1
|
Mdluli T, Wollen-Roberts S, Merbah M, Beckman B, Li Y, Alrubayyi A, Curtis DJ, Shubin Z, Barrera MD, Boeckelman J, Duncan S, Thapa P, Kim D, Costanzo MC, Bai H, Dearlove BL, Hooper JW, Kwilas SA, Paquin-Proulx D, Eller MA, Eller LA, Kibuuka H, Mwesigwa B, Kosgei J, Sawe F, Oyieko J, Ntinginya N, Mwakisisile J, Jani I, Viegas E, Iroezindu M, Akintunde A, Paolino K, Robb ML, Ward L, McLean C, Luhn K, Robinson C, Ake JA, Rolland M. Ebola virus vaccination elicits Ebola virus-specific immune responses without substantial cross-reactivity to other filoviruses. Sci Transl Med 2025; 17:eadq2496. [PMID: 40173257 DOI: 10.1126/scitranslmed.adq2496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/30/2024] [Accepted: 03/05/2025] [Indexed: 04/04/2025]
Abstract
The Janssen Ebola virus (EBOV) vaccine consists of the adenovirus type 26 vector encoding the EBOV glycoprotein (GP) (Ad26.ZEBOV) and the modified vaccinia Ankara (MVA) vector encoding GP from EBOV, Sudan virus, and Marburg virus and nucleoprotein from Tai Forest virus (MVA-BN-Filo) administered 8 weeks later. We conducted a systems immunology analysis of antibody-mediated and cellular immune responses induced after two immunizations with either vaccine used first. The response to vaccination was EBOV GP specific and defined by high antibody binding, Fc effector, and neutralizing responses with CD4 T cell responses also contributing. The vaccine-induced antibody profile did not distinguish people living with or without HIV-1. Samples from 48 survivors and 121 contacts from the 2007 Ugandan Bundibugyo virus epidemic also showed minimal cross-reactivity to other filovirus proteins after infection and exposure. The lack of cross-reactivity suggests that different multivalent vaccine candidates are required to provide broad protection across filoviruses.
Collapse
Affiliation(s)
- Thembi Mdluli
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Suzanne Wollen-Roberts
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Mélanie Merbah
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Bradley Beckman
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Yifan Li
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Aljawharah Alrubayyi
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Daniel J Curtis
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Zhanna Shubin
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Michael D Barrera
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Jacob Boeckelman
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Shayla Duncan
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Pallavi Thapa
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Dohoon Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Margaret C Costanzo
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Hongjun Bai
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Bethany L Dearlove
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Jay W Hooper
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD 21702, USA
| | - Steven A Kwilas
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD 21702, USA
| | - Dominic Paquin-Proulx
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Michael A Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Leigh Anne Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Hannah Kibuuka
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Betty Mwesigwa
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Josphat Kosgei
- Henry M. Jackson Foundation Medical Research International, Nairobi, Kenya
- Kenya Medical Research Institute, US Army Medical Research Directorate-Africa, Kericho, Kenya
| | - Fredrick Sawe
- Henry M. Jackson Foundation Medical Research International, Nairobi, Kenya
- Kenya Medical Research Institute, US Army Medical Research Directorate-Africa, Kericho, Kenya
| | - Janet Oyieko
- Henry M. Jackson Foundation Medical Research International, Nairobi, Kenya
- Kenya Medical Research Institute, US Army Medical Research Directorate-Africa, Kericho, Kenya
| | - Nyanda Ntinginya
- National Institute for Medical Research-Mbeya Medical Research Center, Mbeya, Tanzania
| | - Joel Mwakisisile
- National Institute for Medical Research-Mbeya Medical Research Center, Mbeya, Tanzania
| | - Ilesh Jani
- Polana Caniço Health Research and Training Center, Maputo, Mozambique
| | - Edna Viegas
- Polana Caniço Health Research and Training Center, Maputo, Mozambique
| | - Michael Iroezindu
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation Medical Research International, Abuja, Nigeria
| | - Akindiran Akintunde
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
- Henry M. Jackson Foundation Medical Research International, Abuja, Nigeria
- US Army Medical Research Directorate-Africa, Abuja, Nigeria
| | - Kristopher Paolino
- Clinical Trials Center, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Institute of Global Health and Translational Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Merlin L Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Lucy Ward
- US Department of Defense (DOD) Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, MD 21702, USA
| | | | - Kerstin Luhn
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | | | - Julie A Ake
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Morgane Rolland
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| |
Collapse
|
2
|
Robinson-McCarthy LR, Zirckel KE, Simmons HC, Le Sage V, McCarthy KR. A replicating recombinant vesicular stomatitis virus model for dairy cattle H5N1 influenza virus glycoprotein evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640582. [PMID: 40060653 PMCID: PMC11888439 DOI: 10.1101/2025.02.27.640582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
A panzootic of highly pathogenic avian influenza (HPAI) H5N1 viruses from clade 2.3.4.4b has triggered a multistate outbreak in United States dairy cattle and an unknown number of human infections. HPAI viruses are handled in specialized biocontainment facilities. Ethical considerations limit certain experimental evolution experiments aimed at assessing viral resistance to potential therapeutics. We have developed a replicating recombinant vesicular stomatitis virus (rVSV) where we replaced its glycoprotein with the hemagglutinin (HA) and neuraminidase (NA) genes of a 2.3.4.4b H5N1 virus (rVSV-H5N1dc2024), which is free of these constraints. This virus grows to high titers and encodes a fluorescent reporter to track infection. We demonstrate the utility of rVSV-H5N1dc2024 in neutralization experiments, evaluating antibody escape and characterization of resistance mutations to NA inhibitors. rVSV-H5N1dc2024 or similar viruses may accelerate efforts to develop and evaluate interventions against this emerging threat to human and animal health.
Collapse
Affiliation(s)
- Lindsey R Robinson-McCarthy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kylie E Zirckel
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Holly C Simmons
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Valerie Le Sage
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kevin R McCarthy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Rex EA, Seo D, Embry A, Arulanandam R, Spinelli MM, Diallo JS, Gammon DB. Activation and Evasion of the FEAR Pathway by RNA Viruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.22.609092. [PMID: 40060670 PMCID: PMC11888236 DOI: 10.1101/2024.08.22.609092] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
We recently identified the FACT-ETS-1 Antiviral Response (FEAR) pathway as an interferon-independent innate immune response that restricts DNA virus replication and is countered by poxvirus-encoded A51R proteins (Rex et al., 2024, Nature Microbiology). The human FEAR pathway is mediated by the FACT complex, consisting of hSpt16 and SSRP1 subunits, that remodels chromatin to activate expression of the antiviral transcription factor, ETS-1. To counter this pathway, poxvirus A51R proteins tether SUMOylated hSpt16 subunits to microtubules to prevent ETS-1 expression. While these observations indicate a role for the FEAR pathway in DNA virus restriction, it was unclear if RNA viruses interact with this pathway. Here, we show that RNA viruses are also restricted by the FEAR pathway, yet encode mechanisms distinct from poxviruses to counter this response. We show vesicular stomatitis virus (VSV), a rhabdovirus, utilizes its matrix (M) protein to promote proteasome-dependent degradation of SUMOylated hSpt16 and to block ETS-1 nuclear import. Strains encoding mutant M proteins that cannot antagonize the FEAR pathway exhibit replication defects in human cells that can be rescued by hSpt16 or ETS-1 depletion. Moreover, FACT inhibitor treatment enhanced the replication of oncolytic VSV strains encoding defective M proteins in restrictive cancer cells, suggesting FEAR pathway inhibition may improve oncolytic virotherapy. Strikingly, we provide evidence that the inability of VSV M to degrade SUMOylated Spt16 in lepidopteran insect cells results in abortive infection, suggesting VSV-Spt16 interactions influence virus host range. Lastly, we show that human and murine paramyxovirus target SUMOylated Spt16 proteins for degradation in human and murine cells utilizing a conserved N-terminal motif in their accessory "C" proteins. Collectively, our study illustrates that DNA and RNA viruses have independently evolved diverse mechanisms to antagonize SUMOylated host Spt16 proteins, underscoring the physiological importance of the FEAR pathway to antiviral immunity.
Collapse
Affiliation(s)
- Emily A Rex
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Dahee Seo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Aaron Embry
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Rozanne Arulanandam
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Marcus M Spinelli
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Jean-Simon Diallo
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Don B Gammon
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
4
|
Yi J, Luo H, Zhang K, Lv L, Li S, Jiang Y, Zhou Y, Wei Z, Liu C. The Pseudotyped Replication-Deficient VSV with Spike from PEDV Induces Neutralizing Antibody Against PEDV. Vaccines (Basel) 2025; 13:223. [PMID: 40266086 DOI: 10.3390/vaccines13030223] [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: 01/21/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Porcine epidemic diarrhea virus (PEDV) is a significant pathogen in swine, causing substantial economic losses worldwide. Despite the availability of existing vaccines, there is a critical need for novel vaccine platforms that ensure robust protection while maintaining safety. METHODS A recombinant replication-deficient vesicular stomatitis virus (VSV) vaccine, rVSV∆G-PEDV-S, was developed by pseudotyping the virus with the spike (S) protein from PEDV. To achieve high-titer pseudotyped rVSV particles, a stable Huh7 cell line expressing the PEDV S protein (Huh7-PEDV-S) was generated. The infectivity and replication capacity of rVSV∆G-PEDV-S were evaluated in PEDV-susceptible cell lines and Huh7-PEDV-S cells. The vaccine's immunogenicity and safety were assessed in BALB/c mice vaccinated intramuscularly with rVSV∆G-PEDV-S. RESULTS The pseudotyped rVSV∆G-PEDV-S demonstrated infectivity in PEDV-susceptible cell lines and robust replication in Huh7-PEDV-S cells, while remaining replication-deficient in non-complementary cells. In vaccinated BALB/c mice, the vaccine elicited a strong humoral immune response, characterized by high levels of PEDV S1-specific IgG and neutralizing antibodies. No adverse effects, including weight loss or behavioral changes, were observed in the vaccinated mice, confirming the vaccine's safety. CONCLUSIONS The rVSV∆G-PEDV-S vaccine represents a promising platform for controlling PEDV outbreaks. Its replication-deficient design and pseudotyping methodology ensure safety and adaptability to emerging PEDV variants. These findings highlight the potential of rVSV∆G-PEDV-S as a safe and effective solution to the ongoing challenges posed by PEDV.
Collapse
Affiliation(s)
- Jingxuan Yi
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Huaye Luo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Kang Zhang
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Lilei Lv
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Siqi Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yifeng Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Yanjun Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Zuzhang Wei
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
| | - Changlong Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
5
|
Chai S, Wang L, Du H, Jiang H. Achievement and Challenges in Orthohantavirus Vaccines. Vaccines (Basel) 2025; 13:198. [PMID: 40006744 PMCID: PMC11861054 DOI: 10.3390/vaccines13020198] [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: 12/31/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Orthohantaviruses (also known as hantaviruses) are pathogens that cause two distinct, yet related forms of severe human disease: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). These diseases pose a significant threat to global public health due to their high case fatality rates, which can range from 1% to 50%. In recent years, an increasing number of countries and regions have reported human cases, underscoring the urgent need for improved understanding, prevention, and treatment strategies. Given the severity of these diseases and the lack of specific post-exposure antiviral treatments, preventive measures are critical. For several decades, substantial efforts have been dedicated to developing orthohantavirus vaccines, leading to significant advancements. The first large-scale deployment involved inactivated vaccines, which played a crucial role in reducing HFRS incidence in South Korea and China. Subunit vaccines, viral vector vaccines, and virus-like particle (VLP) vaccines have also been extensively researched. Nucleic acid vaccines, including both mRNA and DNA vaccines, hold the greatest potential for future development due to their rapid design and production cycles, ability to elicit robust immune responses, ease of storage and transportation, and adaptable production platforms. Ongoing advancements in computer technology and artificial intelligence promise to further enhance the development of more effective orthohantavirus vaccines.
Collapse
Affiliation(s)
- Shiqi Chai
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| | - Limei Wang
- Department of Microbiology and Pathogenic Biology, School of Basic Medicine, The Fourth Military Medical University, Xi’an 710032, China;
| | - Hong Du
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| | - Hong Jiang
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| |
Collapse
|
6
|
Adeleke RA, Sahler J, Choi A, Roth K, Upadhye V, Ezzatpour S, Imbiakha B, Khomandiak S, Diaz A, Whittaker GR, Jager MC, August A, Buchholz DW, Aguilar HC. Replication-incompetent VSV-based vaccine elicits protective responses against SARS-CoV-2 and influenza virus. SCIENCE ADVANCES 2025; 11:eadq4545. [PMID: 39879304 PMCID: PMC11777205 DOI: 10.1126/sciadv.adq4545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses lead to severe respiratory illnesses and death in humans, exacerbated in individuals with underlying health conditions, remaining substantial global public health concerns. Here, we developed a bivalent replication-incompetent single-cycle pseudotyped vesicular stomatitis virus vaccine that incorporates both a prefusion-stabilized SARS-CoV-2 spike protein lacking a furin cleavage site and a full-length influenza A virus neuraminidase protein. Vaccination of K18-hACE2 or C57BL/6J mouse models generated durable levels of neutralizing antibodies, T cell responses, and protection from morbidity and mortality upon challenge with either virus. Furthermore, the vaccine provided heterologous protection upon challenge with a different influenza virus strain, supporting the advantage of using NA to increase the breadth of vaccine protection. Now, no bivalent vaccine is approved for use against both SARS-CoV-2 and influenza virus. Our study supports using this platform to develop safe and efficient vaccines against multiple viruses.
Collapse
Affiliation(s)
- Richard A. Adeleke
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Julie Sahler
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Kyle Roth
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Viraj Upadhye
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Brian Imbiakha
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Solomiia Khomandiak
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Annika Diaz
- Department of Population Medicine and Diagnostic Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Mason C. Jager
- Department of Population Medicine and Diagnostic Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - David W. Buchholz
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| |
Collapse
|
7
|
Anhalt H, Marzi A. Generation, Recovery, and Propagation of a Recombinant Vesicular Stomatitis Virus Expressing the Marburg Virus Glycoprotein. Methods Mol Biol 2025; 2877:67-74. [PMID: 39585614 DOI: 10.1007/978-1-0716-4256-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Vesicular stomatitis virus (VSV)-based vaccination has shown protective efficacy against filovirus infection. Following the approval of a VSV-based vaccine against Ebola virus, there have been efforts toward applying the same platform for other filoviruses, including Marburg virus (MARV) and Sudan virus. Because these vaccines express filovirus glycoproteins, they are also a valuable tool to study filovirus entry under biosafety level 2 conditions. In the protocol described below, we outline how to genetically manipulate a full-length VSV vector by removing the native VSV glycoprotein and replacing it with the surface-expressed MARV glycoprotein. In addition, we describe the recovery procedure of these recombinant, full-length VSVs and detail the necessary steps of virus propagation.
Collapse
Affiliation(s)
- Hanna Anhalt
- 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
|
8
|
Suder EL, Hume AJ. Transcription- and Replication-Competent Virus-like Particle Systems for Marburg Virus. Methods Mol Biol 2025; 2877:141-154. [PMID: 39585619 DOI: 10.1007/978-1-0716-4256-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Here, we describe the transcription- and replication-competent virus-like particle (trVLP) system for Marburg virus (MARV), which recapitulates transcription and replication of the viral genome in addition to viral particle assembly, egress, and entry. This protocol includes instructions for transfections for producer and acceptor cells and the use of trVLPs for infection.
Collapse
Affiliation(s)
- Ellen L Suder
- Department of Virology, Immunology & Microbiology, Chobanian & Avedisian School of Medicine and National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Adam J Hume
- Department of Virology, Immunology & Microbiology, Chobanian & Avedisian School of Medicine and National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA.
| |
Collapse
|
9
|
Richards PT, Briseño JAA, Brunton BA, Maury W. In Vivo Investigation of Filovirus Glycoprotein-Mediated Infection in a BSL2 Setting. Methods Mol Biol 2025; 2877:183-198. [PMID: 39585622 PMCID: PMC11727417 DOI: 10.1007/978-1-0716-4256-6_13] [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: 11/26/2024]
Abstract
Highly pathogenic viruses in the Filoviridae family are causative agents of filovirus disease (FVD). Ebola virus (EBOV) is one such member and, of all filoviruses, represents the largest threat to global public health. The study of FVD has been hampered by the lack of tools to study filovirus infection outside maximum containment laboratories. Recombinant vesicular stomatitis virus (VSV) lacking its native glycoprotein and expressing a filovirus glycoprotein (VSV-filo GP) has improved our understanding of GP-mediated host-cell interactions as well as adaptive and humoral immune responses in in vitro and in vivo studies. Furthermore, mouse models suitable for these studies are readily available. Here, we describe multiple injection routes for investigating filovirus GP-mediated infection and pathogenesis using VSV-filo GP and interferon α/β receptor-deficient (Ifnar-/-) mice as models. These tools can be safely used outside maximum containment laboratories, are cost effective, and easy to manipulate.
Collapse
Affiliation(s)
- Paige T Richards
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | | | - Bethany A Brunton
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA.
- Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
10
|
Hicks P, Manzoni TB, Westover JB, Petch RJ, Roper B, Gowen BB, Bates P. Safety, Immunogenicity, and Efficacy of a Recombinant Vesicular Stomatitis Virus Vectored Vaccine Against Severe Fever with Thrombocytopenia Syndrome Virus and Heartland Bandavirus. Vaccines (Basel) 2024; 12:1403. [PMID: 39772063 PMCID: PMC11728676 DOI: 10.3390/vaccines12121403] [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: 11/20/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Severe fever with thrombocytopenia syndrome virus (SFTSV) is a recently emerged tickborne virus in east Asia with over 18,000 confirmed cases. With a high case fatality ratio, SFTSV has been designated a high priority pathogen by the WHO and the NIAID. Despite this, there are currently no approved therapies or vaccines to treat or prevent SFTS. Vesicular stomatitis virus (VSV) represents an FDA-approved vaccine platform that has been considered for numerous viruses due to its low sero-prevalence in humans, ease in genetic manipulation, and promiscuity in incorporating foreign glycoproteins into its virions. METHODS In this study, we developed a recombinant VSV (rVSV) expressing the SFTSV glycoproteins Gn/Gc (rVSV-SFTSV) and assessed its safety, immunogenicity, and efficacy in C57BL/6, Ifnar-/-, and AG129 mice. RESULTS We demonstrate that rVSV-SFTSV is safe when given to immunocompromised animals and is not neuropathogenic when injected intracranially into young immunocompetent mice. Immunization of wild type (C57BL/6) and Ifnar-/- mice with rVSV-SFTSV resulted in high levels of neutralizing antibodies and protection in a lethal SFTSV challenge model. Additionally, passive transfer of sera from immunized Ifnar-/- mice into naïve animals was protective when given pre- or post-exposure. Finally, we demonstrate that immunization with rVSV-SFTSV cross protects AG129 mice against challenge with the closely related Heartland bandavirus despite negligible neutralizing titers to the virus. CONCLUSIONS Taken together, these data suggest that rVSV-SFTSV is a promising vaccine candidate for SFTSV and Heartland bandavirus with a favorable safety profile.
Collapse
Affiliation(s)
- Philip Hicks
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.H.); (T.B.M.); (R.J.P.); (B.R.)
| | - Tomaz B. Manzoni
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.H.); (T.B.M.); (R.J.P.); (B.R.)
| | - Jonna B. Westover
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (J.B.W.); (B.B.G.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Raegan J. Petch
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.H.); (T.B.M.); (R.J.P.); (B.R.)
| | - Brianne Roper
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.H.); (T.B.M.); (R.J.P.); (B.R.)
| | - Brian B. Gowen
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (J.B.W.); (B.B.G.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.H.); (T.B.M.); (R.J.P.); (B.R.)
| |
Collapse
|
11
|
Zinovieva M, Ryapolova A, Karabelsky A, Minskaia E. Oncolytic Vesicular Stomatitis Virus: Optimisation Strategies for Anti-Cancer Therapies. FRONT BIOSCI-LANDMRK 2024; 29:374. [PMID: 39614430 DOI: 10.31083/j.fbl2911374] [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: 06/01/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 12/01/2024]
Abstract
Oncolytic viruses (OVs) represent a targeted anti-cancer therapy approach due to their ability not only to selectively infect and destroy malignant cells but also to induce an immune response. Vesicular stomatitis virus (VSV) offers a promising platform due to its low prevalence and pathogenicity in humans, lack of pre-existing immunity, easily manipulated genome, rapid growth to high titers in a broad range of cell lines, and inability to integrate into the host genome. However, despite its many advantages, many unresolved problems remain: problematic production based on the reverse genetics system, oncological selectivity, and the overall effectiveness of VSV monotherapy. This review will discuss various attempts at viral genome modifications aimed at improving the oncolytic properties of VSV. These strategies include inhibition of viral genes, modification of genes responsible for targeting cancer cells over healthy ones, insertion of foreign genes for boosting immune response, and changing the order of viral and inserted foreign genes. In addition, possible ways to improve VSV-based anti-tumor therapy and achieve higher efficiency will be considered by evaluating the effectiveness of various delivery methods as well as discussing treatment options by combining VSV with other groups of anticancer drugs.
Collapse
Affiliation(s)
- Margarita Zinovieva
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasia Ryapolova
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Alexander Karabelsky
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ekaterina Minskaia
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
12
|
Metko M, Tonne J, Veliz Rios A, Thompson J, Mudrick H, Masopust D, Diaz RM, Barry MA, Vile RG. Intranasal Prime-Boost with Spike Vectors Generates Antibody and T-Cell Responses at the Site of SARS-CoV-2 Infection. Vaccines (Basel) 2024; 12:1191. [PMID: 39460356 PMCID: PMC11511174 DOI: 10.3390/vaccines12101191] [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: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime-boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon viral infection. Although most vaccines are currently delivered intramuscularly (IM), the initial site of infection is the nasal cavity. METHODS Here, we tested the hypothesis that a heterologous prime and boost vaccine regimen delivered intranasally (IN) will generate improved immune responses locally at the site of virus infection compared to intramuscular vaccine/booster regimens. RESULTS In a transgenic human ACE2 murine model, both a Spike-expressing single-cycle adenovirus (SC-Ad) and an IFNß safety-enhanced replication-competent Vesicular Stomatitis Virus (VSV) platform generated anti-Spike antibody and T-cell responses that diminished with age. Although SC-Ad-Spike boosted a prime with VSV-Spike-mIFNß, SC-Ad-Spike alone induced maximal levels of IgG, IgA, and CD8+ T-cell responses. CONCLUSIONS There were significant differences in T-cell responses in spleens compared to lungs, and the intranasal boost was significantly superior to the intramuscular boost in generating sentinel immune effectors at the site of the virus encounter in the lungs. These data show that serious consideration should be given to intranasal boosting with anti-SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Muriel Metko
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Alexa Veliz Rios
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Haley Mudrick
- Molecular Pharmacology and Experimental Therapeutics Program, Mayo Clinic, Rochester, MN 55905, USA;
| | - David Masopust
- Department of Microbiology & Immunology, University of Minnesota Medical School, 2101 6th St. SE, Minneapolis, MN 55455, USA;
| | - Rosa Maria Diaz
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Michael A. Barry
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
- Department of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard G. Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
13
|
Khadka RB, Karki K, Pandey J, Gyawali R, Chaudhary GP. Strengthening global health resilience: Marburg virus-like particle vaccines and the One Health approach. SCIENCE IN ONE HEALTH 2024; 3:100076. [PMID: 39309209 PMCID: PMC11415973 DOI: 10.1016/j.soh.2024.100076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024]
Abstract
The Marburg virus (MARV), belonging to the Filoviridae family, poses a significant global health threat, emphasizing the urgency to develop Marburg virus-like particle (VLP) vaccines for outbreak mitigation. The virus's menacing traits accentuate the need for such vaccines, which can be addressed by VLPs that mimic its structure safely, potentially overcoming past limitations. Early Marburg vaccine endeavors and their challenges are examined in the historical perspectives section, followed by an exploration of VLPs as transformative tools, capable of eliciting immune responses without conventional risks. Noteworthy milestones and achievements in Marburg VLP vaccine development, seen through preclinical and clinical trials, indicate potential cross-protection. Ongoing challenges, encompassing durability, strain diversity, and equitable distribution, are addressed, with proposed innovations like novel adjuvant, mRNA technology, and structure-based design poised to enhance Marburg VLP vaccines. This review highlights the transformative potential of Marburg VLPs in countering the virus, showcasing global collaboration, regulatory roles, and health equity for a safer future through the harmonious interplay of science, regulation, and global efforts.
Collapse
Affiliation(s)
- Ram Bahadur Khadka
- Department of Laboratory Science, Crimson College of Technology, Affiliated with Pokhara University, Butwal-11, Devinagar, Rupandehi 32907, Nepal
| | - Khimdhoj Karki
- Department of Laboratory Science, Crimson College of Technology, Affiliated with Pokhara University, Butwal-11, Devinagar, Rupandehi 32907, Nepal
| | - Jitendra Pandey
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Rabin Gyawali
- Padmodaya Campus, Affiliated to Tribhuwan University, Dang 21906, Nepal
| | - Gautam Prasad Chaudhary
- Department of Pharmacy, Crimson College of Technology, Affiliated with Pokhara University, Butwal-11, Devinagar, Rupandehi 32907, Nepal
| |
Collapse
|
14
|
Yu C, Lin Y, Dai Y, Wu B, Qi Z, Qian X. Recent research advances in the development of Dabie Banda virus vaccines. PLoS Negl Trop Dis 2024; 18:e0012411. [PMID: 39207951 PMCID: PMC11361446 DOI: 10.1371/journal.pntd.0012411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a newly identified tick-borne viral hemorrhagic fever caused by Dabie Banda virus (DBV). The virus was first discovered in eastern China in 2009 and is now considered an infectious disease with a mortality rate ranging from 6.3% to 30%. The best strategy for controlling SFTS is to develop effective vaccines. However, no approved vaccines are currently available to prevent this disease, despite the number of extensive and in-depth studies conducted on DBV in the past few years. This review focuses on the structure of DBV and the induced host immune responses which are the fundamental factors in vaccine development, and thoroughly summarizes the current research progress on DBV vaccines. The developing DBV vaccines include protein subunit vaccines, live attenuated vaccines, recombinant virus vector vaccines, and DNA vaccines. At present, almost all candidate vaccines for DBV are in the laboratory development or preclinical stages. There remain challenges in successfully developing clinically approved DBV vaccines.
Collapse
Affiliation(s)
- Chenyang Yu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Yuxiang Lin
- College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Yixin Dai
- Nursing Department, Faculty of Health and Wellness, Linxia Modern Career College, Gansu, China
| | - Bingan Wu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xijing Qian
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
15
|
Goetz M, Thotathil N, Zhao Z, Mitragotri S. Vaccine adjuvants for infectious disease in the clinic. Bioeng Transl Med 2024; 9:e10663. [PMID: 39036089 PMCID: PMC11256182 DOI: 10.1002/btm2.10663] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Adjuvants, materials added to vaccines to enhance the resulting immune response, are important components of vaccination that are many times overlooked. While vaccines always include an antigen to tell the body what to vaccinate to, of equal importance the adjuvant provides the how, a significant factor in producing a complete response. The adjuvant space has been slow to develop with the first use of an adjuvant in a licensed vaccine occurring in the 1930s, and remaining the only adjuvant in licensed vaccines for the next 80 years. However, with vaccination at the forefront of protection against new and complex pathogens, it is important to consider all components when designing an effective vaccine. Here we summarize the adjuvant space in licensed vaccines as well as the novel adjuvant space in clinical trials with a specific focus on the materials utilized and their resulting impact on the immune response. We discuss five major categories of adjuvant materials: aluminum salts, nanoparticles, viral vectors, TLR agonists, and emulsions. For each category, we delve into the current clinical trials space, the impact of these materials on vaccination, as well as some of the ways in which they could be improved. Adjuvants present an exciting opportunity to improve vaccine responses and stability, this review will help inform about the current progress of this space. Translational impact statement In the aftermath of the COVID-19 pandemic, vaccines for infectious diseases have come into the spotlight. While antigens have always been an important focus of vaccine design, the adjuvant is a significant tool for enhancing the immune response to the vaccine that has been largely underdeveloped. This article provides a broad review of the history of adjuvants and, the current vaccine adjuvant space, and the progress seen in adjuvants in clinical trials. There is specific emphasis on the material landscape for adjuvants and their resulting mechanism of action. Looking ahead, while the novel vaccine adjuvant space features exciting new technologies and materials, there is still a need for more to meet the protective needs of new and complex pathogens.
Collapse
Affiliation(s)
- Morgan Goetz
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Naaz Thotathil
- University of Massachusetts AmherstAmherstMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
16
|
Rasmussen AL, Gronvall G, Lowen AC, Goodrum F. Reply to Ebright et al., "Implementing governmental oversight of enhanced potential pandemic pathogen research". J Virol 2024; 98:e0025624. [PMID: 38477587 PMCID: PMC11019956 DOI: 10.1128/jvi.00256-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024] Open
Affiliation(s)
- Angela L. Rasmussen
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Gigi Gronvall
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anice C. Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Felicia Goodrum
- Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
17
|
Todesco HM, Gafuik C, John CM, Roberts EL, Borys BS, Pawluk A, Kallos MS, Potts KG, Mahoney DJ. High-titer manufacturing of SARS-CoV-2 Spike-pseudotyped VSV in stirred-tank bioreactors. Mol Ther Methods Clin Dev 2024; 32:101189. [PMID: 38327804 PMCID: PMC10847022 DOI: 10.1016/j.omtm.2024.101189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic highlighted the importance of vaccine innovation in public health. Hundreds of vaccines built on numerous technology platforms have been rapidly developed against SARS-CoV-2 since 2020. Like all vaccine platforms, an important bottleneck to viral-vectored vaccine development is manufacturing. Here, we describe a scalable manufacturing protocol for replication-competent SARS-CoV-2 Spike-pseudotyped vesicular stomatitis virus (S-VSV)-vectored vaccines using Vero cells grown on microcarriers in a stirred-tank bioreactor. Using Cytodex 1 microcarriers over 6 days of fed-batch culture, Vero cells grew to a density of 3.95 ± 0.42 ×106 cells/mL in 1-L stirred-tank bioreactors. Ancestral strain S-VSV reached a peak titer of 2.05 ± 0.58 ×108 plaque-forming units (PFUs)/mL at 3 days postinfection. When compared to growth in plate-based cultures, this was a 29-fold increase in virus production, meaning a 1-L bioreactor produces the same amount of virus as 1,284 plates of 15 cm. In addition, the omicron BA.1 S-VSV reached a peak titer of 5.58 ± 0.35 × 106 PFU/mL. Quality control testing showed plate- and bioreactor-produced S-VSV had similar particle-to-PFU ratios and elicited comparable levels of neutralizing antibodies in immunized hamsters. This method should enhance preclinical and clinical development of pseudotyped VSV-vectored vaccines in future pandemics.
Collapse
Affiliation(s)
- Hayley M. Todesco
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Chris Gafuik
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Cini M. John
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Erin L. Roberts
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Breanna S. Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Alexis Pawluk
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Michael S. Kallos
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Kyle G. Potts
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Douglas J. Mahoney
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
18
|
Göbel S, Pelz L, Silva CAT, Brühlmann B, Hill C, Altomonte J, Kamen A, Reichl U, Genzel Y. Production of recombinant vesicular stomatitis virus-based vectors by tangential flow depth filtration. Appl Microbiol Biotechnol 2024; 108:240. [PMID: 38413399 PMCID: PMC10899354 DOI: 10.1007/s00253-024-13078-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
Cell culture-based production of vector-based vaccines and virotherapeutics is of increasing interest. The vectors used not only retain their ability to infect cells but also induce robust immune responses. Using two recombinant vesicular stomatitis virus (rVSV)-based constructs, we performed a proof-of-concept study regarding an integrated closed single-use perfusion system that allows continuous virus harvesting and clarification. Using suspension BHK-21 cells and a fusogenic oncolytic hybrid of vesicular stomatitis virus and Newcastle disease virus (rVSV-NDV), a modified alternating tangential flow device (mATF) or tangential flow depth filtration (TFDF) systems were used for cell retention. As the hollow fibers of the former are characterized by a large internal lumen (0.75 mm; pore size 0.65 μm), membrane blocking by the multi-nucleated syncytia formed during infection could be prevented. However, virus particles were completely retained. In contrast, the TFDF filter unit (lumen 3.15 mm, pore size 2-5 μm) allowed not only to achieve high viable cell concentrations (VCC, 16.4-20.6×106 cells/mL) but also continuous vector harvesting and clarification. Compared to an optimized batch process, 11-fold higher infectious virus titers were obtained in the clarified permeate (maximum 7.5×109 TCID50/mL). Using HEK293-SF cells and a rVSV vector expressing a green fluorescent protein, perfusion cultivations resulted in a maximum VCC of 11.3×106 cells/mL and infectious virus titers up to 7.1×1010 TCID50/mL in the permeate. Not only continuous harvesting but also clarification was possible. Although the cell-specific virus yield decreased relative to a batch process established as a control, an increased space-time yield was obtained. KEY POINTS: • Viral vector production using a TFDF perfusion system resulted in a 460% increase in space-time yield • Use of a TFDF system allowed continuous virus harvesting and clarification • TFDF perfusion system has great potential towards the establishment of an intensified vector production.
Collapse
Affiliation(s)
- Sven Göbel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Lars Pelz
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Cristina A T Silva
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Québec, Canada
| | | | | | - Jennifer Altomonte
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montréal, Québec, Canada
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
- Chair for Bioprocess Engineering, Otto von Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany.
| |
Collapse
|
19
|
Cohen N, Simon I, Hazan O, Tal A, Tzadok H, Levin L, Girshengorn M, Mimran LC, Natan N, Baruhi T, David AB, Rosen O, Shmaya S, Borni S, Cohen N, Lupu E, Kedmi A, Zilberman O, Jayson A, Monash A, Dor E, Diamant E, Goldvaser M, Cohen-Gihon I, Israeli O, Lazar S, Shifman O, Beth-Din A, Zvi A, Oren Z, Makovitzki A, Lerer E, Mimran A, Toister E, Zichel R, Adar Y, Epstein E. Enhanced production yields of rVSV-SARS-CoV-2 vaccine using Fibra-Cel ® macrocarriers. Front Bioeng Biotechnol 2024; 12:1333548. [PMID: 38449674 PMCID: PMC10915211 DOI: 10.3389/fbioe.2024.1333548] [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: 11/05/2023] [Accepted: 02/02/2024] [Indexed: 03/08/2024] Open
Abstract
The COVID-19 pandemic has led to high global demand for vaccines to safeguard public health. To that end, our institute has developed a recombinant viral vector vaccine utilizing a modified vesicular stomatitis virus (VSV) construct, wherein the G protein of VSV is replaced with the spike protein of SARS-CoV-2 (rVSV-ΔG-spike). Previous studies have demonstrated the production of a VSV-based vaccine in Vero cells adsorbed on Cytodex 1 microcarriers or in suspension. However, the titers were limited by both the carrier surface area and shear forces. Here, we describe the development of a bioprocess for rVSV-ΔG-spike production in serum-free Vero cells using porous Fibra-Cel® macrocarriers in fixed-bed BioBLU®320 5p bioreactors, leading to high-end titers. We identified core factors that significantly improved virus production, such as the kinetics of virus production, the use of macrospargers for oxygen supply, and medium replenishment. Implementing these parameters, among others, in a series of GMP production processes improved the titer yields by at least two orders of magnitude (2e9 PFU/mL) over previously reported values. The developed process was highly effective, repeatable, and robust, creating potent and genetically stable vaccine viruses and introducing new opportunities for application in other viral vaccine platforms.
Collapse
Affiliation(s)
- Noam Cohen
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Irit Simon
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ophir Hazan
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Arnon Tal
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Hanan Tzadok
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Lilach Levin
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Meni Girshengorn
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Lilach Cherry Mimran
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Niva Natan
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Tzadok Baruhi
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Alon Ben David
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Osnat Rosen
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shlomo Shmaya
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Sarah Borni
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Noa Cohen
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Edith Lupu
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Adi Kedmi
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Orian Zilberman
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Avital Jayson
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Arik Monash
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Eyal Dor
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Eran Diamant
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Michael Goldvaser
- Department of Organic Chemistry, Israel Institute for Biological, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Inbar Cohen-Gihon
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ofir Israeli
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shirley Lazar
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ohad Shifman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Adi Beth-Din
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Anat Zvi
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ziv Oren
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Arik Makovitzki
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Elad Lerer
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Avishai Mimran
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Einat Toister
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ran Zichel
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Yaakov Adar
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Eyal Epstein
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| |
Collapse
|
20
|
Xu D, Powell AE, Utz A, Sanyal M, Do J, Patten JJ, Moliva JI, Sullivan NJ, Davey RA, Kim PS. Design of universal Ebola virus vaccine candidates via immunofocusing. Proc Natl Acad Sci U S A 2024; 121:e2316960121. [PMID: 38319964 PMCID: PMC10873634 DOI: 10.1073/pnas.2316960121] [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: 09/29/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024] Open
Abstract
The Ebola virus causes hemorrhagic fever in humans and poses a significant threat to global public health. Although two viral vector vaccines have been approved to prevent Ebola virus disease, they are distributed in the limited ring vaccination setting and only indicated for prevention of infection from orthoebolavirus zairense (EBOV)-one of three orthoebolavirus species that have caused previous outbreaks. Ebola virus glycoprotein GP mediates viral infection and serves as the primary target of neutralizing antibodies. Here, we describe a universal Ebola virus vaccine approach using a structure-guided design of candidates with hyperglycosylation that aims to direct antibody responses away from variable regions and toward conserved epitopes of GP. We first determined the hyperglycosylation landscape on Ebola virus GP and used that to generate hyperglycosylated GP variants with two to four additional glycosylation sites to mask the highly variable glycan cap region. We then created vaccine candidates by displaying wild-type or hyperglycosylated GP variants on ferritin nanoparticles (Fer). Immunization with these antigens elicited potent neutralizing antisera against EBOV in mice. Importantly, we observed consistent cross-neutralizing activity against Bundibugyo virus and Sudan virus from hyperglycosylated GP-Fer with two or three additional glycans. In comparison, elicitation of cross-neutralizing antisera was rare in mice immunized with wild-type GP-Fer. These results demonstrate a potential strategy to develop universal Ebola virus vaccines that confer cross-protective immunity against existing and emerging filovirus species.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Abigail E. Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA94305
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA94305
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Jonathan Do
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - J. J. Patten
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Juan I. Moliva
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Nancy J. Sullivan
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
- Department of Biology, Boston University, Boston, MA02118
| | - Robert A. Davey
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Chan Zuckerberg Biohub, San Francisco, CA94158
| |
Collapse
|
21
|
Gholap AD, Gupta J, Kamandar P, Bhowmik DD, Rojekar S, Faiyazuddin M, Hatvate NT, Mohanto S, Ahmed MG, Subramaniyan V, Kumarasamy V. Harnessing Nanovaccines for Effective Immunization─A Special Concern on COVID-19: Facts, Fidelity, and Future Prospective. ACS Biomater Sci Eng 2024; 10:271-297. [PMID: 38096426 DOI: 10.1021/acsbiomaterials.3c01247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Nanotechnology has emerged as a transformative pathway in vaccine research and delivery. Nanovaccines, encompassing lipid and nonlipid formulations, exhibit considerable advantages over traditional vaccine techniques, including enhanced antigen stability, heightened immunogenicity, targeted distribution, and the potential for codelivery with adjuvants or immune modulators. This review provides a comprehensive overview of the latest advancements and applications of lipid and non-lipid-based nanovaccines in current vaccination strategies for immunization. The review commences by outlining the fundamental concepts underlying lipid and nonlipid nanovaccine design before delving into the diverse components and production processes employed in their development. Subsequently, a comparative analysis of various nanocarriers is presented, elucidating their distinct physicochemical characteristics and impact on the immune response, along with preclinical and clinical studies. The discussion also highlights how nanotechnology enables the possibility of personalized and combined vaccination techniques, facilitating the creation of tailored nanovaccines to meet the individual patient needs. The ethical aspects concerning the use of nanovaccines, as well as potential safety concerns and public perception, are also addressed. The study underscores the gaps and challenges that must be overcome before adopting nanovaccines in clinical practice. This comprehensive analysis offers vital new insights into lipid and nonlipid nanovaccine status. It emphasizes the significance of continuous research, collaboration among interdisciplinary experts, and regulatory measures to fully unlock the potential of nanotechnology in enhancing immunization and ensuring a healthier, more resilient society.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Juhi Gupta
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Pallavi Kamandar
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Deblina D Bhowmik
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Md Faiyazuddin
- Department of Pharmaceutics, School of Pharmacy, Al-Karim University, Katihar 854106, Bihar, India
| | - Navnath T Hatvate
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
22
|
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
- College of Veterinary Medicine Northwest A&F University, Yangling, Shanxi, 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
|
23
|
Wu J, Cai Y, Jiang N, Qian Y, Lyu R, You Q, Zhang F, Tao H, Zhu H, Nawaz W, Chen D, Wu Z. Pralatrexate inhibited the replication of varicella zoster virus and vesicular stomatitis virus: An old dog with new tricks. Antiviral Res 2024; 221:105787. [PMID: 38145756 DOI: 10.1016/j.antiviral.2023.105787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
Varicella zoster virus (VZV) is associated with herpes zoster (HZ) or herpes zoster ophthalmicus (HZO). All antiviral agents currently licensed for the management of VZV replication via modulating different mechanisms, and the resistance is on the rise. There is a need to develop new antiviral agents with distinct mechanisms of action and adequate safety profiles. Pralatrexate (PDX) is a fourth-generation anti-folate agent with an inhibitory activity on folate (FA) metabolism and has been used as an anti-tumor drug. We observed that PDX possessed potent inhibitory activity against VZV infection. In this study, we reported the antiviral effects and the underlying mechanism of PDX against VZV infection. The results showed that PDX not only inhibited VZV replication in vitro and in mice corneal tissues but also reduced the inflammatory response and apoptosis induced by viral infection. Furthermore, PDX treatment showed a similar anti-VSV inhibitory effect in both in vitro and in vivo models. Mechanistically, PDX inhibited viral replication by interrupting the substrate supply for de novo purine and thymidine synthesis. In conclusion, this study discovered the potent antiviral activity of PDX with a novel mechanism and presented a new strategy for VZV treatment that targets a cellular metabolic mechanism essential for viral replication. The present study provided a new insight into the development of broad-spectrum antiviral agents.
Collapse
Affiliation(s)
- Jing Wu
- Medical School of Nanjing University, Nanjing, China
| | - Yurong Cai
- School of Life Science, Ningxia University, Yinchuan, China
| | - Na Jiang
- Medical School of Nanjing University, Nanjing, China
| | - Yajie Qian
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ruining Lyu
- Medical School of Nanjing University, Nanjing, China
| | - Qiao You
- Medical School of Nanjing University, Nanjing, China
| | - Fang Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hongji Tao
- Medical School of Nanjing University, Nanjing, China
| | - Haotian Zhu
- Medical School of Nanjing University, Nanjing, China
| | - Waqas Nawaz
- Hȏpital Maisonneuve-Rosemont, School of Medicine, University of Montreal, Canada
| | - Deyan Chen
- Medical School of Nanjing University, Nanjing, China.
| | - Zhiwei Wu
- Medical School of Nanjing University, Nanjing, China; Northern Jiangsu People's Hospital, Affiliated Teaching Hospital of Medical School, Nanjing University, Yangzhou, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China; School of Life Science, Ningxia University, Yinchuan, China.
| |
Collapse
|
24
|
Xu D, Powell AE, Utz A, Sanyal M, Do J, Patten J, Moliva JI, Sullivan NJ, Davey RA, Kim PS. Design of universal Ebola virus vaccine candidates via immunofocusing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.14.562364. [PMID: 37904982 PMCID: PMC10614775 DOI: 10.1101/2023.10.14.562364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Ebola virus causes hemorrhagic fever in humans and poses a significant threat to global public health. Although two viral vector vaccines have been approved to prevent Ebola virus disease, they are distributed in the limited ring vaccination setting and only indicated for prevention of infection from orthoebolavirus zairense (EBOV) - one of three orthoebolavirus species that have caused previous outbreaks. Ebola virus glycoprotein GP mediates viral infection and serves as the primary target of neutralizing antibodies. Here we describe a universal Ebola virus vaccine approach using structure-guided design of candidates with hyperglycosylation that aims to direct antibody responses away from variable regions and toward conserved epitopes of GP. We first determined the hyperglycosylation landscape on Ebola virus GP and used that to generate hyperglycosylated GP variants with two to four additional glycosylation sites to mask the highly variable glycan cap region. We then created vaccine candidates by displaying wild-type or hyperglycosylated GP variants on ferritin nanoparticles (Fer). Immunization with these antigens elicited potent neutralizing antisera against EBOV in mice. Importantly, we observed consistent cross-neutralizing activity against Bundibugyo virus and Sudan virus from hyperglycosylated GP-Fer with two or three additional glycans. In comparison, elicitation of cross-neutralizing antisera was rare in mice immunized with wild-type GP-Fer. These results demonstrate a potential strategy to develop universal Ebola virus vaccines that confer cross-protective immunity against existing and emerging filovirus species.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Abigail E. Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Jonathan Do
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - J.J. Patten
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Juan I. Moliva
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nancy J. Sullivan
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biology, Boston University, Boston, MA 02118, USA
| | - Robert A. Davey
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
25
|
Aguilar-Briseño JA, Elliff JM, Patten JJ, Wilson LR, Davey RA, Bailey AL, Maury WJ. Effect of Interferon Gamma on Ebola Virus Infection of Primary Kupffer Cells and a Kupffer Cell Line. Viruses 2023; 15:2077. [PMID: 37896854 PMCID: PMC10611415 DOI: 10.3390/v15102077] [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: 08/27/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Ebola virus disease (EVD) represents a global health threat. The etiological agents of EVD are six species of Orthoebolaviruses, with Orthoebolavirus zairense (EBOV) having the greatest public health and medical significance. EVD pathogenesis occurs as a result of broad cellular tropism of the virus, robust viral replication and a potent and dysregulated production of cytokines. In vivo, tissue macrophages are some of the earliest cells infected and contribute significantly to virus load and cytokine production. While EBOV is known to infect macrophages and to generate high titer virus in the liver, EBOV infection of liver macrophages, Kupffer cells, has not previously been examined in tissue culture or experimentally manipulated in vivo. Here, we employed primary murine Kupffer cells (KC) and an immortalized murine Kupffer cell line (ImKC) to assess EBOV-eGFP replication in liver macrophages. KCs and ImKCs were highly permissive for EBOV infection and IFN-γ polarization of these cells suppressed their permissiveness to infection. The kinetics of IFN-γ-elicited antiviral responses were examined using a biologically contained model of EBOV infection termed EBOV ΔVP30. The antiviral activity of IFN-γ was transient, but a modest ~3-fold reduction of infection persisted for as long as 6 days post-treatment. To assess the interferon-stimulated gene products (ISGs) responsible for protection, the efficacy of secreted ISGs induced by IFN-γ was evaluated and secreted ISGs failed to block EBOV ΔVP30. Our studies define new cellular tools for the study of EBOV infection that can potentially aid the development of new antiviral therapies. Furthermore, our data underscore the importance of macrophages in EVD pathogenesis and those IFN-γ-elicited ISGs that help to control EBOV infection.
Collapse
Affiliation(s)
| | - Jonah M. Elliff
- Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Justin J. Patten
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA; (J.J.P.); (R.A.D.)
| | - Lindsay R. Wilson
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA; (L.R.W.); (A.L.B.)
| | - Robert A. Davey
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA; (J.J.P.); (R.A.D.)
| | - Adam L. Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA; (L.R.W.); (A.L.B.)
| | - Wendy J. Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA;
- Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
| |
Collapse
|
26
|
Sène MA, Xia Y, Kamen AA. Comparative Transcriptomic Analyses of a Vero Cell Line in Suspension versus Adherent Culture Conditions. Int J Cell Biol 2023; 2023:9364689. [PMID: 37680537 PMCID: PMC10482560 DOI: 10.1155/2023/9364689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 07/13/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023] Open
Abstract
The Vero cell line is the most used continuous cell line for viral vaccine manufacturing. Its anchorage-dependent use renders scaling up challenging and operations very labor-intensive which affects cost effectiveness. Thus, efforts to adapt Vero cells to suspension cultures have been invested, but hurdles such as the long doubling time and low cell viability remain to be addressed. In this study, building on the recently published Vero cell line annotated genome, a functional genomics analysis of the Vero cells adapted to suspension is performed to better understand the genetic and phenotypic switches at play during the adaptation of Vero cells from anchorage-dependent to suspension cultures. Results show downregulation of the epithelial-to-mesenchymal transition (EMT) pathway, highlighting the dissociation between the adaptation to suspension process and EMT. Surprisingly, an upregulation of cell adhesion components is observed, notably the CDH18 gene, the cytoskeleton pathway, and the extracellular pathway. Moreover, a downregulation of the glycolytic pathway is balanced by an upregulation of the asparagine metabolism pathway, promoting cell adaptation to nutrient deprivation. A downregulation of the adherens junctions and the folate pathways alongside with the FYN gene are possible explanations behind the currently observed low-cell viability and long doubling time.
Collapse
Affiliation(s)
| | - Yu Xia
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Amine A. Kamen
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Ouyang MJ, Ao Z, Olukitibi TA, Lawrynuik P, Shieh C, Kung SKP, Fowke KR, Kobasa D, Yao X. Oral Immunization with rVSV Bivalent Vaccine Elicits Protective Immune Responses, Including ADCC, against Both SARS-CoV-2 and Influenza A Viruses. Vaccines (Basel) 2023; 11:1404. [PMID: 37766083 PMCID: PMC10534613 DOI: 10.3390/vaccines11091404] [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: 07/26/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
COVID-19 and influenza both cause enormous disease burdens, and vaccines are the primary measures for their control. Since these viral diseases are transmitted through the mucosal surface of the respiratory tract, developing an effective and convenient mucosal vaccine should be a high priority. We previously reported a recombinant vesicular stomatitis virus (rVSV)-based bivalent vaccine (v-EM2/SPΔC1Delta) that protects animals from both SARS-CoV-2 and influenza viruses via intramuscular and intranasal immunization. Here, we further investigated the immune response induced by oral immunization with this vaccine and its protective efficacy in mice. The results demonstrated that the oral delivery, like the intranasal route, elicited strong and protective systemic immune responses against SARS-CoV-2 and influenza A virus. This included high levels of neutralizing antibodies (NAbs) against SARS-CoV-2, as well as strong anti-SARS-CoV-2 spike protein (SP) antibody-dependent cellular cytotoxicity (ADCC) and anti-influenza M2 ADCC responses in mice sera. Furthermore, it provided efficient protection against challenge with influenza H1N1 virus in a mouse model, with a 100% survival rate and a significantly low lung viral load of influenza virus. All these findings provide substantial evidence for the effectiveness of oral immunization with the rVSV bivalent vaccine.
Collapse
Affiliation(s)
- Maggie Jing Ouyang
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Titus A. Olukitibi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Peter Lawrynuik
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Christopher Shieh
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Sam K. P. Kung
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W3, Canada;
| | - Keith R. Fowke
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Darwyn Kobasa
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| |
Collapse
|
28
|
Hotez PJ, Gilbert S, Saville M, Privor-Dumm L, Abdool-Karim S, Thompson D, Excler JL, Kim JH. COVID-19 vaccines and the pandemic: lessons learnt for other neglected diseases and future threats. BMJ Glob Health 2023; 8:bmjgh-2023-011883. [PMID: 37277196 DOI: 10.1136/bmjgh-2023-011883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/09/2023] [Indexed: 06/07/2023] Open
Abstract
Through the experiences gained by accelerating new vaccines for both Ebola virus infection and COVID-19 in a public health emergency, vaccine development has benefited from a 'multiple shots on goal' approach to new vaccine targets. This approach embraces simultaneous development of candidates with differing technologies, including, when feasible, vesicular stomatitis virus or adenovirus vectors, messenger RNA (mRNA), whole inactivated virus, nanoparticle and recombinant protein technologies, which led to multiple effective COVID-19 vaccines. The challenge of COVID-19 vaccine inequity, as COVID-19 spread globally, created a situation where cutting-edge mRNA technologies were preferentially supplied by multinational pharmaceutical companies to high-income countries while low and middle-income countries (LMICs) were pushed to the back of the queue and relied more heavily on adenoviral vector, inactivated virus and recombinant protein vaccines. To prevent this from occurring in future pandemics, it is essential to expand the scale-up capacity for both traditional and new vaccine technologies at individual or simultaneous hubs in LMICs. In parallel, a process of tech transfer of new technologies to LMIC producers needs to be facilitated and funded, while building LMIC national regulatory capacity, with the aim of several reaching 'stringent regulator' status. Access to doses is an essential start but is not sufficient, as healthcare infrastructure for vaccination and combating dangerous antivaccine programmes both require support. Finally, there is urgency to establish an international framework through a United Nations Pandemic Treaty to promote, support and harmonise a more robust, coordinated and effective global response.
Collapse
Affiliation(s)
- Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah Gilbert
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| | | | - Lois Privor-Dumm
- International Vaccine Access Center, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Salim Abdool-Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA) and Department of Epidemiology, Columbia University, New York, New York, USA
| | | | | | - Jerome H Kim
- International Vaccine Institute, Seoul, South Korea
| |
Collapse
|
29
|
Chen T, Ding Z, Lan J, Wong G. Advances and perspectives in the development of vaccines against highly pathogenic bunyaviruses. Front Cell Infect Microbiol 2023; 13:1174030. [PMID: 37274315 PMCID: PMC10234439 DOI: 10.3389/fcimb.2023.1174030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Increased human activities around the globe and the rapid development of once rural regions have increased the probability of contact between humans and wild animals. A majority of bunyaviruses are of zoonotic origin, and outbreaks may result in the substantial loss of lives, economy contraction, and social instability. Many bunyaviruses require manipulation in the highest levels of biocontainment, such as Biosafety Level 4 (BSL-4) laboratories, and the scarcity of this resource has limited the development speed of vaccines for these pathogens. Meanwhile, new technologies have been created, and used to innovate vaccines, like the mRNA vaccine platform and bioinformatics-based antigen design. Here, we summarize current vaccine developments for three different bunyaviruses requiring work in the highest levels of biocontainment: Crimean-Congo Hemorrhagic Fever Virus (CCHFV), Rift Valley Fever Virus (RVFV), and Hantaan virus (HTNV), and provide perspectives and potential future directions that can be further explored to advance specific vaccines for humans and livestock.
Collapse
Affiliation(s)
- Tong Chen
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhe Ding
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaming Lan
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Gary Wong
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
| |
Collapse
|
30
|
LaBauve AE, Saada EA, Jones IKA, Mosesso R, Noureddine A, Techel J, Gomez A, Collette N, Sherman MB, Serda RE, Butler KS, Brinker CJ, Schoeniger JS, Sasaki D, Negrete OA. Lipid-coated mesoporous silica nanoparticles for anti-viral applications via delivery of CRISPR-Cas9 ribonucleoproteins. Sci Rep 2023; 13:6873. [PMID: 37105997 PMCID: PMC10133914 DOI: 10.1038/s41598-023-33092-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Emerging and re-emerging viral pathogens present a unique challenge for anti-viral therapeutic development. Anti-viral approaches with high flexibility and rapid production times are essential for combating these high-pandemic risk viruses. CRISPR-Cas technologies have been extensively repurposed to treat a variety of diseases, with recent work expanding into potential applications against viral infections. However, delivery still presents a major challenge for these technologies. Lipid-coated mesoporous silica nanoparticles (LCMSNs) offer an attractive delivery vehicle for a variety of cargos due to their high biocompatibility, tractable synthesis, and amenability to chemical functionalization. Here, we report the use of LCMSNs to deliver CRISPR-Cas9 ribonucleoproteins (RNPs) that target the Niemann-Pick disease type C1 gene, an essential host factor required for entry of the high-pandemic risk pathogen Ebola virus, demonstrating an efficient reduction in viral infection. We further highlight successful in vivo delivery of the RNP-LCMSN platform to the mouse liver via systemic administration.
Collapse
Affiliation(s)
- Annette E LaBauve
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, USA
| | - Edwin A Saada
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA
- Biotechnology and Biosciences Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, USA
| | - Iris K A Jones
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA
| | - Richard Mosesso
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA
| | - Achraf Noureddine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, USA
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, USA
| | - Jessica Techel
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, USA
| | - Andrew Gomez
- Department of Active Ceramics Value Stream, Sandia National Laboratories, Albuquerque, USA
| | - Nicole Collette
- Biotechnology and Biosciences Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, USA
| | - Michael B Sherman
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, USA
| | - Rita E Serda
- Department of Internal Medicine, Health Sciences Center, University of New Mexico, Albuquerque, USA
| | - Kimberly S Butler
- Department of Molecular and Microbiology, Sandia National Laboratories, Albuquerque, USA
| | - C Jeffery Brinker
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, USA
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, USA
- Advanced Materials Laboratory, Sandia National Laboratories, Albuquerque, USA
| | | | - Darryl Sasaki
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, USA
| | - Oscar A Negrete
- Department of Systems Biology, Sandia National Laboratories, Livermore, USA.
| |
Collapse
|
31
|
Yang Z, Paes BCMF, Fulber JPC, Tran MY, Farnós O, Kamen AA. Development of an Integrated Continuous Manufacturing Process for the rVSV-Vectored SARS-CoV-2 Candidate Vaccine. Vaccines (Basel) 2023; 11:vaccines11040841. [PMID: 37112753 PMCID: PMC10143285 DOI: 10.3390/vaccines11040841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The administration of viral vectored vaccines remains one of the most effective ways to respond to the ongoing novel coronavirus disease 2019 (COVID-19) pandemic. However, pre-existing immunity to the viral vector hinders its potency, resulting in a limited choice of viral vectors. Moreover, the basic batch mode of manufacturing vectored vaccines does not allow one to cost-effectively meet the global demand for billions of doses per year. To date, the exposure of humans to VSV infection has been limited. Therefore, a recombinant vesicular stomatitis virus (rVSV), which expresses the spike protein of SARS-CoV-2, was selected as the vector. To determine the operating upstream process conditions for the most effective production of an rVSV-SARS-CoV-2 candidate vaccine, a set of critical process parameters was evaluated in an Ambr 250 modular system, whereas in the downstream process, a streamlined process that included DNase treatment, clarification, and a membrane-based anion exchange chromatography was developed. The design of the experiment was performed with the aim to obtain the optimal conditions for the chromatography step. Additionally, a continuous mode manufacturing process integrating upstream and downstream steps was evaluated. rVSV-SARS-CoV-2 was continuously harvested from the perfusion bioreactor and purified by membrane chromatography in three columns that were operated sequentially under a counter-current mode. Compared with the batch mode, the continuous mode of operation had a 2.55-fold increase in space-time yield and a reduction in the processing time by half. The integrated continuous manufacturing process provides a reference for the efficient production of other viral vectored vaccines.
Collapse
Affiliation(s)
- Zeyu Yang
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | | | - Julia Puppin Chaves Fulber
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Michelle Yen Tran
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Omar Farnós
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Amine A Kamen
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
32
|
Yankowski C, Kurup D, Wirblich C, Schnell MJ. Effects of adjuvants in a rabies-vectored Ebola virus vaccine on protection from surrogate challenge. NPJ Vaccines 2023; 8:10. [PMID: 36754965 PMCID: PMC9906604 DOI: 10.1038/s41541-023-00615-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
Ebola virus is the primary contributor to the global threat of filovirus severe hemorrhagic fever, and Ebola virus disease has a case fatality rate of 50-90%. An inactivated, bivalent filovirus/rabies virus vaccine, FILORAB1, consists of recombinant rabies virus virions expressing the Ebola virus glycoprotein. FILORAB1 is immunogenic and protective from Ebola virus challenge in mice and non-human primates, and protection is enhanced when formulated with toll-like receptor 4 agonist Glucopyranosyl lipid adjuvant (GLA) in a squalene oil-in-water emulsion (SE). Through an adjuvant comparison in mice, we demonstrate that GLA-SE improves FILORAB1 efficacy by activating the innate immune system and shaping a Th1-biased adaptive immune response. GLA-SE adjuvanted mice and those adjuvanted with the SE component are better protected from surrogate challenge, while Th2 alum adjuvanted mice are not. Additionally, the immune response to FILORAB1 is long-lasting, as exhibited by highly-maintained serum antibody titers and long-lived cells in the spleen and bone marrow.
Collapse
Affiliation(s)
- Catherine Yankowski
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Drishya Kurup
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson Vaccine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson Vaccine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Vijver SV, Danklmaier S, Pipperger L, Gronauer R, Floriani G, Hackl H, Das K, Wollmann G. Prediction and validation of murine MHC class I epitopes of the recombinant virus VSV-GP. Front Immunol 2023; 13:1100730. [PMID: 36741416 PMCID: PMC9893851 DOI: 10.3389/fimmu.2022.1100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
Oncolytic viruses are currently tested as a novel platform for cancer therapy. These viruses preferentially replicate in and kill malignant cells. Due to their microbial origin, treatment with oncolytic viruses naturally results in anti-viral responses and general immune activation. Consequently, the oncolytic virus treatment also induces anti-viral T cells. Since these can constitute the dominant activated T cell pool, monitoring of the anti-viral T cell response may aid in better understanding of the immune responses post oncolytic virotherapy. This study aimed to identify the anti-viral T cells raised by VSV-GP virotherapy in C57BL/6J mice, one of the most widely used models for preclinical studies. VSV-GP is a novel oncolytic agent that recently entered a clinical phase I study. To identify the VSV-GP epitopes to which mouse anti-viral T cells react, we used a multilevel adapted bioinformatics viral epitope prediction approach based on the tools netMHCpan, MHCflurry and netMHCstabPan, which are commonly used in neoepitope identification. Predicted viral epitopes were ranked based on consensus binding strength categories, predicted stability, and dissimilarity to the mouse proteome. The top ranked epitopes were selected and included in the peptide candidate matrix in order to use a matrix deconvolution approach. Using ELISpot, we showed which viral epitopes presented on C57BL/6J mouse MHC-I alleles H2-Db and H2-Kb trigger IFN-γ secretion due to T cell activation. Furthermore, we validated these findings using an intracellular cytokine staining. Collectively, identification of the VSV-GP T cell epitopes enables monitoring of the full range of anti-viral T cell responses upon VSV-GP virotherapy in future studies with preclinical mouse models to more comprehensively delineate anti-viral from anti-tumor T cell responses. These findings also support the development of novel VSV-GP variants expressing immunomodulatory transgenes and can improve the assessment of anti-viral immunity in preclinical models.
Collapse
Affiliation(s)
- Saskia V. Vijver
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Danklmaier
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Lisa Pipperger
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Raphael Gronauer
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriel Floriani
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Guido Wollmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
34
|
Ithinji DG, Buchholz DW, Ezzatpour S, Monreal IA, Cong Y, Sahler J, Bangar AS, Imbiakha B, Upadhye V, Liang J, Ma A, Bradel-Tretheway B, Kaza B, Yeo YY, Choi EJ, Johnston GP, Huzella L, Kollins E, Dixit S, Yu S, Postnikova E, Ortega V, August A, Holbrook MR, Aguilar HC. Multivalent viral particles elicit safe and efficient immunoprotection against Nipah Hendra and Ebola viruses. NPJ Vaccines 2022; 7:166. [PMID: 36528644 PMCID: PMC9759047 DOI: 10.1038/s41541-022-00588-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Experimental vaccines for the deadly zoonotic Nipah (NiV), Hendra (HeV), and Ebola (EBOV) viruses have focused on targeting individual viruses, although their geographical and bat reservoir host overlaps warrant creation of multivalent vaccines. Here we explored whether replication-incompetent pseudotyped vesicular stomatitis virus (VSV) virions or NiV-based virus-like particles (VLPs) were suitable multivalent vaccine platforms by co-incorporating multiple surface glycoproteins from NiV, HeV, and EBOV onto these virions. We then enhanced the vaccines' thermotolerance using carbohydrates to enhance applicability in global regions that lack cold-chain infrastructure. Excitingly, in a Syrian hamster model of disease, the VSV multivalent vaccine elicited safe, strong, and protective neutralizing antibody responses against challenge with NiV, HeV, or EBOV. Our study provides proof-of-principle evidence that replication-incompetent multivalent viral particle vaccines are sufficient to provide protection against multiple zoonotic deadly viruses with high pandemic potential.
Collapse
Affiliation(s)
- Duncan G Ithinji
- School for Global Animal Health, Washington State University, Pullman, WA, USA.,Kenya Agricultural and Livestock Research Organization, Nairobi, Kenya
| | - David W Buchholz
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - I Abrrey Monreal
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Yu Cong
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Julie Sahler
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | | | - Brian Imbiakha
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Viraj Upadhye
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Janie Liang
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Andrew Ma
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | | | - Benjamin Kaza
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Yao Yu Yeo
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Eun Jin Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Gunner P Johnston
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Louis Huzella
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Erin Kollins
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Saurabh Dixit
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Shuiqing Yu
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Elena Postnikova
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Victoria Ortega
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Michael R Holbrook
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
35
|
New vector and vaccine platforms: mRNA, DNA, viral vectors. Curr Opin HIV AIDS 2022; 17:338-344. [DOI: 10.1097/coh.0000000000000763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Sène M, Xia Y, Kamen AA. From functional genomics of vero cells to CRISPR-based genomic deletion for improved viral production rates. Biotechnol Bioeng 2022; 119:2794-2805. [PMID: 35869699 PMCID: PMC9540595 DOI: 10.1002/bit.28190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/07/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022]
Abstract
Despite their wide use in the vaccine manufacturing field for over 40 years, one of the main limitations to recent efforts to develop Vero cells as high-throughput vaccine manufacturing platforms is the lack of understanding of virus-host interactions during infection and cell-based virus production in Vero cells. To overcome this limitation, this manuscript uses the recently generated reference genome for the Vero cell line to identify the factors at play during influenza A virus (IAV) and recombinant vesicular stomatitis virus (rVSV) infection and replication in Vero host cells. The best antiviral gene candidate for gene editing was selected using Differential Gene Expression analysis, Gene Set Enrichment Analysis and Network Topology-based Analysis. After selection of the ISG15 gene for targeted CRISPR genomic deletion, the ISG15 genomic sequence was isolated for CRISPR guide RNAs design and the guide RNAs with the highest knockout efficiency score were selected. The CRISPR experiment was then validated by confirmation of genomic deletion via PCR and further assessed via quantification of ISG15 protein levels by western blot. The gene deletion effect was assessed thereafter via quantification of virus production yield in the edited Vero cell line. A 70-fold and an 87-fold increase of total viral particles productions in ISG15-/- Vero cells was achieved for, respectively, IAV and rVSV while the ratio of infectious viral particles/total viral particles also significantly increased from 0.0316 to 0.653 for IAV and from 0.0542 to 0.679 for rVSV-GFP.
Collapse
Affiliation(s)
| | - Yu Xia
- Department of BioengineeringMcGill UniversityMontrealQuébecCanada
| | - Amine A. Kamen
- Department of BioengineeringMcGill UniversityMontrealQuébecCanada
| |
Collapse
|
37
|
Ao Z, Ouyang MJ, Olukitibi TA, Warner B, Vendramelli R, Truong T, Meilleur C, Zhang M, Kung S, Fowke KR, Kobasa D, Yao X. A Recombinant VSV-Based Bivalent Vaccine Effectively Protects against Both SARS-CoV-2 and Influenza A Virus Infection. J Virol 2022; 96:e0133722. [PMID: 36069551 PMCID: PMC9517730 DOI: 10.1128/jvi.01337-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
COVID-19 and influenza are both highly contagious respiratory diseases that have been serious threats to global public health. It is necessary to develop a bivalent vaccine to control these two infectious diseases simultaneously. In this study, we generated three attenuated replicating recombinant vesicular stomatitis virus (rVSV)-based vaccine candidates against both SARS-CoV-2 and influenza viruses. These rVSV-based vaccines coexpress SARS-CoV-2 Delta spike protein (SP) bearing the C-terminal 17 amino acid (aa) deletion (SPΔC) and I742A point mutation, or the SPΔC with a deletion of S2 domain, or the RBD domain, and a tandem repeat harboring four copies of the highly conserved influenza M2 ectodomain (M2e) that fused with the Ebola glycoprotein DC-targeting/activation domain. Animal immunization studies have shown that these rVSV bivalent vaccines induced efficient humoral and cellular immune responses against both SARS-CoV-2 SP and influenza M2 protein, including high levels of neutralizing antibodies against SARS-CoV-2 Delta and other variant SP-pseudovirus infections. Importantly, immunization of the rVSV bivalent vaccines effectively protected hamsters or mice against the challenges of SARS-CoV-2 Delta variant and lethal H1N1 and H3N2 influenza viruses and significantly reduced respiratory viral loads. Overall, this study provides convincing evidence for the high efficacy of this bivalent vaccine platform to be used and/or easily adapted to produce new vaccines against new or reemerging SARS-CoV-2 variants and influenza A virus infections. IMPORTANCE Given that both COVID-19 and influenza are preferably transmitted through respiratory droplets during the same seasons, it is highly advantageous to develop a bivalent vaccine that could simultaneously protect against both COVID-19 and influenza. In this study, we generated the attenuated replicating recombinant vesicular stomatitis virus (rVSV)-based vaccine candidates that target both spike protein of SARS-Cov-2 Delta variant and the conserved influenza M2 domain. Importantly, these vaccine candidates effectively protected hamsters or mice against the challenges of SARS-CoV-2 Delta variant and lethal H1N1 and H3N2 influenza viruses and significantly reduced respiratory viral loads.
Collapse
Affiliation(s)
- Zhujun Ao
- Laboratory of Molecular Human Retrovirology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maggie J. Ouyang
- Laboratory of Molecular Human Retrovirology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Titus A. Olukitibi
- Laboratory of Molecular Human Retrovirology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bryce Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Robert Vendramelli
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Thang Truong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Courtney Meilleur
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Manli Zhang
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sam Kung
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Keith R. Fowke
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
38
|
Khoshnood S, Ghanavati R, Shirani M, Ghahramanpour H, Sholeh M, Shariati A, Sadeghifard N, Heidary M. Viral vector and nucleic acid vaccines against COVID-19: A narrative review. Front Microbiol 2022; 13:984536. [PMID: 36118203 PMCID: PMC9470835 DOI: 10.3389/fmicb.2022.984536] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/12/2022] [Indexed: 12/14/2022] Open
Abstract
After about 2 years since the first detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in Wuhan, China, in December 2019 that resulted in a worldwide pandemic, 6.2 million deaths have been recorded. As a result, there is an urgent need for the development of a safe and effective vaccine for coronavirus disease 2019 (COVID-19). Endeavors for the production of effective vaccines inexhaustibly are continuing. At present according to the World Health Organization (WHO) COVID-19 vaccine tracker and landscape, 153 vaccine candidates are developing in the clinical phase all over the world. Some new and exciting platforms are nucleic acid-based vaccines such as Pfizer Biontech and Moderna vaccines consisting of a messenger RNA (mRNA) encoding a viral spike protein in host cells. Another novel vaccine platform is viral vector vaccine candidates that could be replicating or nonreplicating. These types of vaccines that have a harmless viral vector like adenovirus contain a genome encoding the spike protein of SARS-CoV-2, which induces significant immune responses. This technology of vaccine manufacturing has previously been used in many human clinical trials conducted for adenoviral vector-based vaccines against different infectious agents, including Ebola virus, Zika virus, HIV, and malaria. In this paper, we have a review of nucleic acid-based vaccines that are passing their phase 3 and 4 clinical trials and discuss their efficiency and adverse effects.
Collapse
Affiliation(s)
- Saeed Khoshnood
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
- Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran
| | - Roya Ghanavati
- School of Paramedical Sciences, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Maryam Shirani
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Ghahramanpour
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Sholeh
- Department of Microbiology, Pasteur Institute of Iran, Tehran, Iran
| | - Aref Shariati
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
| | - Nourkhoda Sadeghifard
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohsen Heidary
- Department of Laboratory Sciences, School of Paramedical Sciences, Sabzevar University of Medical Sciences, Sabzevar, Iran
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
- *Correspondence: Mohsen Heidary,
| |
Collapse
|
39
|
Taylor KL, Lanning L, Wolfraim L, Shrivastava Gales S, Sico C, Dowling WE, Ward LA, Florence WC, Nuzum E, Bryant PR. A U.S. Government-Coordinated Effort to Leverage Non-Human Primate Data to Facilitate Ebolavirus Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10081201. [PMID: 36016089 PMCID: PMC9412622 DOI: 10.3390/vaccines10081201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
A United States Government (USG) interagency group, the Filovirus Animal Non-Clinical Group (FANG), has been established to support the development of biodefense medical countermeasures (MCMs). As both vaccines and therapeutics are licensed using “non-traditional pathways”, such as the U.S. Food and Drug Administration’s (FDA) Animal Rule (AR), non-human primate (NHP) models and associated assays have been developed and standardized across BSL4 testing sites to evaluate candidate products. Vaccine candidates are evaluated using these NHP models, and through this public–private partnership, a meta-analysis of NHP control data has been conducted and submitted to the FDA as a master file. This is an example of how existing NHP control data can be leveraged in lieu of conducting separate natural history studies at multiple testing facilities to demonstrate the consistency of a standardized animal model for vaccine development. As a result, animal use can be minimized and the duplication of effort avoided, thus reducing the amount of time needed to conduct additional studies, as well as the cost of vaccine candidate development. This successful strategy may be applied to other pathogens of high consequence for vaccine development, and shows how strategic preparedness for biodefense can be leveraged in response to outbreaks and public health emergencies.
Collapse
Affiliation(s)
- Kimberly L. Taylor
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (L.W.); (S.S.G.); (C.S.); (W.C.F.); (E.N.); (P.R.B.)
- Correspondence:
| | - Lynda Lanning
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA;
| | - Lawrence Wolfraim
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (L.W.); (S.S.G.); (C.S.); (W.C.F.); (E.N.); (P.R.B.)
| | - Sonia Shrivastava Gales
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (L.W.); (S.S.G.); (C.S.); (W.C.F.); (E.N.); (P.R.B.)
| | - Colleen Sico
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (L.W.); (S.S.G.); (C.S.); (W.C.F.); (E.N.); (P.R.B.)
| | - William E. Dowling
- Coalition for Epidemic Preparedness Innovations, Washington, DC 20006, USA;
| | - Lucy A. Ward
- U.S. Department of Defense (DOD), Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager for Chemical, Biological, Radiological and Nuclear Medical (JPM CBRN Medical), Fort Detrick, MD 21702, USA;
| | - William C. Florence
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (L.W.); (S.S.G.); (C.S.); (W.C.F.); (E.N.); (P.R.B.)
| | - Edwin Nuzum
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (L.W.); (S.S.G.); (C.S.); (W.C.F.); (E.N.); (P.R.B.)
| | - Paula R. Bryant
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (L.W.); (S.S.G.); (C.S.); (W.C.F.); (E.N.); (P.R.B.)
| |
Collapse
|
40
|
Travieso T, Li J, Mahesh S, Mello JDFRE, Blasi M. The use of viral vectors in vaccine development. NPJ Vaccines 2022; 7:75. [PMID: 35787629 PMCID: PMC9253346 DOI: 10.1038/s41541-022-00503-y] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/15/2022] [Indexed: 12/22/2022] Open
Abstract
Vaccines represent the single most cost-efficient and equitable way to combat and eradicate infectious diseases. While traditional licensed vaccines consist of either inactivated/attenuated versions of the entire pathogen or subunits of it, most novel experimental vaccines against emerging infectious diseases employ nucleic acids to produce the antigen of interest directly in vivo. These include DNA plasmid vaccines, mRNA vaccines, and recombinant viral vectors. The advantages of using nucleic acid vaccines include their ability to induce durable immune responses, high vaccine stability, and ease of large-scale manufacturing. In this review, we present an overview of pre-clinical and clinical data on recombinant viral vector vaccines and discuss the advantages and limitations of the different viral vector platforms.
Collapse
Affiliation(s)
- Tatianna Travieso
- 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
| | - Jenny Li
- Duke University, Durham, NC, USA
| | - Sneha Mahesh
- 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
| | - Juliana Da Fonzeca Redenze E Mello
- 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
| | - 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.
| |
Collapse
|
41
|
Rupani N, Ngole ME, Lee JA, Aluisio AR, Gainey M, Perera SM, Ntamwinja LK, Matafali RM, Muhayangabo RF, Makoyi FN, Laghari R, Levine AC, Kearney AS. Effect of Recombinant Vesicular Stomatitis Virus–Zaire Ebola Virus Vaccination on Ebola Virus Disease Illness and Death, Democratic Republic of the Congo. Emerg Infect Dis 2022; 28:1180-1188. [PMID: 35608607 PMCID: PMC9155898 DOI: 10.3201/eid2806.212223] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We conducted a retrospective cohort study to assess the effect vaccination with the live-attenuated recombinant vesicular stomatitis virus–Zaire Ebola virus vaccine had on deaths among patients who had laboratory-confirmed Ebola virus disease (EVD). We included EVD-positive patients coming to an Ebola Treatment Center in eastern Democratic Republic of the Congo during 2018–2020. Overall, 25% of patients vaccinated before symptom onset died compared with 63% of unvaccinated patients. Vaccinated patients reported fewer EVD-associated symptoms, had reduced time to clearance of viral load, and had reduced length of stay at the Ebola Treatment Center. After controlling for confounders, vaccination was strongly associated with decreased deaths. Reduction in deaths was not affected by timing of vaccination before or after EVD exposure. These findings support use of preexposure and postexposure recombinant vesicular stomatitis virus–Zaire Ebola virus vaccine as an intervention associated with improved death rates, illness, and recovery time among patients with EVD.
Collapse
|
42
|
rVSV-ΔG-SARS-CoV-2-S vaccine: repeated intramuscular (IM) toxicity, local tolerance, immunogenicity and biodistribution study in NZW rabbits. Arch Toxicol 2022; 96:2329-2339. [PMID: 35577986 PMCID: PMC9110212 DOI: 10.1007/s00204-022-03302-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 11/06/2022]
Abstract
BriLife®, a vector-based vaccine that utilizes the recombinant vesicular stomatitis virus (VSV) platform to express and present the spike antigen of SARS-CoV-2, is undergoing testing in a phase 2 clinical trial in Israel. A nonclinical repeated-dose (GLP) toxicity study in New Zealand white rabbits was performed to evaluate the potential toxicity, local tolerance, immunogenicity and biodistribution of the vaccine. rVSV-ΔG-SARS-CoV-2-S (or vehicle) was administered intramuscularly to two groups of animals (106, 107 PFU/animal, n = 10/sex/group) on three occasions, at 2-week intervals, followed by a 3-week recovery period. Systemic clinical signs, local reactions, body weight, body temperature, food consumption, ophthalmology, urinalysis, clinical pathology, C-reactive protein, viremia and antibody levels were monitored. Gross pathology was performed, followed by organs/tissues collection for biodistribution and histopathological evaluation. Treatment-related changes were restricted to multifocal minimal myofiber necrosis at the injection sites, and increased lymphocytic cellularity in the iliac and mesenteric lymph nodes and in the spleen. These changes were considered related to the inflammatory reaction elicited, and correlated with a trend for recovery. Detection of rVSV-ΔG-SARS-CoV-2-S vaccine RNA was noted in the regional iliac lymph node in animals assigned to the high-dose group, at both termination time points. A significant increase in binding and neutralizing antibody titers was observed following vaccination at both vaccine doses. In view of the findings, it was concluded that the rVSV-ΔG-SARS-CoV-2-S vaccine is safe. These results supported the initiation of clinical trials.
Collapse
|
43
|
Lin JJ, Tien CF, Kuo YP, Lin EJ, Tsai WH, Chen MY, Tsai PJ, Su YW, Pathak N, Yang JM, Yu CY, Chuang ZS, Wu HC, Tsai WT, Dai SS, Liao HC, Chai KM, Su YS, Chuang TH, Liu SJ, Chen HW, Dou HY, Chen FJ, Chen CT, Liao CL, Yu GY. Furin and TMPRSS2 Resistant Spike Induces Robust Humoral and Cellular Immunity Against SARS-CoV-2 Lethal Infection. Front Immunol 2022; 13:872047. [PMID: 35585971 PMCID: PMC9108258 DOI: 10.3389/fimmu.2022.872047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/24/2022] [Indexed: 12/23/2022] Open
Abstract
An effective COVID-19 vaccine against broad SARS-CoV-2 variants is still an unmet need. In the study, the vesicular stomatitis virus (VSV)-based vector was used to express the SARS-CoV-2 Spike protein to identify better vaccine designs. The replication-competent of the recombinant VSV-spike virus with C-terminal 19 amino acid truncation (SΔ19 Rep) was generated. A single dose of SΔ19 Rep intranasal vaccination is sufficient to induce protective immunity against SARS-CoV-2 infection in hamsters. All the clones isolated from the SΔ19 Rep virus contained R682G mutation located at the Furin cleavage site. An additional S813Y mutation close to the TMPRSS2 cleavage site was identified in some clones. The enzymatic processing of S protein was blocked by these mutations. The vaccination of the R682G-S813Y virus produced a high antibody response against S protein and a robust S protein-specific CD8+ T cell response. The vaccinated animals were protected from the lethal SARS-CoV-2 (delta variant) challenge. The S antigen with resistance to enzymatic processes by Furin and TMPRSS2 will provide better immunogenicity for vaccine design.
Collapse
Affiliation(s)
- Jhe-Jhih Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Feng Tien
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Ping Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - En-Ju Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Wei-Hsiang Tsai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Ming-Yu Chen
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Pei-Ju Tsai
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Nikhil Pathak
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jinn-Moon Yang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Zih-Shiuan Chuang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Han-Chieh Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Wan-Ting Tsai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Syong Dai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Siang Su
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Horng-Yunn Dou
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Feng-Jui Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chin-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- *Correspondence: Guann-Yi Yu,
| |
Collapse
|
44
|
Sandbrink JB, Koblentz GD. Biosecurity risks associated with vaccine platform technologies. Vaccine 2022; 40:2514-2523. [PMID: 33640142 PMCID: PMC7904460 DOI: 10.1016/j.vaccine.2021.02.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/18/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
Vaccine platforms have been critical for accelerating the timeline of COVID-19 vaccine development. Faster vaccine timelines demand further development of these technologies. Currently investigated platform approaches include virally vectored and RNA-based vaccines, as well as DNA vaccines and recombinant protein expression system platforms, each featuring different advantages and challenges. Viral vector-based and DNA vaccines in particular have received a large share of research funding to date. Platform vaccine technologies may feature dual-use potential through informing or enabling pathogen engineering, which may raise the risk for the occurrence of deliberate, anthropogenic biological events. Research on virally vectored vaccines exhibits relatively high dual-use potential for two reasons. First, development of virally vectored vaccines may generate insights of particular dual-use concern such as techniques for circumventing pre-existing anti-vector immunity. Second, while the amount of work on viral vectors for gene therapy exceeds that for vaccine research, work on virally vectored vaccines may increase the number of individuals capable of engineering viruses of particular concern, such as ones closely related to smallpox. Other platform vaccine approaches, such as RNA vaccines, feature relatively little dual-use potential. The biosecurity risk associated with platform advancement may be minimised by focusing preferentially on circumventing anti-vector immunity with non-genetic rather than genetic modifications, using vectors that are not based on viruses pathogenic to humans, or preferential investment into promising RNA-based vaccine approaches. To reduce the risk of anthropogenic pandemics, structures for the governance of biotechnology and life science research with dual-use potential need to be reworked. Scientists outside of the pathogen research community, for instance those who work on viral vectors or oncolytic viruses, need to become more aware of the dual-use risks associated with their research. Both public and private research-funding bodies need to prioritise the evaluation and reduction of biosecurity risks.
Collapse
Affiliation(s)
- Jonas B Sandbrink
- Future of Humanity Institute, University of Oxford, Trajan House, Mill St, Oxford, OX2 0AN, UK; Medical Sciences Division, University of Oxford, Medical Sciences Office, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK.
| | - Gregory D Koblentz
- Schar School of Policy and Government, George Mason University, Van Metre Hall, 678 3351 Fairfax Drive Arlington, VA 22201, USA.
| |
Collapse
|
45
|
Peng KW, Carey T, Lech P, Vandergaast R, Muñoz-Alía MÁ, Packiriswamy N, Gnanadurai C, Krotova K, Tesfay M, Ziegler C, Haselton M, Sevola K, Lathrum C, Reiter S, Narjari R, Balakrishnan B, Suksanpaisan L, Sakuma T, Recker J, Zhang L, Waniger S, Russell L, Petro CD, Kyratsous CA, Baum A, Janecek JL, Lee RM, Ramachandran S, Graham ML, Russell SJ. Boosting of SARS-CoV-2 immunity in nonhuman primates using an oral rhabdoviral vaccine. Vaccine 2022; 40:2342-2351. [PMID: 35282925 PMCID: PMC8743387 DOI: 10.1016/j.vaccine.2021.12.063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 01/29/2023]
Abstract
An orally active vaccine capable of boosting SARS-CoV-2 immune responses in previously infected or vaccinated individuals would help efforts to achieve and sustain herd immunity. Unlike mRNA-loaded lipid nanoparticles and recombinant replication-defective adenoviruses, replicating vesicular stomatitis viruses with SARS-CoV-2 spike glycoproteins (VSV-SARS2) were poorly immunogenic after intramuscular administration in clinical trials. Here, by G protein trans-complementation, we generated VSV-SARS2(+G) virions with expanded target cell tropism. Compared to parental VSV-SARS2, G-supplemented viruses were orally active in virus-naive and vaccine-primed cynomolgus macaques, powerfully boosting SARS-CoV-2 neutralizing antibody titers. Clinical testing of this oral VSV-SARS2(+G) vaccine is planned.
Collapse
Affiliation(s)
- Kah-Whye Peng
- Vyriad Inc, Rochester MN 55901, USA; Imanis Life Sciences, Rochester MN 55901, USA; Department of Molecular Medicine, Mayo Clinic, MN 55905, USA
| | | | | | | | | | | | | | | | - Mulu Tesfay
- Imanis Life Sciences, Rochester MN 55901, USA
| | | | | | - Kara Sevola
- Imanis Life Sciences, Rochester MN 55901, USA
| | | | | | | | | | | | | | | | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, MN 55905, USA
| | | | | | | | | | - Alina Baum
- Regeneron Pharmaceuticals Inc, Tarrytown, NY 10591, USA
| | | | | | | | | | - Stephen J Russell
- Vyriad Inc, Rochester MN 55901, USA; Imanis Life Sciences, Rochester MN 55901, USA; Department of Molecular Medicine, Mayo Clinic, MN 55905, USA.
| |
Collapse
|
46
|
Vianello E, Gonzalez-Dias P, van Veen S, Engele CG, Quinten E, Monath TP, Medaglini D, Santoro F, Huttner A, Dubey S, Eichberg M, Ndungu FM, Kremsner PG, Essone PN, Agnandji ST, Siegrist CA, Nakaya HI, Ottenhoff THM, Haks MC. Transcriptomic signatures induced by the Ebola virus vaccine rVSVΔG-ZEBOV-GP in adult cohorts in Europe, Africa, and North America: a molecular biomarker study. THE LANCET. MICROBE 2022; 3:e113-e123. [PMID: 35544042 PMCID: PMC7613316 DOI: 10.1016/s2666-5247(21)00235-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/05/2021] [Accepted: 08/13/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND A recombinant vesicular stomatitis virus vector expressing the Zaire Ebola virus glycoprotein (rVSVΔG-ZEBOV-GP) vaccine has been reported as safe, immunogenic, and highly protective in a ring vaccination trial. We aimed to identify transcriptomic immune response biomarker signatures induced by vaccination and associated signatures with its immunogenicity and reactogenicity to better understand the potential mechanisms of action of the vaccine. METHODS 354 healthy adult volunteers were vaccinated in randomised, double-blind, placebo-controlled trials in Europe (Geneva, Switzerland [November, 2014, to January, 2015]) and North America (USA [Dec 5, 2014, to June 23, 2015]), and dose-escalation trials in Africa (Lambaréné, Gabon [November, 2014, to January, 2015], and Kilifi, Kenya [December, 2014, to January, 2015]) using different doses of the recombinant vesicular stomatitis virus vector expressing the Zaire Ebola virus glycoprotein (rVSVΔG-ZEBOV-GP; 3 × 105 to 1 × 108 plaque-forming units [pfu]). Longitudinal transcriptomic responses (days 0, 1, 2, 3, 7, 14, and 28) were measured in whole blood using a targeted gene expression profiling platform (dual-colour reverse-transcriptase multiplex ligation-dependent probe amplification) focusing on 144 immune-related genes. The effect of time and dose on transcriptomic response was also assessed. Logistic regression with lasso regularisation was applied to identify host signatures with optimal discriminatory capability of vaccination at day 1 or day 7 versus baseline, whereas random-effects models and recursive feature elimination combined with regularised logistic regression were used to associate signatures with immunogenicity and reactogenicity. FINDINGS Our results indicated that perturbation of gene expression peaked on day 1 and returned to baseline levels between day 7 and day 28. The magnitude of the response was dose-dependent, with vaccinees receiving a high dose (≥9 × 106 pfu) of rVSVΔG-ZEBOV-GP exhibiting the largest amplitude. The most differentially expressed genes that were significantly upregulated following vaccination consisted of type I and II interferon-related genes and myeloid cell-associated markers, whereas T cell, natural killer cell, and cytotoxicity-associated genes were downregulated. A gene signature associated with immunogenicity (common to all four cohorts) was identified correlating gene expression profiles with ZEBOV-GP antibody titres and a gene signatures associated with reactogenicity (Geneva cohort) was identified correlating gene expression profiles with an adverse event (ie, arthritis). INTERPRETATION Collectively, our results identify and cross-validate immune-related transcriptomic signatures induced by rVSVΔG-ZEBOV-GP vaccination in four cohorts of adult participants from different genetic and geographical backgrounds. These signatures will aid in the rational development, testing, and evaluation of novel vaccines and will allow evaluation of the effect of host factors such as age, co-infection, and comorbidity on responses to vaccines. FUNDING Innovative Medicines Initiative 2 Joint Undertaking.
Collapse
Affiliation(s)
- Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| | - Patricia Gonzalez-Dias
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Carmen G Engele
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Edwin Quinten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Sclavo Vaccines Association, Siena, Italy
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Angela Huttner
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Center for Vaccinology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Sheri Dubey
- Department of Vaccine and Biologics Research, Merck Research Laboratories, West Point, PA, USA
| | - Michael Eichberg
- Department of Vaccine and Biologics Research, Merck Research Laboratories, West Point, PA, USA
| | - Francis M Ndungu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, and German Center for Infection Research, Tübingen, Germany
| | - Paulin N Essone
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, and German Center for Infection Research, Tübingen, Germany
| | - Claire-Anne Siegrist
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Center for Vaccinology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur-USP, São Paulo, Brazil
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
47
|
Madar-Balakirski N, Rosner A, Melamed S, Politi B, Steiner M, Tamir H, Yahalom-Ronen Y, Bar-David E, Ben-Shmuel A, Sittner A, Glinert I, Weiss S, Bar-Haim E, Cohen H, Elia U, Achdout H, Erez N, Rotem S, Lazar S, Nyska A, Yitzhaki S, Beth-Din A, Levy H, Paran N, Israely T, Marcus H. Preliminary nonclinical safety and immunogenicity of an rVSV-ΔG-SARS-CoV-2-S vaccine in mice, hamsters, rabbits and pigs. Arch Toxicol 2022; 96:859-875. [PMID: 35032184 PMCID: PMC8760087 DOI: 10.1007/s00204-021-03214-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/21/2021] [Indexed: 12/18/2022]
Abstract
rVSV-ΔG-SARS-CoV-2-S is a clinical stage (Phase 2) replication competent recombinant vaccine against SARS-CoV-2. To evaluate the safety profile of the vaccine, a series of non-clinical safety, immunogenicity and efficacy studies were conducted in four animal species, using multiple doses (up to 108 Plaque Forming Units/animal) and dosing regimens. There were no treatment-related mortalities or any noticeable clinical signs in any of the studies. Compared to unvaccinated controls, hematology and biochemistry parameters were unremarkable and no adverse histopathological findings. There was no detectable viral shedding in urine, nor viral RNA detected in whole blood or serum samples seven days post vaccination. The rVSV-ΔG-SARS-CoV-2-S vaccination gave rise to neutralizing antibodies, cellular immune responses, and increased lymphocytic cellularity in the spleen germinal centers and regional lymph nodes. No evidence for neurovirulence was found in C57BL/6 immune competent mice or in highly sensitive type I interferon knock-out mice. Vaccine virus replication and distribution in K18-human Angiotensin-converting enzyme 2-transgenic mice showed a gradual clearance from the vaccination site with no vaccine virus recovered from the lungs. The nonclinical data suggest that the rVSV-ΔG-SARS-CoV-2-S vaccine is safe and immunogenic. These results supported the initiation of clinical trials, currently in Phase 2.
Collapse
Affiliation(s)
- Noa Madar-Balakirski
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Amir Rosner
- Veterinary Center for Preclinical Research, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Boaz Politi
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | | | - Hadas Tamir
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Yfat Yahalom-Ronen
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Elad Bar-David
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Amir Ben-Shmuel
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Assa Sittner
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Itai Glinert
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shay Weiss
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Hila Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Uri Elia
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Hagit Achdout
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Noam Erez
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shahar Rotem
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shlomi Lazar
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Abraham Nyska
- Sackler School of Medicine, Tel Aviv University, and Consultant in Toxicologic Pathology, Tel Aviv, Israel
| | - Shmuel Yitzhaki
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Adi Beth-Din
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Haim Levy
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel.
| | - Hadar Marcus
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel.
| |
Collapse
|
48
|
Murin CD, Gilchuk P, Crowe JE, Ward AB. Structural Biology Illuminates Molecular Determinants of Broad Ebolavirus Neutralization by Human Antibodies for Pan-Ebolavirus Therapeutic Development. Front Immunol 2022; 12:808047. [PMID: 35082794 PMCID: PMC8784787 DOI: 10.3389/fimmu.2021.808047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/06/2021] [Indexed: 01/13/2023] Open
Abstract
Monoclonal antibodies (mAbs) have proven effective for the treatment of ebolavirus infection in humans, with two mAb-based drugs Inmazeb™ and Ebanga™ receiving FDA approval in 2020. While these drugs represent a major advance in the field of filoviral therapeutics, they are composed of antibodies with single-species specificity for Zaire ebolavirus. The Ebolavirus genus includes five additional species, two of which, Bundibugyo ebolavirus and Sudan ebolavirus, have caused severe disease and significant outbreaks in the past. There are several recently identified broadly neutralizing ebolavirus antibodies, including some in the clinical development pipeline, that have demonstrated broad protection in preclinical studies. In this review, we describe how structural biology has illuminated the molecular basis of broad ebolavirus neutralization, including details of common antigenic sites of vulnerability on the glycoprotein surface. We begin with a discussion outlining the history of monoclonal antibody therapeutics for ebolaviruses, with an emphasis on how structural biology has contributed to these efforts. Next, we highlight key structural studies that have advanced our understanding of ebolavirus glycoprotein structures and mechanisms of antibody-mediated neutralization. Finally, we offer examples of how structural biology has contributed to advances in anti-viral medicines and discuss what opportunities the future holds, including rationally designed next-generation therapeutics with increased potency, breadth, and specificity against ebolaviruses.
Collapse
MESH Headings
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antiviral Agents/immunology
- Antiviral Agents/therapeutic use
- Drug Combinations
- Ebolavirus/drug effects
- Ebolavirus/immunology
- Ebolavirus/physiology
- Epitopes/chemistry
- Epitopes/immunology
- Glycoproteins/chemistry
- Glycoproteins/immunology
- Hemorrhagic Fever, Ebola/drug therapy
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/virology
- Humans
- Models, Molecular
- Protein Domains/immunology
- Viral Proteins/chemistry
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Charles D. Murin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
49
|
Kim GN, Choi JA, Wu K, Saeedian N, Yang E, Park H, Woo SJ, Lim G, Kim SG, Eo SK, Jeong HW, Kim T, Chang JH, Seo SH, Kim NH, Choi E, Choo S, Lee S, Winterborn A, Li Y, Parham K, Donovan JM, Fenton B, Dikeakos JD, Dekaban GA, Haeryfar SMM, Troyer RM, Arts EJ, Barr SD, Song M, Kang CY. A vesicular stomatitis virus-based prime-boost vaccination strategy induces potent and protective neutralizing antibodies against SARS-CoV-2. PLoS Pathog 2021; 17:e1010092. [PMID: 34914812 PMCID: PMC8675757 DOI: 10.1371/journal.ppat.1010092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 11/04/2021] [Indexed: 12/17/2022] Open
Abstract
The development of safe and effective vaccines to prevent SARS-CoV-2 infections remains an urgent priority worldwide. We have used a recombinant vesicular stomatitis virus (rVSV)-based prime-boost immunization strategy to develop an effective COVID-19 vaccine candidate. We have constructed VSV genomes carrying exogenous genes resulting in the production of avirulent rVSV carrying the full-length spike protein (SF), the S1 subunit, or the receptor-binding domain (RBD) plus envelope (E) protein of SARS-CoV-2. Adding the honeybee melittin signal peptide (msp) to the N-terminus enhanced the protein expression, and adding the VSV G protein transmembrane domain and the cytoplasmic tail (Gtc) enhanced protein incorporation into pseudotype VSV. All rVSVs expressed three different forms of SARS-CoV-2 spike proteins, but chimeras with VSV-Gtc demonstrated the highest rVSV-associated expression. In immunized mice, rVSV with chimeric S protein-Gtc derivatives induced the highest level of potent neutralizing antibodies and T cell responses, and rVSV harboring the full-length msp-SF-Gtc proved to be the superior immunogen. More importantly, rVSV-msp-SF-Gtc vaccinated animals were completely protected from a subsequent SARS-CoV-2 challenge. Overall, we have developed an efficient strategy to induce a protective response in SARS-CoV-2 challenged immunized mice. Vaccination with our rVSV-based vector may be an effective solution in the global fight against COVID-19. The COVID-19 pandemic has had unprecedented global health, economic and societal impact globally. Vaccinating the majority of the world’s population is the best way to help prevent new infections. Many vaccines have been developed to prevent various viral diseases that are currently in use around the world. This has generated a high demand for these vaccines, putting a strain on production capacity and delivery. With new variants of concern starting to dominate the human pandemic, new derivatives of the current vaccines may be necessary for continued protection from SARS-CoV-2 infection. We have developed a vaccine that uses a safe vesicular stomatitis virus-based delivery vehicle to present a key SARS-CoV-2 protein to our immune system in order to train it to recognize and prevent SARS-CoV-2 infection. Our vaccine completely protected vaccinated animals from SARS-CoV-2 infection and significantly reduced lung damage, a major hallmark of COVID-19.
Collapse
Affiliation(s)
- Gyoung Nyoun Kim
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Jung-ah Choi
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Kunyu Wu
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Nasrin Saeedian
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Eunji Yang
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Hayan Park
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Sun-Je Woo
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Gippeum Lim
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Seong-Gyu Kim
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Su-Kyeong Eo
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Hoe Won Jeong
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Taewoo Kim
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Jae-Hyung Chang
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Sang Hwan Seo
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Na Hyung Kim
- Sumagen, 4F Dongwon Bldg, Teheran-ro 77-gil, Gangnam-gu, Seoul, Korea
| | - Eunsil Choi
- Sumagen, 4F Dongwon Bldg, Teheran-ro 77-gil, Gangnam-gu, Seoul, Korea
| | - Seungho Choo
- Sumagen, 4F Dongwon Bldg, Teheran-ro 77-gil, Gangnam-gu, Seoul, Korea
| | - Sangkyun Lee
- Sumagen, 4F Dongwon Bldg, Teheran-ro 77-gil, Gangnam-gu, Seoul, Korea
| | | | - Yue Li
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Kate Parham
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Justin M. Donovan
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Brock Fenton
- Department of Biology, Faculty of Science, The University of Western Ontario, London, Ontario, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Gregory A. Dekaban
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Ryan M. Troyer
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Eric J. Arts
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Stephen D. Barr
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Manki Song
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - C. Yong Kang
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
50
|
Martinón-Torres F, García-Sastre A, Pollard AJ, Martín C, Osterhaus A, Ladhani SN, Ramilo O, Gómez Rial J, Salas A, Bosch FX, Martinón-Torres M, Mina MJ, Cherry J. TIPICO XI: report of the first series and podcast on infectious diseases and vaccines (aTIPICO). Hum Vaccin Immunother 2021; 17:4299-4327. [PMID: 34762551 PMCID: PMC8828069 DOI: 10.1080/21645515.2021.1953351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TIPiCO is an annual expert meeting and workshop on infectious diseases and vaccination. The edition of 2020 changed its name and format to aTIPiCO, the first series and podcasts on infectious diseases and vaccines. A total of 13 prestigious experts from different countries participated in this edition launched on the 26 November 2020. The state of the art of coronavirus disease-2019 (COVID-19) and the responsible pathogen, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), and the options to tackle the pandemic situation were discussed in light of the knowledge in November 2020. Despite COVID-19, the status of other infectious diseases, including influenza infections, respiratory syncytial virus disease, human papillomavirus infection, measles, pertussis, tuberculosis, meningococcal disease, and pneumococcal disease, were also addressed. The essential lessons that can be learned from these diseases and their vaccines to use in the COVID-19 pandemic were also commented with the experts.
Collapse
Affiliation(s)
- Federico Martinón-Torres
- Department of Paediatrics Translational Paediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, Universidad de Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Carlos Martín
- Department of Microbiology, Faculty of Medicine, IIS Aragon, Universidad de Zaragoza, CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Albert Osterhaus
- Research Center Emerging Infections and Zoonoses (RIZ, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Octavio Ramilo
- Nationwide Children's Hospital and the Ohio State University, Columbus, Ohio, US
| | - Jose Gómez Rial
- Immunology Department, Hospital Clínico Universitario de Santiago de Compostela, Spain
| | - Antonio Salas
- Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
| | | | | | - Michael J Mina
- Harvard School of Public Health and Harvard Medical School, Boston, MA, US
| | - James Cherry
- The David Geffen School of Medicine at UCLA, Los Angeles, CA, US
| |
Collapse
|