site as well as in draining lymph nodes; neither virus induced significant weight loss, pathologic signs, or evidence of neuroinvasion. In contrast, following IN inoculation, the prototypic virus was detected in all tissues tested and caused significant weight loss leading to death. IN administration of rVSVIN-N4CT9-Gag1 resulted in detection in numerous tissues (brain, lung, nasal turbinates, and lymph nodes) albeit in significantly reduced levels, which caused little or no weight loss nor any mortality. Following IV inoculation, both prototypic and attenuated viruses were detected by Q-PCR in all tissues tested. In contrast to the prototype, rVSVIN-N4CT9-Gag1 viral loads were significantly lower in all organs tested, and no infectious virus was detected in the brain following IV inoculation, despite the presence of viral RNA. These studies demonstrated significant differences in the biodistribution patterns of and the associated pathogenicity engendered by the prototypic and attenuated vectors in a highly susceptible host.
Collapse
|