1
|
Jackson Cullison SR, Flemming JP, Karagoz K, Wermuth PJ, Mahoney MG. Mechanisms of extracellular vesicle uptake and implications for the design of cancer therapeutics. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70017. [PMID: 39483807 PMCID: PMC11522837 DOI: 10.1002/jex2.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024]
Abstract
The translation of pre-clinical anti-cancer therapies to regulatory approval has been promising, but slower than hoped. While innovative and effective treatments continue to achieve or seek approval, setbacks are often attributed to a lack of efficacy, failure to achieve clinical endpoints, and dose-limiting toxicities. Successful efforts have been characterized by the development of therapeutics designed to specifically deliver optimal and effective dosing to tumour cells while minimizing off-target toxicity. Much effort has been devoted to the rational design and application of synthetic nanoparticles to serve as targeted therapeutic delivery vehicles. Several challenges to the successful application of this modality as delivery vehicles include the induction of a protracted immune response that results in their rapid systemic clearance, manufacturing cost, lack of stability, and their biocompatibility. Extracellular vesicles (EVs) are a heterogeneous class of endogenous biologically produced lipid bilayer nanoparticles that mediate intercellular communication by carrying bioactive macromolecules capable of modifying cellular phenotypes to local and distant cells. By genetic, chemical, or metabolic methods, extracellular vesicles (EVs) can be engineered to display targeting moieties on their surface while transporting specific cargo to modulate pathological processes following uptake by target cell populations. This review will survey the types of EVs, their composition and cargoes, strategies employed to increase their targeting, uptake, and cargo release, and their potential as targeted anti-cancer therapeutic delivery vehicles.
Collapse
Affiliation(s)
| | - Joseph P. Flemming
- Rowan‐Virtua School of Osteopathic MedicineRowan UniversityStratfordNew JerseyUSA
| | - Kubra Karagoz
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Mỹ G. Mahoney
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Otolaryngology – Head and Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Florova M, Abreu-Mota T, Paesen GC, Beetschen AS, Cornille K, Marx AF, Narr K, Sahin M, Dimitrova M, Swarnalekha N, Beil-Wagner J, Savic N, Pelczar P, Buch T, King CG, Bowden TA, Pinschewer DD. Central tolerance shapes the neutralizing B cell repertoire against a persisting virus in its natural host. Proc Natl Acad Sci U S A 2024; 121:e2318657121. [PMID: 38446855 PMCID: PMC10945855 DOI: 10.1073/pnas.2318657121] [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: 10/25/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Viral mimicry of host cell structures has been postulated to curtail the B cell receptor (BCR) repertoire against persisting viruses through tolerance mechanisms. This concept awaits, however, experimental testing in a setting of natural virus-host relationship. We engineered mouse models expressing a monoclonal BCR specific for the envelope glycoprotein of lymphocytic choriomeningitis virus (LCMV), a naturally persisting mouse pathogen. When the heavy chain of the LCMV-neutralizing antibody KL25 was paired with its unmutated ancestor light chain, most B cells underwent receptor editing, a behavior reminiscent of autoreactive clones. In contrast, monoclonal B cells expressing the same heavy chain in conjunction with the hypermutated KL25 light chain did not undergo receptor editing but exhibited low levels of surface IgM, suggesting that light chain hypermutation had lessened KL25 autoreactivity. Upon viral challenge, these IgMlow cells were not anergic but up-regulated IgM, participated in germinal center reactions, produced antiviral antibodies, and underwent immunoglobulin class switch as well as further affinity maturation. These studies on a persisting virus in its natural host species suggest that central tolerance mechanisms prune the protective antiviral B cell repertoire.
Collapse
Affiliation(s)
- Marianna Florova
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Tiago Abreu-Mota
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Guido C. Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Anna Sophia Beetschen
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Karen Cornille
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Anna-Friederike Marx
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Kerstin Narr
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Mehmet Sahin
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Mirela Dimitrova
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| | - Nivedya Swarnalekha
- Department of Biomedicine, Immune Cell Biology Laboratory, University Hospital Basel, Basel4031, Switzerland
| | - Jane Beil-Wagner
- Institute of Laboratory Animal Science, University of Zurich, Zurich8093, Switzerland
| | - Natasa Savic
- ETH Phenomics Center, ETH Zürich, Zürich8093, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel4001, Switzerland
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich8093, Switzerland
| | - Carolyn G. King
- Department of Biomedicine, Immune Cell Biology Laboratory, University Hospital Basel, Basel4031, Switzerland
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Daniel D. Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel4009, Switzerland
| |
Collapse
|
3
|
Reyna RA, Littlefield KE, Shehu N, Makishima T, Maruyama J, Paessler S. The Importance of Lassa Fever and Its Disease Management in West Africa. Viruses 2024; 16:266. [PMID: 38400041 PMCID: PMC10892767 DOI: 10.3390/v16020266] [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: 12/11/2023] [Revised: 01/21/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Lassa virus (LASV) is a zoonotic pathogen endemic throughout western Africa and is responsible for a human disease known as Lassa fever (LF). Historically, LASV has been emphasized as one of the greatest public health threats in West Africa, with up to 300,000 cases and 5000 associated deaths per year. This, and the fact that the disease has been reported in travelers, has driven a rapid production of various vaccine candidates. Several of these vaccines are currently in clinical development, despite limitations in understanding the immune response to infection. Alarmingly, the host immune response has been implicated in the induction of sensorineural hearing loss in LF survivors, legitimately raising safety questions about any future vaccines as well as efficacy in preventing potential hearing loss. The objective of this article is to revisit the importance and prevalence of LF in West Africa, with focus on Nigeria, and discuss current therapeutic approaches and ongoing vaccine development. In addition, we aim to emphasize the need for more scientific studies relating to LF-associated hearing loss, and to promote critical discussion about potential risks and benefits of vaccinating the population in endemic regions of West Africa.
Collapse
Affiliation(s)
- Rachel A. Reyna
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kirsten E. Littlefield
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nathan Shehu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Infectious Disease Unit, Department of Medicine, Jos University Teaching Hospital, Jos P.M.B. 2076, Nigeria
| | - Tomoko Makishima
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Junki Maruyama
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
4
|
Ma Y, Shao J, Liu W, Gao S, Peng D, Miao C, Yang S, Hou Z, Zhou G, Qi X, Chang H. A vesicular stomatitis virus-based African swine fever vaccine prototype effectively induced robust immune responses in mice following a single-dose immunization. Front Microbiol 2024; 14:1310333. [PMID: 38249478 PMCID: PMC10797088 DOI: 10.3389/fmicb.2023.1310333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/04/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction African swine fever (ASF) is a highly contagious hemorrhagic fever disease in pigs caused by African swine fever virus (ASFV). It is very difficult to control and prevent ASF outbreaks due to the absence of safe and effective vaccines. Methods In order to develop a safe and effective ASF vaccine for the control and prevention of ASF, two ASFV recombinant vesicular stomatitis virus (VSV) live vector vaccine prototypes, containing the gene of p72, and a chimera of p30 and p54, were developed based on the replication-competent VSV, and named VSV-p72 and VSV-p35. The immune potency of VSV-p72 or VSV-p35 alone and in combination was evaluated in BALB/c mice via intramuscular and intranasal vaccination. Results The results indicated that whether administered alone or in combination, the two vaccine prototypes showed acceptable safety in mice and, more importantly, induced high-level specific antibodies against p72, p30, and p54 of ASFV and a strong cellular immune response 28 days after vaccination. The sera from mice vaccinated with the vaccine prototypes significantly inhibited ASFV from infecting porcine alveolar macrophages (PAMs) in vitro. Most notably, the immunized sera from a mixture of VSV-p35 and VSV-p72 inhibited ASFV from infecting PAMs, with an inhibition rate of up to 78.58%. Conclusion Overall, our findings suggest that ASFV recombinant VSV live vector vaccine prototypes may become a promising candidate vaccine for the control and prevention of ASF.
Collapse
Affiliation(s)
- Yunyun Ma
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Junjun Shao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Wei Liu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shandian Gao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Decai Peng
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Chun Miao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Sicheng Yang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhuo Hou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Guangqing Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xuefeng Qi
- College of Veterinary Medicine Northwest A&F University, Yangling, Shanxi, China
| | - Huiyun Chang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Taveira N, Figueiredo I, Calado R, Martin F, Bártolo I, Marcelino JM, Borrego P, Cardoso F, Barroso H. An HIV-1/HIV-2 Chimeric Envelope Glycoprotein Generates Binding and Neutralising Antibodies against HIV-1 and HIV-2 Isolates. Int J Mol Sci 2023; 24:ijms24109077. [PMID: 37240423 DOI: 10.3390/ijms24109077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The development of immunogens that elicit broadly reactive neutralising antibodies (bNAbs) is the highest priority for an HIV vaccine. We have shown that a prime-boost vaccination strategy with vaccinia virus expressing the envelope glycoprotein gp120 of HIV-2 and a polypeptide comprising the envelope regions C2, V3 and C3 elicits bNAbs against HIV-2. We hypothesised that a chimeric envelope gp120 containing the C2, V3 and C3 regions of HIV-2 and the remaining parts of HIV-1 would elicit a neutralising response against HIV-1 and HIV-2. This chimeric envelope was synthesised and expressed in vaccinia virus. Balb/c mice primed with the recombinant vaccinia virus and boosted with an HIV-2 C2V3C3 polypeptide or monomeric gp120 from a CRF01_AG HIV-1 isolate produced antibodies that neutralised >60% (serum dilution 1:40) of a primary HIV-2 isolate. Four out of nine mice also produced antibodies that neutralised at least one HIV-1 isolate. Neutralising epitope specificity was assessed using a panel of HIV-1 TRO.11 pseudoviruses with key neutralising epitopes disrupted by alanine substitution (N160A in V2; N278A in the CD4 binding site region; N332A in the high mannose patch). The neutralisation of the mutant pseudoviruses was reduced or abolished in one mouse, suggesting that neutralising antibodies target the three major neutralising epitopes in the HIV-1 envelope gp120. These results provide proof of concept for chimeric HIV-1/HIV-2 envelope glycoproteins as vaccine immunogens that can direct the antibody response against neutralising epitopes in the HIV-1 and HIV-2 surface glycoproteins.
Collapse
Affiliation(s)
- Nuno Taveira
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health and Science, 2829-511 Caparica, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
| | - Inês Figueiredo
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health and Science, 2829-511 Caparica, Portugal
| | - Rita Calado
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
| | - Francisco Martin
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
| | - Inês Bártolo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
| | - José M Marcelino
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health and Science, 2829-511 Caparica, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
| | - Pedro Borrego
- Centre for Public Administration and Public Policies, Institute of Social and Political Sciences, Universidade de Lisboa, 1300-663 Lisbon, Portugal
| | - Fernando Cardoso
- Unidade de Microbiologia Médica, Saúde Global e Medicina Tropical, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, 1099-085 Lisbon, Portugal
| | - Helena Barroso
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health and Science, 2829-511 Caparica, Portugal
| |
Collapse
|
6
|
Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther 2023; 8:149. [PMID: 37029123 PMCID: PMC10081433 DOI: 10.1038/s41392-023-01408-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- China National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
7
|
Yuan F, Zheng A. Replicating-Competent VSV-Vectored Pseudotyped Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:329-348. [PMID: 36920706 DOI: 10.1007/978-981-99-0113-5_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Vesicular stomatitis virus (VSV) is prototype virus in the family of Rhabdoviridae. Reverse genetic platform has enabled the genetic manipulation of VSV as a powerful live viral vector. Replicating-competent VSV is constructed by replacing the original VSV glycoprotein gene with heterologous envelope genes. The resulting recombinant viruses are able to replicate in permissive cells and incorporate the foreign envelope proteins on the surface of the viral particle without changing the bullet-shape morphology. Correspondingly, the cell tropism of replicating-competent VSV is determined by the foreign envelope proteins. Replicating-competent VSVs have been successfully used for selecting critical viral receptors or host factors, screening mutants that escape therapeutic antibodies, and developing VSV-based live viral vaccines.
Collapse
Affiliation(s)
- Fei Yuan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Aihua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
8
|
Gallinaro A, Pirillo MF, Aldon Y, Cecchetti S, Michelini Z, Tinari A, Borghi M, Canitano A, McKay PF, Bona R, Vescio MF, Grasso F, Blasi M, Baroncelli S, Scarlatti G, LaBranche C, Montefiori D, Klotman ME, Sanders RW, Shattock RJ, Negri D, Cara A. Persistent immunogenicity of integrase defective lentiviral vectors delivering membrane-tethered native-like HIV-1 envelope trimers. NPJ Vaccines 2022; 7:44. [PMID: 35449174 PMCID: PMC9023570 DOI: 10.1038/s41541-022-00465-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 03/18/2022] [Indexed: 11/09/2022] Open
Abstract
Integrase Defective Lentiviral Vectors (IDLVs) represent an attractive vaccine platform for delivering HIV-1 antigens, given their ability to induce specific and persistent immune responses in both mice and non-human primates (NHPs). Recent advances in HIV-1 immunogen design demonstrated that native-like HIV-1 Envelope (Env) trimers that mimic the structure of virion-associated Env induce neutralization breadth in rabbits and macaques. Here, we describe the development of an IDLV-based HIV-1 vaccine expressing either soluble ConSOSL.UFO.664 or membrane-tethered ConSOSL.UFO.750 native-like Env immunogens with enhanced bNAb epitopes exposure. We show that IDLV can be pseudotyped with properly folded membrane-tethered native-like UFO.750 trimers. After a single IDLV injection in BALB/c mice, IDLV-UFO.750 induced a faster humoral kinetic as well as higher levels of anti-Env IgG compared to IDLV-UFO.664. IDLV-UFO.750 vaccinated cynomolgus macaques developed unusually long-lasting anti-Env IgG antibodies, as underlined by their remarkable half-life both after priming and boost with IDLV. After boosting with recombinant ConM SOSIP.v7 protein, two animals developed neutralization activity against the autologous tier 1B ConS virus mediated by V1/V2 and V3 glycan sites responses. By combining the possibility to display stabilized trimeric Env on the vector particles with the ability to induce sustained humoral responses, IDLVs represent an appropriate strategy for delivering rationally designed antigens to progress towards an effective HIV-1 vaccine.
Collapse
Affiliation(s)
| | | | - Yoann Aldon
- Department of Infectious Disease, Imperial College London, Norfolk Place, London, UK
- Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, the Netherlands
| | - Serena Cecchetti
- Confocal Microscopy Unit NMR, Confocal Microscopy Area Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Tinari
- Center for Gender Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Paul F McKay
- Department of Infectious Disease, Imperial College London, Norfolk Place, London, UK
| | - Roberta Bona
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Felicia Grasso
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Blasi
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Silvia Baroncelli
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Celia LaBranche
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Mary E Klotman
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Rogier W Sanders
- Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, the Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, 1300 York Avenue,, New York, NY, USA
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, Norfolk Place, London, UK
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
9
|
O’Donnell KL, Gourdine T, Fletcher P, Shifflett K, Furuyama W, Clancy CS, Marzi A. VSV-Based Vaccines Reduce Virus Shedding and Viral Load in Hamsters Infected with SARS-CoV-2 Variants of Concern. Vaccines (Basel) 2022; 10:435. [PMID: 35335067 PMCID: PMC8951568 DOI: 10.3390/vaccines10030435] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022] Open
Abstract
The continued progression of the COVID-19 pandemic can partly be attributed to the ability of SARS-CoV-2 to mutate and introduce new viral variants. Some of these variants with the potential to spread quickly and conquer the globe are termed variants of concern (VOC). The existing vaccines implemented on a global scale are based on the ancestral strain, which has resulted in increased numbers of breakthrough infections as these VOC have emerged. It is imperative to show protection against VOC infection with newly developed vaccines. Previously, we evaluated two vesicular stomatitis virus (VSV)-based vaccines expressing the SARS-CoV-2 spike protein alone (VSV-SARS2) or in combination with the Ebola virus glycoprotein (VSV-SARS2-EBOV) and demonstrated their fast-acting potential. Here, we prolonged the time to challenge; we vaccinated hamsters intranasally (IN) or intramuscularly 28 days prior to infection with three SARS-CoV-2 VOC-the Alpha, Beta, and Delta variants. IN vaccination with either the VSV-SARS2 or VSV-SARS2-EBOV resulted in the highest protective efficacy as demonstrated by decreased virus shedding and lung viral load of vaccinated hamsters. Histopathologic analysis of the lungs revealed the least amount of lung damage in the IN-vaccinated animals regardless of the challenge virus. This data demonstrates the ability of a VSV-based vaccine to not only protect from disease caused by SARS-CoV-2 VOC but also reduce viral shedding.
Collapse
Affiliation(s)
- Kyle L. O’Donnell
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Tylisha Gourdine
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Paige Fletcher
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Kyle Shifflett
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Chad S. Clancy
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| |
Collapse
|
10
|
Chiale C, Marchese AM, Furuya Y, Robek MD. Virus-based vaccine vectors with distinct replication mechanisms differentially infect and activate dendritic cells. NPJ Vaccines 2021; 6:138. [PMID: 34811393 PMCID: PMC8608815 DOI: 10.1038/s41541-021-00400-w] [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: 11/22/2020] [Accepted: 10/22/2021] [Indexed: 11/09/2022] Open
Abstract
The precise mechanism by which many virus-based vectors activate immune responses remains unknown. Dendritic cells (DCs) play key roles in priming T cell responses and controlling virus replication, but their functions in generating protective immunity following vaccination with viral vectors are not always well understood. We hypothesized that highly immunogenic viral vectors with identical cell entry pathways but unique replication mechanisms differentially infect and activate DCs to promote antigen presentation and activation of distinctive antigen-specific T cell responses. To evaluate differences in replication mechanisms, we utilized a rhabdovirus vector (vesicular stomatitis virus; VSV) and an alphavirus-rhabdovirus hybrid vector (virus-like vesicles; VLV), which replicates like an alphavirus but enters the cell via the VSV glycoprotein. We found that while virus replication promotes CD8+ T cell activation by VLV, replication is absolutely required for VSV-induced responses. DC subtypes were differentially infected in vitro with VSV and VLV, and displayed differences in activation following infection that were dependent on vector replication but were independent of interferon receptor signaling. Additionally, the ability of the alphavirus-based vector to generate functional CD8+ T cells in the absence of replication relied on cDC1 cells. These results highlight the differential activation of DCs following infection with unique viral vectors and indicate potentially discrete roles of DC subtypes in activating the immune response following immunization with vectors that have distinct replication mechanisms.
Collapse
Affiliation(s)
- Carolina Chiale
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.,Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Anthony M Marchese
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Yoichi Furuya
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Michael D Robek
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
11
|
Pinski AN, Messaoudi I. Therapeutic vaccination strategies against EBOV by rVSV-EBOV-GP: the role of innate immunity. Curr Opin Virol 2021; 51:179-189. [PMID: 34749265 DOI: 10.1016/j.coviro.2021.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022]
Abstract
Zaire Ebola virus (EBOV) is a member of the Filoviridae family. Infection with EBOV causes Ebola virus disease (EVD) characterized by excessive inflammation, lymphocyte death, coagulopathy, and multi-organ failure. In 2019, the FDA-approved the first anti-EBOV vaccine, rVSV-EBOV-GP (Ervebo® by Merck). This live-recombinant vaccine confers both prophylactic and therapeutic protection to nonhuman primates and humans. While mechanisms conferring prophylactic protection are well-investigated, those underlying protection conferred shortly before and after exposure to EBOV remain poorly understood. In this review, we review data from in vitro and in vivo studies analyzing early immune responses to rVSV-EBOV-GP and discuss the role of innate immune activation in therapeutic protection.
Collapse
Affiliation(s)
- Amanda N Pinski
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
12
|
Sharma NR, Gadhave K, Kumar P, Saif M, Khan MM, Sarkar DP, Uversky VN, Giri R. Analysis of the dark proteome of Chandipura virus reveals maximum propensity for intrinsic disorder in phosphoprotein. Sci Rep 2021; 11:13253. [PMID: 34168211 PMCID: PMC8225862 DOI: 10.1038/s41598-021-92581-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/07/2021] [Indexed: 02/05/2023] Open
Abstract
Chandipura virus (CHPV, a member of the Rhabdoviridae family) is an emerging pathogen that causes rapidly progressing influenza-like illness and acute encephalitis often leading to coma and death of the human host. Given several CHPV outbreaks in Indian sub-continent, recurring sporadic cases, neurological manifestation, and high mortality rate of this infection, CHPV is gaining global attention. The 'dark proteome' includes the whole proteome with special emphasis on intrinsically disordered proteins (IDP) and IDP regions (IDPR), which are proteins or protein regions that lack unique (or ordered) three-dimensional structures within the cellular milieu. These proteins/regions, however, play a number of vital roles in various biological processes, such as cell cycle regulation, control of signaling pathways, etc. and, therefore, are implicated in many human diseases. IDPs and IPPRs are also abundantly found in many viral proteins enabling their multifunctional roles in the viral life cycles and their capability to highjack various host systems. The unknown abundance of IDP and IDPR in CHPV, therefore, prompted us to analyze the dark proteome of this virus. Our analysis revealed a varying degree of disorder in all five CHPV proteins, with the maximum level of intrinsic disorder propensity being found in Phosphoprotein (P). We have also shown the flexibility of P protein using extensive molecular dynamics simulations up to 500 ns (ns). Furthermore, our analysis also showed the abundant presence of the disorder-based binding regions (also known as molecular recognition features, MoRFs) in CHPV proteins. The identification of IDPs/IDPRs in CHPV proteins suggests that their disordered regions may function as potential interacting domains and may also serve as novel targets for disorder-based drug designs.
Collapse
Affiliation(s)
- Nishi R Sharma
- School of Interdisciplinary Studies, Jamia Hamdard-Institute of Molecular Medicine (JH-IMM), Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India.
| | - Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, VPO Kamand, Kamand, Himachal Pradesh, 175005, India
| | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, VPO Kamand, Kamand, Himachal Pradesh, 175005, India
| | - Mohammad Saif
- School of Interdisciplinary Studies, Jamia Hamdard-Institute of Molecular Medicine (JH-IMM), Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Md M Khan
- School of Interdisciplinary Studies, Jamia Hamdard-Institute of Molecular Medicine (JH-IMM), Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Debi P Sarkar
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA.
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, 142290, Moscow, Russia.
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, VPO Kamand, Kamand, Himachal Pradesh, 175005, India.
| |
Collapse
|
13
|
Fallet B, Hao Y, Florova M, Cornille K, de Los Aires AV, Girelli Zubani G, Ertuna YI, Greiff V, Menzel U, Hammad K, Merkler D, Reddy ST, Weill JC, Reynaud CA, Pinschewer DD. Chronic Viral Infection Promotes Efficient Germinal Center B Cell Responses. Cell Rep 2020; 30:1013-1026.e7. [PMID: 31995746 PMCID: PMC6996002 DOI: 10.1016/j.celrep.2019.12.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/20/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022] Open
Abstract
Persistent viral infections subvert key elements of adaptive immunity. To compare germinal center (GC) B cell responses in chronic and acute lymphocytic choriomeningitis virus infection, we exploit activation-induced deaminase (AID) fate-reporter mice and perform adoptive B cell transfer experiments. Chronic infection yields GC B cell responses of higher cellularity than acute infections do, higher memory B cell and antibody secreting cell output for longer periods of time, a better representation of the late B cell repertoire in serum immunoglobulin, and higher titers of protective neutralizing antibodies. GC B cells of chronically infected mice are similarly hypermutated as those emerging from acute infection. They efficiently adapt to viral escape variants and even in hypermutation-impaired AID mutant mice, chronic infection selects for GC B cells with hypermutated B cell receptors (BCRs) and neutralizing antibody formation. These findings demonstrate that, unlike for CD8+ T cells, chronic viral infection drives a functional, productive, and protective GC B cell response. Chronic viral infection elicits potent and sustained germinal center (GC) responses Chronic infection triggers prolonged plasma cell and memory B cell output from GCs GC B cells hypermutate efficiently and are potently selected in chronic infection
Collapse
Affiliation(s)
- Bénédict Fallet
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Haus Petersplatz, 4009 Basel, Switzerland
| | - Yi Hao
- Development of the Immune System, Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, U1151-Centre National de la Recherche Scientifique, UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Marianna Florova
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Haus Petersplatz, 4009 Basel, Switzerland
| | - Karen Cornille
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Haus Petersplatz, 4009 Basel, Switzerland
| | - Alba Verge de Los Aires
- Development of the Immune System, Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, U1151-Centre National de la Recherche Scientifique, UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Giulia Girelli Zubani
- Development of the Immune System, Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, U1151-Centre National de la Recherche Scientifique, UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Yusuf I Ertuna
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Haus Petersplatz, 4009 Basel, Switzerland
| | - Victor Greiff
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland; Department of Immunology, University of Oslo, Oslo, Norway
| | - Ulrike Menzel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Karim Hammad
- Department of Pathology and Immunology, Division of Clinical Pathology, University & University Hospital of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, University & University Hospital of Geneva, Geneva, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jean-Claude Weill
- Development of the Immune System, Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, U1151-Centre National de la Recherche Scientifique, UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claude-Agnès Reynaud
- Development of the Immune System, Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, U1151-Centre National de la Recherche Scientifique, UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Daniel D Pinschewer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Haus Petersplatz, 4009 Basel, Switzerland.
| |
Collapse
|
14
|
Scher G, Schnell MJ. Rhabdoviruses as vectors for vaccines and therapeutics. Curr Opin Virol 2020; 44:169-182. [PMID: 33130500 PMCID: PMC8331071 DOI: 10.1016/j.coviro.2020.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/12/2020] [Accepted: 09/13/2020] [Indexed: 12/24/2022]
Abstract
Appropriate choice of vaccine vector is crucial for effective vaccine development. Rhabdoviral vectors, such as rabies virus and vesicular stomatitis virus, have been used in a variety of vaccine strategies. These viruses have small, easily manipulated genomes that can stably express foreign glycoproteins due to a well-established reverse genetics system for virus recovery. Both viruses have well-described safety profiles and have been demonstrated to be effective vaccine vectors. This review will describe how these Rhabdoviruses can be manipulated for use as vectors, their various applications as vaccines or therapeutics, and the advantages and disadvantages of their use.
Collapse
Affiliation(s)
- Gabrielle Scher
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA; Jefferson Vaccine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
15
|
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
|
16
|
Chiale C, Yarovinsky TO, Mason SW, Madina BR, Menon M, Krady MM, Moshkani S, Chattopadhyay Pal A, Almassian B, Rose JK, Robek MD, Nakaar V. Modified Alphavirus-Vesiculovirus Hybrid Vaccine Vectors for Homologous Prime-Boost Immunotherapy of Chronic Hepatitis B. Vaccines (Basel) 2020; 8:vaccines8020279. [PMID: 32517032 PMCID: PMC7349932 DOI: 10.3390/vaccines8020279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/12/2022] Open
Abstract
Virus-like vesicles (VLV) are hybrid vectors based on an evolved Semliki Forest virus (SFV) RNA replicon and the envelope glycoprotein (G) from vesicular stomatitis virus (VSV). Previously, we showed that VLV can be used to express protein antigens and generate protective antigen-specific CD8+ T cells. This report describes VLV vectors designed for enhanced protein expression and immunogenicity. Expressing hepatitis B virus (HBV) middle S antigen (MHBs) from VLV using a dual subgenomic promoter significantly increased MHBs-specific CD8+ T cell and antibody production in mice. Furthermore, envelope glycoprotein switch from VSV Indiana to the glycoprotein of Chandipura virus enabled prime-boost immunization and further increased responses to MHBs. Therapeutic efficacy was evaluated in a mouse model of chronic HBV infection initiated by HBV delivery with adeno-associated virus. Mice with lower or intermediate HBV antigen levels demonstrated a significant and sustained reduction of HBV replication following VLV prime-boost immunization. However, mice with higher HBV antigen levels showed no changes in HBV replication, emphasizing the importance of HBV antigenemia for implementing immunotherapies. This report highlights the potential of VLV dual promoter vectors to induce effective antigen-specific immune responses and informs the further development and evaluation of hybrid viral vaccine platforms for preventative and therapeutic purposes.
Collapse
Affiliation(s)
- Carolina Chiale
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (C.C.); (S.M.); (M.D.R.)
| | - Timur O. Yarovinsky
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA; (A.C.P.); (J.K.R.)
- CaroGen Corporation, Farmington, CT 06032, USA; (S.W.M.); (B.R.M.); (M.M.); (M.M.K.); (B.A.)
- Correspondence: (T.O.Y.); (V.N.)
| | - Stephen W. Mason
- CaroGen Corporation, Farmington, CT 06032, USA; (S.W.M.); (B.R.M.); (M.M.); (M.M.K.); (B.A.)
| | - Bhaskara R. Madina
- CaroGen Corporation, Farmington, CT 06032, USA; (S.W.M.); (B.R.M.); (M.M.); (M.M.K.); (B.A.)
| | - Manisha Menon
- CaroGen Corporation, Farmington, CT 06032, USA; (S.W.M.); (B.R.M.); (M.M.); (M.M.K.); (B.A.)
| | - Marie M. Krady
- CaroGen Corporation, Farmington, CT 06032, USA; (S.W.M.); (B.R.M.); (M.M.); (M.M.K.); (B.A.)
| | - Safiehkhatoon Moshkani
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (C.C.); (S.M.); (M.D.R.)
| | - Anasuya Chattopadhyay Pal
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA; (A.C.P.); (J.K.R.)
| | - Bijan Almassian
- CaroGen Corporation, Farmington, CT 06032, USA; (S.W.M.); (B.R.M.); (M.M.); (M.M.K.); (B.A.)
| | - John K. Rose
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA; (A.C.P.); (J.K.R.)
| | - Michael D. Robek
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (C.C.); (S.M.); (M.D.R.)
| | - Valerian Nakaar
- CaroGen Corporation, Farmington, CT 06032, USA; (S.W.M.); (B.R.M.); (M.M.); (M.M.K.); (B.A.)
- Correspondence: (T.O.Y.); (V.N.)
| |
Collapse
|
17
|
McWilliams IL, Kielczewski JL, Ireland DDC, Sykes JS, Lewkowicz AP, Konduru K, Xu BC, Chan CC, Caspi RR, Manangeeswaran M, Verthelyi D. Pseudovirus rVSVΔG-ZEBOV-GP Infects Neurons in Retina and CNS, Causing Apoptosis and Neurodegeneration in Neonatal Mice. Cell Rep 2020; 26:1718-1726.e4. [PMID: 30759384 DOI: 10.1016/j.celrep.2019.01.069] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/15/2018] [Accepted: 01/17/2019] [Indexed: 02/07/2023] Open
Abstract
Zaire Ebola virus (ZEBOV) survivors experience visual and CNS sequelae that suggests the ZEBOV glycoprotein can mediate neurotropism. Replication-competent rVSVΔG-ZEBOV-GP vaccine candidate is generally well tolerated; however, its potential neurotropism requires careful study. Here, we show that a single inoculation of rVSVΔG-ZEBOV-GP virus in neonatal C57BL/6 mice results in transient viremia, neurological symptoms, high viral titers in eyes and brains, and death. rVSVΔG-ZEBOV-GP infects the inner layers of the retina, causing severe retinitis. In the cerebellum, rVSVΔG-ZEBOV-GP infects neurons in the granular and Purkinje layers, resulting in progressive foci of apoptosis and neurodegeneration. The susceptibility to infection is not due to impaired type I IFN responses, although MDA5-/-, IFNβ-/-, and IFNAR1-/- mice have accelerated mortality. However, boosting interferon levels by co-administering poly(I:C) reduces viral titers in CNS and improves survival. Although these data should not be directly extrapolated to humans, they challenge the hypothesis that VSV-based vaccines are non-neurotropic.
Collapse
Affiliation(s)
- Ian L McWilliams
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | | | - Derek D C Ireland
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Jacob S Sykes
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Aaron P Lewkowicz
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Krishnamurthy Konduru
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Biying C Xu
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Chi-Chao Chan
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Mohanraj Manangeeswaran
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Daniela Verthelyi
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
18
|
Calado R, Duarte J, Borrego P, Marcelino JM, Bártolo I, Martin F, Figueiredo I, Almeida S, Graça L, Vítor J, Aires da Silva F, Dias I, Carrapiço B, Taveira N. A Prime-Boost Immunization Strategy with Vaccinia Virus Expressing Novel gp120 Envelope Glycoprotein from a CRF02_AG Isolate Elicits Cross-Clade Tier 2 HIV-1 Neutralizing Antibodies. Vaccines (Basel) 2020; 8:E171. [PMID: 32272637 PMCID: PMC7349027 DOI: 10.3390/vaccines8020171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 12/22/2022] Open
Abstract
Development of new immunogens eliciting broadly neutralizing antibodies (bNAbs) is a main priority for the HIV-1 vaccine field. Envelope glycoproteins from non-B-non-C HIV-1clades have not been fully explored as components of a vaccine. We produced Vaccinia viruses expressing a truncated version of gp120 (gp120t) from HIV-1 clades CRF02_AG, H, J, B, and C and examined their immunogenicity in mice and rabbits. Mice primed with the recombinant Vaccinia viruses and boosted with the homologous gp120t or C2V3C3 polypeptides developed antibodies that bind potently to homologous and heterologous envelope glycoproteins. Notably, a subset of mice immunized with the CRF02_AG-based envelope immunogens developed a cross-reactive neutralizing response against tier 2 HIV-1 Env-pseudoviruses and primary isolates. Rabbits vaccinated with the CRF02_AG-based envelope immunogens also generated potent binding antibodies, and one animal elicited antibodies that neutralized almost all (13 of 16, 81.3%) tier 2 HIV-1 isolates tested. Overall, the results suggest that the novel CRF02_AG-based envelope immunogens and prime-boost immunization strategy elicit the type of immune responses required for a preventive HIV-1 vaccine.
Collapse
Affiliation(s)
- Rita Calado
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
| | - Joana Duarte
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
| | - Pedro Borrego
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
| | - José Maria Marcelino
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, 2829-511 Monte de Caparica, Portugal
| | - Inês Bártolo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
| | - Francisco Martin
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
| | - Inês Figueiredo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
| | - Silvia Almeida
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, 1649-02 Lisboa, Portugal; (S.A.); (L.G.)
- Post-Graduate Program in Infectious Diseases, and Department of Social Medicine, Center of Health Sciences, Federal University of Espirito Santo, Vitória 29075-910, Brazil
| | - Luís Graça
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, 1649-02 Lisboa, Portugal; (S.A.); (L.G.)
| | - Jorge Vítor
- Biochemistry and Human Biology Dept, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Frederico Aires da Silva
- Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (F.A.d.S.); (I.D.); (B.C.)
| | - Inês Dias
- Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (F.A.d.S.); (I.D.); (B.C.)
| | - Belmira Carrapiço
- Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (F.A.d.S.); (I.D.); (B.C.)
| | - Nuno Taveira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (R.C.); (J.D.); (P.B.); (J.M.M.); (I.B.); (F.M.); (I.F.)
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, 2829-511 Monte de Caparica, Portugal
| |
Collapse
|
19
|
O'Donnell K, Marzi A. The Ebola virus glycoprotein and its immune responses across multiple vaccine platforms. Expert Rev Vaccines 2020; 19:267-277. [PMID: 32129120 DOI: 10.1080/14760584.2020.1738225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: For over 40 years, ebolaviruses have been responsible for sporadic outbreaks of severe and often fatal hemorrhagic fever in humans and nonhuman primates across western and central Africa. In December 2013, an unprecedented Ebola virus (EBOV) epidemic began in West Africa and resulted in the largest outbreak to date. The past and current epidemics in West Africa and the Democratic Republic of the Congo has focused attention on the potential vaccine platforms developed over the past 20 years.Areas covered: This review summarizes the extraordinary progress using a variety of vaccination platforms including DNA, subunit, and several viral vector approaches, replicating and non-replicating, incorporating the primary antigen of EBOV, the glycoprotein. These vaccine constructs have shown varying degrees of protective efficacy in the 'gold-standard' nonhuman primate model for EBOV infections and were immunogenic in human clinical trials.Expert commentary: A number of these vaccine platforms have moved into phase III clinical trials over the past years and with the recent approval of the first EBOV vaccine in the European Union and the USA there is a strong potential to prevent future outbreaks/epidemics of EBOV infections on the scale of the West African epidemic.
Collapse
Affiliation(s)
- Kyle O'Donnell
- Laboratory of Virology, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
20
|
Suschak JJ, Schmaljohn CS. Vaccines against Ebola virus and Marburg virus: recent advances and promising candidates. Hum Vaccin Immunother 2019; 15:2359-2377. [PMID: 31589088 DOI: 10.1080/21645515.2019.1651140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The filoviruses Ebola virus and Marburg virus are among the most dangerous pathogens in the world. Both viruses cause viral hemorrhagic fever, with case fatality rates of up to 90%. Historically, filovirus outbreaks had been relatively small, with only a few hundred cases reported. However, the recent West African Ebola virus outbreak underscored the threat that filoviruses pose. The three year-long outbreak resulted in 28,646 Ebola virus infections and 11,323 deaths. The lack of Food and Drug Administration (FDA) licensed vaccines and antiviral drugs hindered early efforts to contain the outbreak. In response, the global scientific community has spurred the advanced development of many filovirus vaccine candidates. Novel vaccine platforms, such as viral vectors and DNA vaccines, have emerged, leading to the investigation of candidate vaccines that have demonstrated protective efficacy in small animal and nonhuman primate studies. Here, we will discuss several of these vaccine platforms with a particular focus on approaches that have advanced into clinical development.
Collapse
Affiliation(s)
- John J Suschak
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | - Connie S Schmaljohn
- Headquarters Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
21
|
Wilmschen S, Schmitz JE, Kimpel J. Viral Vectors for the Induction of Broadly Neutralizing Antibodies against HIV. Vaccines (Basel) 2019; 7:vaccines7030119. [PMID: 31546894 PMCID: PMC6789710 DOI: 10.3390/vaccines7030119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 01/10/2023] Open
Abstract
Extensive research on generating an efficient HIV vaccine is ongoing. A major aim of HIV vaccines is the induction of long-lasting, broadly neutralizing antibodies (bnAbs) that can confer sterile immunity for a prolonged period of time. Several strategies have been explored to reach this goal, i.e. protein immunization, DNA, or viral vectors, or a combination thereof. In this review, we give an overview of approaches using viral vectors for the induction of HIV-specific bnAbs. Many pre-clinical studies were performed using various replication-competent and -incompetent vectors. Amongst them, poxviral and adenoviral vectors were the most prevalent ones. In many studies, viral vectors were combined with a DNA prime or a protein boost. However, neutralizing antibodies were mainly induced against the homologous HIV-1 vaccine strain or tier 1 viruses, and in rare cases, against tier 2 viruses, indicating the need for improved antigens and vaccination strategies. Furthermore, we also review next generation Env antigens that are currently being used in protein vaccination approaches and point out how they could be utilized in viral vectors.
Collapse
Affiliation(s)
- Sarah Wilmschen
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
22
|
Fathi A, Dahlke C, Addo MM. Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Hum Vaccin Immunother 2019; 15:2269-2285. [PMID: 31368826 PMCID: PMC6816421 DOI: 10.1080/21645515.2019.1649532] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The devastating Ebola virus (EBOV) outbreak in West Africa in 2013-2016 has flagged the need for the timely development of vaccines for high-threat pathogens. To be better prepared for new epidemics, the WHO has compiled a list of priority pathogens that are likely to cause future outbreaks and for which R&D efforts are, therefore, paramount (R&D Blueprint: https://www.who.int/blueprint/priority-diseases/en/ ). To this end, the detailed characterization of vaccine platforms is needed. The vesicular stomatitis virus (VSV) has been established as a robust vaccine vector backbone for infectious diseases for well over a decade. The recent clinical trials testing the vaccine candidate VSV-EBOV against EBOV disease now have added a substantial amount of clinical data and suggest VSV to be an ideal vaccine vector candidate for outbreak pathogens. In this review, we discuss insights gained from the clinical VSV-EBOV vaccine trials as well as from animal studies investigating vaccine candidates for Blueprint pathogens.
Collapse
Affiliation(s)
- Anahita Fathi
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Christine Dahlke
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Marylyn M Addo
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| |
Collapse
|
23
|
Bresk CA, Hofer T, Wilmschen S, Krismer M, Beierfuß A, Effantin G, Weissenhorn W, Hogan MJ, Jordan APO, Gelman RS, Montefiori DC, Liao HX, Schmitz JE, Haynes BF, von Laer D, Kimpel J. Induction of Tier 1 HIV Neutralizing Antibodies by Envelope Trimers Incorporated into a Replication Competent Vesicular Stomatitis Virus Vector. Viruses 2019; 11:v11020159. [PMID: 30769947 PMCID: PMC6409518 DOI: 10.3390/v11020159] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/04/2019] [Accepted: 02/12/2019] [Indexed: 12/21/2022] Open
Abstract
A chimeric vesicular stomatitis virus with the glycoprotein of the lymphocytic choriomeningitis virus, VSV-GP, is a potent viral vaccine vector that overcomes several of the limitations of wild-type VSV. Here, we evaluated the potential of VSV-GP as an HIV vaccine vector. We introduced genes for different variants of the HIV-1 envelope protein Env, i.e., secreted or membrane-anchored, intact or mutated furin cleavage site or different C-termini, into the genome of VSV-GP. We found that the addition of the Env antigen did not attenuate VSV-GP replication. All HIV-1 Env variants were expressed in VSV-GP infected cells and some were incorporated very efficiently into VSV-GP particles. Crucial epitopes for binding of broadly neutralizing antibodies against HIV-1 such as MPER (membrane-proximal external region), CD4 binding site, V1V2 and V3 loop were present on the surface of VSV-GP-Env particles. Binding of quaternary antibodies indicated a trimeric structure of VSV-GP incorporated Env. We detected high HIV-1 antibody titers in mice and showed that vectors expressing membrane-anchored Env elicited higher antibody titers than vectors that secreted Envs. In rabbits, Tier 1A HIV-1 neutralizing antibodies were detectable after prime immunization and titers further increased after boosting with a second immunization. Taken together, VSV-GP-Env is a promising vector vaccine against HIV-1 infection since this vector permits incorporation of native monomeric and/or trimeric HIV-1 Env into a viral membrane.
Collapse
Affiliation(s)
- C Anika Bresk
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Tamara Hofer
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Sarah Wilmschen
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Marina Krismer
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Anja Beierfuß
- Central Laboratory Animal Facility, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Grégory Effantin
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 38044 Grenoble, France.
| | - Winfried Weissenhorn
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 38044 Grenoble, France.
| | - Michael J Hogan
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Andrea P O Jordan
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Rebecca S Gelman
- Dana-Farber Cancer Institute, Harvard Medical School and Harvard School of Public Health, Boston, MA 02215, USA.
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Dorothee von Laer
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
24
|
Abstract
The West African Ebola virus (EBOV) epidemic has fast-tracked countermeasures for this rare, emerging zoonotic pathogen. Until 2013-2014, most EBOV vaccine candidates were stalled between the preclinical and clinical milestones on the path to licensure, because of funding problems, lack of interest from pharmaceutical companies, and competing priorities in public health. The unprecedented and devastating epidemic propelled vaccine candidates toward clinical trials that were initiated near the end of the active response to the outbreak. Those trials did not have a major impact on the epidemic but provided invaluable data on vaccine safety, immunogenicity, and, to a limited degree, even efficacy in humans. There are plenty of lessons to learn from these trials, some of which are addressed in this review. Better preparation is essential to executing an effective response to EBOV in the future; yet, the first indications of waning interest are already noticeable.
Collapse
Affiliation(s)
- Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba 93E 0J9, Canada
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA
| | - Andrea Marzi
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
| |
Collapse
|
25
|
Blasi M, Negri D, LaBranche C, Alam SM, Baker EJ, Brunner EC, Gladden MA, Michelini Z, Vandergrift NA, Wiehe KJ, Parks R, Shen X, Bonsignori M, Tomaras GD, Ferrari G, Montefiori DC, Santra S, Haynes BF, Moody MA, Cara A, Klotman ME. IDLV-HIV-1 Env vaccination in non-human primates induces affinity maturation of antigen-specific memory B cells. Commun Biol 2018; 1:134. [PMID: 30272013 PMCID: PMC6125466 DOI: 10.1038/s42003-018-0131-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 08/06/2018] [Indexed: 01/21/2023] Open
Abstract
HIV continues to be a major global health issue. In spite of successful prevention interventions and treatment methods, the development of an HIV vaccine remains a major priority for the field and would be the optimal strategy to prevent new infections. We showed previously that a single immunization with a SIV-based integrase-defective lentiviral vector (IDLV) expressing the 1086.C HIV-1-envelope induced durable, high-magnitude immune responses in non-human primates (NHPs). In this study, we have further characterized the humoral responses by assessing antibody affinity maturation and antigen-specific memory B-cell persistence in two vaccinated macaques. These animals were also boosted with IDLV expressing the heterologous 1176.C HIV-1-Env to determine if neutralization breadth could be increased, followed by evaluation of the injection sites to assess IDLV persistence. IDLV-Env immunization was associated with persistence of the vector DNA for up to 6 months post immunization and affinity maturation of antigen-specific memory B cells. Maria Blasi et al. report the anti-HIV-1 humoral response elicited in rhesus macaques following vaccination with an SIV-based integrase-defective lentiviral vector (IDLV). They find that a single IDLV-Env immunization induces continuous antibody avidity maturation and boosting with a heterologous HIV-1 Env results in lower peak antibody titers than autologous boost.
Collapse
Affiliation(s)
- Maria Blasi
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA. .,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA.
| | - Donatella Negri
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA.,Istituto Superiore di Sanità, Rome, 00161, Italy
| | - Celia LaBranche
- Department of Surgery, Duke University Medical Center, Durham, 27710, NC, USA
| | - S Munir Alam
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA.,Department of Pathology, Duke University Medical Center, Durham, 27710, NC, USA
| | - Erich J Baker
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Elizabeth C Brunner
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Morgan A Gladden
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | | | - Nathan A Vandergrift
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Kevin J Wiehe
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Robert Parks
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Xiaoying Shen
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Mattia Bonsignori
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA.,Department of Surgery, Duke University Medical Center, Durham, 27710, NC, USA
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, 27710, NC, USA
| | - David C Montefiori
- Department of Surgery, Duke University Medical Center, Durham, 27710, NC, USA
| | - Sampa Santra
- Beth Israel Deaconess Medical Center, Boston, 02215, MA, USA
| | - Barton F Haynes
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA.,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA
| | - Michael A Moody
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA.,Department of Pediatrics, Duke University Medical Center, Durham, 27710, NC, USA
| | - Andrea Cara
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA. .,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA. .,Istituto Superiore di Sanità, Rome, 00161, Italy.
| | - Mary E Klotman
- Department of Medicine, Duke University Medical Center, Durham, 27710, NC, USA. .,Duke Human Vaccine Institute, Duke University Medical Center, Durham, 27710, NC, USA.
| |
Collapse
|
26
|
Zemp F, Rajwani J, Mahoney DJ. Rhabdoviruses as vaccine platforms for infectious disease and cancer. Biotechnol Genet Eng Rev 2018; 34:122-138. [PMID: 29781359 DOI: 10.1080/02648725.2018.1474320] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The family Rhabdoviridae (RV) comprises a large, genetically diverse collection of single-stranded, negative sense RNA viruses from the order Mononegavirales. Several RV members are being developed as live-attenuated vaccine vectors for the prevention or treatment of infectious disease and cancer. These include the prototype recombinant Vesicular Stomatitis Virus (rVSV) and the more recently developed recombinant Maraba Virus, both species within the genus Vesiculoviridae. A relatively strong safety profile in humans, robust immunogenicity and genetic malleability are key features that make the RV family attractive vaccine platforms. Currently, the rVSV vector is in preclinical development for vaccination against numerous high-priority infectious diseases, with clinical evaluation underway for HIV/AIDS and Ebola virus disease. Indeed, the success of the rVSV-ZEBOV vaccine during the 2014-15 Ebola virus outbreak in West Africa highlights the therapeutic potential of rVSV as a vaccine vector for acute, life-threatening viral illnesses. The rVSV and rMaraba platforms are also being tested as 'oncolytic' cancer vaccines in a series of phase 1-2 clinical trials, after being proven effective at eliciting immune-mediated tumour regression in preclinical mouse models. In this review, we discuss the biological and genetic features that make RVs attractive vaccine platforms and the development and ongoing testing of rVSV and rMaraba strains as vaccine vectors for infectious disease and cancer.
Collapse
Affiliation(s)
- Franz Zemp
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada
| | - Jahanara Rajwani
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| | - Douglas J Mahoney
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,c Department of Microbiology, Immunology and Infectious Disease , Faculty of Medicine , University of Calgary , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| |
Collapse
|
27
|
Abstract
Vesicular stomatitis virus (VSV) is an oncolytic rhabdovirus and its glycoprotein G is widely used to pseudotype other viruses for gene therapy. Low-density lipoprotein receptor (LDL-R) serves as a major entry receptor for VSV. Here we report two crystal structures of VSV G in complex with two distinct cysteine-rich domains (CR2 and CR3) of LDL-R, showing that their binding sites on G are identical. We identify two basic residues on G, which are essential for its interaction with CR2 and CR3. Mutating these residues abolishes VSV infectivity even though VSV can use alternative receptors, indicating that all VSV receptors are members of the LDL-R family. Collectively, our data suggest that VSV G has specifically evolved to interact with receptor CR domains. These structural insights into the interaction between VSV G and host cell receptors provide a basis for the design of recombinant viruses with an altered tropism.
Collapse
|
28
|
Abstract
The filoviruses, Ebola virus (EBOV), and Marburg virus (MARV), are among the most pathogenic viruses known to man and the causative agents of viral hemorrhagic fever outbreaks in Africa with case fatality rates of up to 90%. Nearly 30,000 infections were observed in the latest EBOV epidemic in West Africa; previous outbreaks were much smaller, typically only affecting less than a few hundred people. Compared to other diseases such as AIDS or Malaria with millions of cases annually, filovirus hemorrhagic fever (FHF) is one of the neglected infectious diseases. There are no licensed vaccines or therapeutics available to treat EBOV and MARV infections; therefore, these pathogens can only be handled in maximum containment laboratories and are classified as select agents. Under these limitations, a very few laboratories worldwide conducted basic research and countermeasure development for EBOV and MARV since their respective discoveries in 1967 (MARV) and 1976 (EBOV). In this review, we discuss several vaccine platforms against EBOV and MARV, which have been assessed for their protective efficacy in animal models of FHF. The focus is on the most promising approaches, which were accelerated in clinical development (phase I-III trials) during the EBOV epidemic in West Africa.
Collapse
Affiliation(s)
- Pierce Reynolds
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
29
|
Masemann D, Boergeling Y, Ludwig S. Employing RNA viruses to fight cancer: novel insights into oncolytic virotherapy. Biol Chem 2017; 398:891-909. [DOI: 10.1515/hsz-2017-0103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/08/2017] [Indexed: 12/13/2022]
Abstract
Abstract
Within recent decades, viruses that specifically target tumor cells have emerged as novel therapeutic agents against cancer. These viruses do not only act via their cell-lytic properties, but also harbor immunostimulatory features to re-direct the tumor microenvironment and stimulate tumor-directed immune responses. Furthermore, oncolytic viruses are considered to be superior to classical cancer therapies due to higher selectivity towards tumor cell destruction and, consequently, less collateral damage of non-transformed healthy tissue. In particular, the field of oncolytic RNA viruses is rapidly developing since these agents possess alternative tumor-targeting strategies compared to established oncolytic DNA viruses. Thus, oncolytic RNA viruses have broadened the field of virotherapy facilitating new strategies to fight cancer. In addition to several naturally occurring oncolytic viruses, genetically modified RNA viruses that are armed to express foreign factors such as immunostimulatory molecules have been successfully tested in early clinical trials showing promising efficacy. This review aims to provide an overview of the most promising RNA viruses in clinical development, to summarize the current knowledge of clinical trials using these viral agents, and to discuss the main issues as well as future perspectives of clinical approaches using oncolytic RNA viruses.
Collapse
|
30
|
Baquero E, Albertini AA, Raux H, Abou-Hamdan A, Boeri-Erba E, Ouldali M, Buonocore L, Rose JK, Lepault J, Bressanelli S, Gaudin Y. Structural intermediates in the fusion-associated transition of vesiculovirus glycoprotein. EMBO J 2017; 36:679-692. [PMID: 28188244 DOI: 10.15252/embj.201694565] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 11/09/2022] Open
Abstract
Vesiculoviruses enter cells by membrane fusion, driven by a large, low-pH-induced, conformational change in the fusion glycoprotein G that involves transition from a trimeric pre-fusion toward a trimeric post-fusion state via monomeric intermediates. Here, we present the structure of the G fusion protein at intermediate pH for two vesiculoviruses, vesicular stomatitis virus (VSV) and Chandipura virus (CHAV), which is responsible for deadly encephalopathies. First, a CHAV G crystal structure shows two intermediate conformations forming a flat dimer of heterodimers. On virions, electron microscopy (EM) and tomography reveal monomeric spikes similar to one of the crystal conformations. In solution, mass spectrometry shows dimers of G. Finally, mutations at a dimer interface, involving fusion domains associated in an antiparallel manner to form an intermolecular β-sheet, affect G fusion properties. The location of the compensatory mutations restoring fusion activity strongly suggests that this interface is functionally relevant. This work reveals the range of G structural changes and suggests that G monomers can re-associate, through antiparallel interactions between fusion domains, into dimers that play a role at some early stage of the fusion process.
Collapse
Affiliation(s)
- Eduard Baquero
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Aurélie A Albertini
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Hélène Raux
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Abbas Abou-Hamdan
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Elisabetta Boeri-Erba
- CNRS, CEA, Institut de Biologie Structurale (IBS), Université Grenoble Alpes, Grenoble, France
| | - Malika Ouldali
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | | | - John K Rose
- Yale University School of Medicine, New Haven, CT, USA
| | - Jean Lepault
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Stéphane Bressanelli
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Yves Gaudin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| |
Collapse
|
31
|
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
|
32
|
Abstract
For 40 years ebolaviruses have been responsible for sporadic outbreaks of severe and often fatal hemorrhagic fever in humans and nonhuman primates. In December 2013 an unprecedented Zaire ebolavirus epidemic began in West Africa. Although "patient zero" has finally been reached after 2 years, the virus is again causing disease in the region. Currently there are no licensed vaccines or therapeutic countermeasures against ebolaviruses; however, the epidemic in West Africa has focused attention on the potential vaccine platforms developed over the past 15 years. There has been remarkable progress using a variety of platforms including DNA, subunit, and several viral vector approaches, replicating and non-replicating, which have shown varying degrees of protective efficacy in the "gold-standard" nonhuman primate models for Ebolavirus infections. A number of these vaccine platforms have moved into clinical trials over the past year with the hope of finding an efficacious vaccine to prevent future outbreaks/epidemics of Ebola hemorrhagic fever on the scale of the West African epidemic.
Collapse
Affiliation(s)
- Chad E Mire
- a Galveston National Laboratory, and Department of Microbiology and Immunology , University of Texas Medical Branch , Galveston , TX , USA
| | - Thomas W Geisbert
- a Galveston National Laboratory, and Department of Microbiology and Immunology , University of Texas Medical Branch , Galveston , TX , USA
| | - Heinz Feldmann
- b Laboratory of Virology, Division of Intramural Research , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton , MT , USA
| | - Andrea Marzi
- b Laboratory of Virology, Division of Intramural Research , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton , MT , USA
| |
Collapse
|
33
|
Vaccination with Vesicular Stomatitis Virus-Vectored Chimeric Hemagglutinins Protects Mice against Divergent Influenza Virus Challenge Strains. J Virol 2015; 90:2544-50. [PMID: 26676789 DOI: 10.1128/jvi.02598-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/11/2015] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Seasonal influenza virus infections continue to cause significant disease each year, and there is a constant threat of the emergence of reassortant influenza strains causing a new pandemic. Available influenza vaccines are variably effective each season, are of limited scope at protecting against viruses that have undergone significant antigenic drift, and offer low protection against newly emergent pandemic strains. "Universal" influenza vaccine strategies that focus on the development of humoral immunity directed against the stalk domains of the viral hemagglutinin (HA) show promise for protecting against diverse influenza viruses. Here, we describe such a strategy that utilizes vesicular stomatitis virus (VSV) as a vector for chimeric hemagglutinin (cHA) antigens. This vaccination strategy is effective at generating HA stalk-specific, broadly cross-reactive serum antibodies by both intramuscular and intranasal routes of vaccination. We show that prime-boost vaccination strategies provide protection against both lethal homologous and heterosubtypic influenza challenge and that protection is significantly improved with intranasal vaccine administration. Additionally, we show that vaccination with VSV-cHAs generates greater stalk-specific and cross-reactive serum antibodies than does vaccination with VSV-vectored full-length HAs, confirming that cHA-based vaccination strategies are superior at generating stalk-specific humoral immunity. VSV-vectored influenza vaccines that express chimeric hemagglutinin antigens offer a novel means for protecting against widely diverging influenza viruses. IMPORTANCE Universal influenza vaccination strategies should be capable of protecting against a wide array of influenza viruses, and we have developed such an approach utilizing a single viral vector system. The potent antibody responses that these vaccines generate are shown to protect mice against lethal influenza challenges with highly divergent viruses. Notably, intranasal vaccination offers significantly better protection than intramuscular vaccination in a lethal virus challenge model. The results described in this study offer insights into the mechanisms by which chimeric hemagglutinin (HA)-based vaccines confer immunity, namely, that the invariant stalk of cHA antigens is superior to full-length HA antigens at inducing cross-reactive humoral immune responses and that VSV-cHA vaccine-induced protection varies by site of inoculation, and contribute to the further development of universal influenza virus vaccines.
Collapse
|
34
|
Marzi A, Feldmann F, Geisbert TW, Feldmann H, Safronetz D. Vesicular stomatitis virus-based vaccines against Lassa and Ebola viruses. Emerg Infect Dis 2015; 21:305-7. [PMID: 25625358 PMCID: PMC4313664 DOI: 10.3201/eid2102.141649] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We demonstrated that previous vaccination with a vesicular stomatitis virus (VSV)–based Lassa virus vaccine does not alter protective efficacy of subsequent vaccination with a VSV-based Ebola virus vaccine. These findings demonstrate the utility of VSV-based vaccines against divergent viral pathogens, even when preexisting immunity to the vaccine vector is present.
Collapse
|
35
|
Retargeting Oncolytic Vesicular Stomatitis Virus to Human T-Cell Lymphotropic Virus Type 1-Associated Adult T-Cell Leukemia. J Virol 2015; 89:11786-800. [PMID: 26378177 PMCID: PMC4645320 DOI: 10.1128/jvi.01356-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/18/2015] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Adult T cell leukemia/lymphoma (ATL) is an aggressive cancer of CD4/CD25(+) T lymphocytes, the etiological agent of which is human T-cell lymphotropic virus type 1 (HTLV-1). ATL is highly refractory to current therapies, making the development of new treatments a high priority. Oncolytic viruses such as vesicular stomatitis virus (VSV) are being considered as anticancer agents since they readily infect transformed cells compared to normal cells, the former appearing to exhibit defective innate immune responses. Here, we have evaluated the efficacy and safety of a recombinant VSV that has been retargeted to specifically infect and replicate in transformed CD4(+) cells. This was achieved by replacing the single VSV glycoprotein (G) with human immunodeficiency virus type 1 (HIV-1) gp160 to create a hybrid fusion protein, gp160G. The resultant virus, VSV-gp160G, was found to only target cells expressing CD4 and retained robust oncolytic activity against HTLV-1 actuated ATL cells. VSV-gp160G was further noted to be highly attenuated and did not replicate efficiently in or induce significant cell death of primary CD4(+) T cells. Accordingly, VSV-gp160G did not elicit any evidence of neurotoxicity even in severely immunocompromised animals such as NOD/Shi-scid, IL-2Rγ-c-null (NSG) mice. Importantly, VSV-gp160G effectively exerted potent oncolytic activity in patient-derived ATL transplanted into NSG mice and facilitated a significant survival benefit. Our data indicate that VSV-gp160G exerts potent oncolytic efficacy against CD4(+) malignant cells and either alone or in conjunction with established therapies may provide an effective treatment in patients displaying ATL. IMPORTANCE Adult T cell leukemia (ATL) is a serious form of cancer with a high mortality rate. HTLV-1 infection is the etiological agent of ATL and, unfortunately, most patients succumb to the disease within a few years. Current treatment options have failed to significantly improve survival rate. In this study, we developed a recombinant strain of vesicular stomatitis virus (VSV) that specifically targets transformed CD4(+) T cells through replacement of the G protein of VSV with a hybrid fusion protein, combining domains from gp160 of HIV-1 and VSV-G. This modification eliminated the normally broad tropism of VSV and restricted infection to primarily the transformed CD4(+) cell population. This effect greatly reduced neurotoxic risk associated with VSV infection while still allowing VSV to effectively target ATL cells.
Collapse
|
36
|
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
|
37
|
Creation of matrix protein gene variants of two serotypes of vesicular stomatitis virus as prime-boost vaccine vectors. J Virol 2015; 89:6338-51. [PMID: 25855732 DOI: 10.1128/jvi.00222-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/30/2015] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED To take advantage of live recombinant vesicular stomatitis viruses (rVSVs) as vaccine vectors for their high yield and for their induction of strong and long-lasting immune responses, it is necessary to make live vaccine vectors safe for use without losing their immunogenicity. We have generated safer and highly efficient recombinant VSV vaccine vectors by combining the M51R mutation in the M gene of serotype VSV-Indiana (VSVInd) with a temperature-sensitive mutation (tsO23) of the VSVInd Orsay strain. In addition, we have generated two new serotype VSV-New Jersey (VSVNJ) vaccine vectors by combining M48R and M51R mutations with G22E and L110F mutations in the M gene, rVSVNJ(G22E M48R M51R) [rVSVNJ(GMM)] and VSVNJ(G22E M48R M51R L110F) [rVSVNJ(GMML)]. The combined mutations G21E, M51R, and L111F in the M protein of VSVInd significantly reduced the burst size of the virus by up to 10,000-fold at 37°C without affecting the level of protein expression. BHK21 cells and SH-SY5Y human neuroblastoma cells infected with rVSVInd(GML), rVSVNJ(GMM), and rVSVNJ(GMML) showed significantly reduced cytopathic effects in vitro at 37°C, and mice injected with 1 million infectious virus particles of these mutants into the brain showed no neurological dysfunctions or any other adverse effects. In order to increase the stability of the temperature-sensitive mutant, we have replaced the phenylalanine with alanine. This will change all three nucleotides from UUG (leucine) to GCA (alanine). The resulting L111A mutant showed the temperature-sensitive phenotype of rVSVInd(GML) and increased stability. Twenty consecutive passages of rVSVInd(GML) with an L111A mutation did not convert back to leucine (UUG) at position 111 in the M protein gene. IMPORTANCE Recombinant vesicular stomatitis viruses as live vaccine vectors are very effective in expressing foreign genes and inducing adaptive T cell and B cell immune responses. As with any other live viruses in humans or animals, the use of live rVSVs as vaccine vectors demands the utmost safety. Our strategy to attenuate rVSVInd by utilizing a temperature-sensitive assembly-defective mutation of L111A and combining it with an M51R mutation in the M protein of rVSVInd significantly reduced the pathogenicity of the virus while maintaining highly effective virus production. We believe our new temperature-sensitive M gene mutant of rVSVInd(GML) and M gene mutants of rVSVNJ(GMM) and rVSVNJ(GMML) add excellent vaccine vectors to the pool of live viral vectors.
Collapse
|
38
|
Structure of the low pH conformation of Chandipura virus G reveals important features in the evolution of the vesiculovirus glycoprotein. PLoS Pathog 2015; 11:e1004756. [PMID: 25803715 PMCID: PMC4372607 DOI: 10.1371/journal.ppat.1004756] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 02/20/2015] [Indexed: 11/19/2022] Open
Abstract
Chandipura virus (CHAV), a member of the vesiculovirus genus, is an emerging human pathogen. As for other rhabdoviruses, CHAV entry into susceptible cells is mediated by its single envelope glycoprotein G which is both involved in receptor recognition and fusion of viral and cellular membranes. Here, we have characterized the fusion properties of CHAV-G. As for vesicular stomatitis virus (VSV, the prototype of the genus) G, fusion is triggered at low pH below 6.5. We have also analyzed the biochemical properties of a soluble form of CHAV-G ectodomain (CHAV-Gth, generated by thermolysin limited-proteolysis of recombinant VSV particles in which the G gene was replaced by that of CHAV). The overall behavior of CHAV-Gth is similar to that previously reported for VSV-Gth. Particularly, CHAV-Gth pre-fusion trimer is not stable in solution and low-pH-induced membrane association of CHAV-Gth is reversible. Furthermore, CHAV-Gth was crystallized in its low pH post-fusion conformation and its structure was determined at 3.6Å resolution. An overall comparison of this structure with the previously reported VSV-Gth post-fusion conformation, shows a high structural similarity as expected from the comparison of primary structure. Among the three domains of G, the pleckstrin homology domain (PHD) appears to be the most divergent and the largest differences are confined to the secondary structure of the major antigenic site of rhabdoviruses. Finally, local differences indicate that CHAV has evolved alternate structural solutions in hinge regions between PH and fusion domains but also distinct pH sensitive switches. Globally the comparison between the post fusion conformation of CHAV and VSV-G highlights several features essential for the protein's function. It also reveals the remarkable plasticity of G in terms of local structures.
Collapse
|
39
|
Schell JB, Bahl K, Folta-Stogniew E, Rose N, Buonocore L, Marx PA, Gambhira R, Rose JK. Antigenic requirement for Gag in a vaccine that protects against high-dose mucosal challenge with simian immunodeficiency virus. Virology 2015; 476:405-412. [PMID: 25591175 DOI: 10.1016/j.virol.2014.12.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/16/2014] [Accepted: 12/17/2014] [Indexed: 11/28/2022]
Abstract
We reported previously on a vaccine approach that conferred apparent sterilizing immunity to SIVsmE660. The vaccine regimen employed a prime-boost using vectors based on recombinant vesicular stomatitis virus (VSV) and an alphavirus replicon expressing either SIV Gag or SIV Env. In the current study, we tested the ability of vectors expressing only the SIVsmE660 Env protein to protect macaques against the same high-dose mucosal challenge. Animals developed neutralizing antibody levels comparable to or greater than seen in the previous vaccine study. When the vaccinated animals were challenged with the same high-dose of SIVsmE660, all became infected. While average peak viral loads in animals were slightly lower than those of previous controls, the viral set points were not significantly different. These data indicate that Gag, or the combination of Gag and Env are required for the generation of apparent sterilizing immunity to the SIVsmE660 challenge.
Collapse
Affiliation(s)
- John B Schell
- Yale University School of Medicine, New Haven, CT, United States
| | - Kapil Bahl
- Yale University School of Medicine, New Haven, CT, United States
| | - Ewa Folta-Stogniew
- Yale University School of Medicine, New Haven, CT, United States; Keck Biophysical Resource Facility, New Haven, CT, United States
| | - Nina Rose
- Yale University School of Medicine, New Haven, CT, United States
| | - Linda Buonocore
- Yale University School of Medicine, New Haven, CT, United States
| | - Preston A Marx
- Tulane National Primate Research Center, Covington, LA, United States
| | - Ratish Gambhira
- Tulane National Primate Research Center, Covington, LA, United States
| | - John K Rose
- Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
40
|
A viable recombinant rhabdovirus lacking its glycoprotein gene and expressing influenza virus hemagglutinin and neuraminidase is a potent influenza vaccine. J Virol 2014; 89:2820-30. [PMID: 25540378 DOI: 10.1128/jvi.03246-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED The emergence of novel influenza viruses that cause devastating human disease is an ongoing threat and serves as an impetus for the continued development of novel approaches to influenza vaccines. Influenza vaccine development has traditionally focused on producing humoral and/or cell-mediated immunity, often against the viral surface glycoproteins hemagglutinin (HA) and neuraminidase (NA). Here, we describe a new vaccine candidate that utilizes a replication-defective vesicular stomatitis virus (VSV) vector backbone that lacks the native G surface glycoprotein gene (VSVΔG). The expression of the H5 HA of an H5N1 highly pathogenic avian influenza virus (HPAIV), A/Vietnam/1203/04 (VN1203), and the NA of the mouse-adapted H1N1 influenza virus A/Puerto Rico/8/34 (PR8) in the VSVΔG vector restored the ability of the recombinant virus to replicate in cell culture, without the requirement for the addition of trypsin. We show here that this recombinant virus vaccine candidate was nonpathogenic in mice when given by either the intramuscular or intranasal route of immunization and that the in vivo replication of VSVΔG-H5N1 is profoundly attenuated. This recombinant virus also provided protection against lethal H5N1 infection after a single dose. This novel approach to vaccination against HPAIVs may be widely applicable to other emerging strains of influenza virus. IMPORTANCE Preparation for a potentially catastrophic influenza pandemic requires novel influenza vaccines that are safe, can be produced and administered quickly, and are effective, both soon after administration and for a long duration. We have created a new influenza vaccine that utilizes an attenuated vesicular stomatitis virus (VSV) vector, to deliver and express influenza virus proteins against which vaccinated animals develop potent antibody responses. The influenza virus hemagglutinin and neuraminidase proteins, expressed on the surface of VSV particles, allowed this vaccine to grow in cell culture and induced a potent antibody response in mice that was effective against infection with a lethal influenza virus. The mice showed no adverse reactions to the vaccine, and they were protected against an otherwise lethal influenza infection after only 14 days postvaccination and after as many as 140 days postvaccination. The ability to rapidly produce this safe and effective vaccine in cell culture is additionally advantageous.
Collapse
|
41
|
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
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
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
|
46
|
Carter JR, Nawtaisong P, Balaraman V, Fraser MJ. Design and analysis of hammerhead ribozyme activity against an artificial gene target. Methods Mol Biol 2014; 1103:57-66. [PMID: 24318886 PMCID: PMC4219533 DOI: 10.1007/978-1-62703-730-3_5] [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] [Indexed: 06/02/2023]
Abstract
In vitro cleavage assays are routinely conducted to properly assess the catalytic activity of hammerhead ribozymes (HHR) against target RNA molecules like dengue virus RNA. These experiments are performed for initial assessment of HHR catalysis in a cell-free system and have been simplified by the substitution of agarose gel electrophoresis for SDS-PAGE. Substituting mobility assays enables the analysis of ribozymes in a more rapid fashion without radioisotopes. Here we describe the in vitro transcription of an HHR and corresponding target from T7-promoted plasmids into RNA molecules leading to the analysis of HHR activity against the RNA target by in vitro cleavage assays.
Collapse
Affiliation(s)
- James R Carter
- Department of Biology, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA
| | | | | | | |
Collapse
|
47
|
Mire CE, Versteeg KM, Cross RW, Agans KN, Fenton KA, Whitt MA, Geisbert TW. Single injection recombinant vesicular stomatitis virus vaccines protect ferrets against lethal Nipah virus disease. Virol J 2013; 10:353. [PMID: 24330654 PMCID: PMC3878732 DOI: 10.1186/1743-422x-10-353] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/03/2013] [Indexed: 11/10/2022] Open
Abstract
Background Nipah virus (NiV) is a highly pathogenic zoonotic agent in the family Paramyxoviridae that is maintained in nature by bats. Outbreaks have occurred in Malaysia, Singapore, India, and Bangladesh and have been associated with 40 to 75% case fatality rates. There are currently no vaccines or postexposure treatments licensed for combating human NiV infection. Methods and results Four groups of ferrets received a single vaccination with different recombinant vesicular stomatitis virus vectors expressing: Group 1, control with no glycoprotein; Group 2, the NiV fusion protein (F); Group 3, the NiV attachment protein (G); and Group 4, a combination of the NiV F and G proteins. Animals were challenged intranasally with NiV 28 days after vaccination. Control ferrets in Group 1 showed characteristic clinical signs of NiV disease including respiratory distress, neurological disorders, viral load in blood and tissues, and gross lesions and antigen in target tissues; all animals in this group succumbed to infection by day 8. Importantly, all specifically vaccinated ferrets in Groups 2-4 showed no evidence of clinical illness and survived challenged. All animals in these groups developed anti-NiV F and/or G IgG and neutralizing antibody titers. While NiV RNA was detected in blood at day 6 post challenge in animals from Groups 2-4, the levels were orders of magnitude lower than animals from control Group 1. Conclusions These data show protective efficacy against NiV in a relevant model of human infection. Further development of this technology has the potential to yield effective single injection vaccines for NiV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd,, Galveston, TX, USA.
| |
Collapse
|
48
|
Eyles JE, Johnson JE, Megati S, Roopchand V, Cockle PJ, Weeratna R, Makinen S, Brown TP, Lang S, Witko SE, Kotash CS, Li J, West K, Maldonado O, Falconer DJ, Lees C, Smith GJ, White P, Wright P, Loudon PT, Merson JR, Jansen KU, Sidhu MK. Nonreplicating vaccines can protect african green monkeys from the memphis 37 strain of respiratory syncytial virus. J Infect Dis 2013; 208:319-29. [PMID: 23596321 DOI: 10.1093/infdis/jit169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We evaluated the immunological responses of African green monkeys immunized with multiple F and G protein-based vaccines and assessed protection against the Memphis 37 strain of respiratory syncytial virus (RSV). METHODS Monkeys were immunized with F and G proteins adjuvanted with immunostimulatory (CpG) oligodeoxyribonucleotides admixed with either Alhydrogel or ISCOMATRIX adjuvant. Delivery of F and G proteins via replication incompetent recombinant vesicular stomatitis viruses (VSVs) and human adenoviruses was also evaluated. Mucosally or parenterally administered recombinant adenoviruses were used in prime-boost regimens with adjuvanted proteins or recombinant DNA. RESULTS Animals primed by intranasal delivery of recombinant adenoviruses, and boosted by intramuscular injection of adjuvanted F and G proteins, developed neutralizing antibodies and F/G protein-specific T cells and were protected from RSV infection. Intramuscular injections of Alhydrogel (plus CpG) adjuvanted F and G proteins reduced peak viral loads in the lungs of challenged monkeys. Granulocyte numbers were not significantly elevated, relative to controls, in postchallenge bronchoalveolar lavage samples from vaccinated animals. CONCLUSIONS This study has validated the use of RSV (Memphis 37) in an African green monkey model of intranasal infection and identified nonreplicating vaccines capable of eliciting protection in this higher species challenge model.
Collapse
Affiliation(s)
- Jim E Eyles
- Pfizer Vaccine Research, La Jolla, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Delivery of viral-vectored vaccines by B cells represents a novel strategy to accelerate CD8+ T-cell recall responses. Blood 2013; 121:2432-9. [DOI: 10.1182/blood-2012-06-438481] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Key PointsUsing B cells to target antigens into the follicular regions represents a novel approach to accelerate CD8+ T-cell recall responses.
Collapse
|
50
|
|