1
|
de La Vega MA, XIII A, Massey CS, Spengler JR, Kobinger GP, Woolsey C. An update on nonhuman primate usage for drug and vaccine evaluation against filoviruses. Expert Opin Drug Discov 2024; 19:1185-1211. [PMID: 39090822 PMCID: PMC11466704 DOI: 10.1080/17460441.2024.2386100] [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: 06/23/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Due to their faithful recapitulation of human disease, nonhuman primates (NHPs) are considered the gold standard for evaluating drugs against Ebolavirus and other filoviruses. The long-term goal is to reduce the reliance on NHPs with more ethical alternatives. In silico simulations and organoid models have the potential to revolutionize drug testing by providing accurate, human-based systems that mimic disease processes and drug responses without the ethical concerns associated with animal testing. However, as these emerging technologies are still in their developmental infancy, NHP models are presently needed for late-stage evaluation of filovirus vaccines and drugs, as they provide critical insights into the efficacy and safety of new medical countermeasures. AREAS COVERED In this review, the authors introduce available NHP models and examine the existing literature on drug discovery for all medically significant filoviruses in corresponding models. EXPERT OPINION A deliberate shift toward animal-free models is desired to align with the 3Rs of animal research. In the short term, the use of NHP models can be refined and reduced by enhancing replicability and publishing negative data. Replacement involves a gradual transition, beginning with the selection and optimization of better small animal models; advancing organoid systems, and using in silico models to accurately predict immunological outcomes.
Collapse
Affiliation(s)
- Marc-Antoine de La Vega
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Ara XIII
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Christopher S. Massey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Jessica R. Spengler
- Viral Special Pathogens Branch and Infectious Diseases
Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for
Disease Control and Prevention, Atlanta, GA
| | - Gary P. Kobinger
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| |
Collapse
|
2
|
Kisakov DN, Belyakov IM, Kisakova LA, Yakovlev VA, Tigeeva EV, Karpenko LI. The use of electroporation to deliver DNA-based vaccines. Expert Rev Vaccines 2024; 23:102-123. [PMID: 38063059 DOI: 10.1080/14760584.2023.2292772] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Nucleic acids represent a promising platform for creating vaccines. One disadvantage of this approach is its relatively low immunogenicity. Electroporation (EP) is an effective way to increase the DNA vaccines immunogenicity. However, due to the different configurations of devices used for EP, EP protocols optimization is required not only to enhance immunogenicity, but also to ensure greater safety and tolerability of the EP procedure. AREA COVERED An data analysis for recent years on the DNA vaccines delivery against viral and parasitic infections using EP was carried out. The study of various EP physical characteristics, such as frequency, pulse duration, pulse interval, should be considered along with the immunogenic construct design and the site of delivery of the vaccine, through the study of the immunogenic and protective characteristics of the latter. EXPERT OPINION Future research should focus on regulating the humoral and cellular response required for protection against infectious agents by modifying the EP protocol. Significant efforts will be directed to establishing the possibility of redirecting the immune response toward the Th1 or Th2 response by changing the EP physical parameters. It will allow for an individual selective approach during EP, depending on the pathogen type of an infectious disease.
Collapse
Affiliation(s)
- Denis N Kisakov
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Igor M Belyakov
- Department of medico-biological disciplines, Moscow University for Industry and Finance "Synergy", Moscow, Russia
| | - Lubov A Kisakova
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Vladimir A Yakovlev
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Elena V Tigeeva
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Larisa I Karpenko
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| |
Collapse
|
3
|
de Lima MR, Leandro ACCS, de Souza AL, Barradas MM, Roma EH, Fernandes ATG, Galdino-Silva G, Carvalho JKMR, Marchevsky RS, Coelho JMCO, Gonçalves EDC, VandeBerg JL, Silva CL, Bonecini-Almeida MDG. Safety and Immunogenicity of an In Vivo Muscle Electroporation Delivery System for DNA- hsp65 Tuberculosis Vaccine in Cynomolgus Monkeys. Vaccines (Basel) 2023; 11:1863. [PMID: 38140266 PMCID: PMC10747856 DOI: 10.3390/vaccines11121863] [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: 10/20/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
A Bacille Calmette-Guérin (BCG) is still the only licensed vaccine for the prevention of tuberculosis, providing limited protection against Mycobacterium tuberculosis infection in adulthood. New advances in the delivery of DNA vaccines by electroporation have been made in the past decade. We evaluated the safety and immunogenicity of the DNA-hsp65 vaccine administered by intramuscular electroporation (EP) in cynomolgus macaques. Animals received three doses of DNA-hsp65 at 30-day intervals. We demonstrated that intramuscular electroporated DNA-hsp65 vaccine immunization of cynomolgus macaques was safe, and there were no vaccine-related effects on hematological, renal, or hepatic profiles, compared to the pre-vaccination parameters. No tuberculin skin test conversion nor lung X-ray alteration was identified. Further, low and transient peripheral cellular immune response and cytokine expression were observed, primarily after the third dose of the DNA-hsp65 vaccine. Electroporated DNA-hsp65 vaccination is safe but provides limited enhancement of peripheral cellular immune responses. Preclinical vaccine trials with DNA-hsp65 delivered via EP may include a combination of plasmid cytokine adjuvant and/or protein prime-boost regimen, to help the induction of a stronger cellular immune response.
Collapse
Affiliation(s)
- Monique Ribeiro de Lima
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Ana Cristina C. S. Leandro
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
- Division of Human Genetics, South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX 78520, USA;
| | - Andreia Lamoglia de Souza
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Marcio Mantuano Barradas
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Eric Henrique Roma
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Ana Teresa Gomes Fernandes
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Gabrielle Galdino-Silva
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Joyce Katiuccia M. Ramos Carvalho
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Renato Sergio Marchevsky
- Laboratory of Neurovirulence, Instituto de Biotecnologia em Imunobiológicos, Biomanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Janice M. C. Oliveira Coelho
- Laboratory of Pathology, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil;
| | | | - John L. VandeBerg
- Division of Human Genetics, South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX 78520, USA;
| | - Celio Lopes Silva
- Farmacore Biotecnologia Ltda, Ribeirão Preto 14056-680, SP, Brazil; (E.D.C.G.); (C.L.S.)
- Laboratory for Research and Development of Immunobiologicals, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Maria da Gloria Bonecini-Almeida
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| |
Collapse
|
4
|
Zhang HQ, Zhang QY, Yuan ZM, Zhang B. The potential epidemic threat of Ebola virus and the development of a preventive vaccine. JOURNAL OF BIOSAFETY AND BIOSECURITY 2023; 5:67-78. [DOI: 10.1016/j.jobb.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
5
|
Cross RW, Prasad AN, Woolsey CB, Agans KN, Borisevich V, Dobias NS, Comer JE, Deer DJ, Geisbert JB, Rasmussen AL, Lipkin WI, Fenton KA, Geisbert TW. Natural history of nonhuman primates after conjunctival exposure to Ebola virus. Sci Rep 2023; 13:4175. [PMID: 36914721 PMCID: PMC10011569 DOI: 10.1038/s41598-023-31027-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
Transmission of Ebola virus (EBOV) primarily occurs via contact exposure of mucosal surfaces with infected body fluids. Historically, nonhuman primate (NHP) challenge studies have employed intramuscular (i.m.) or small particle aerosol exposure, which are largely lethal routes of infection, but mimic worst-case scenarios such as a needlestick or intentional release, respectively. When exposed by more likely routes of natural infection, limited NHP studies have shown delayed onset of disease and reduced mortality. Here, we performed a series of systematic natural history studies in cynomolgus macaques with a range of conjunctival exposure doses. Challenge with 10,000 plaque forming units (PFU) of EBOV was uniformly lethal, whereas 5/6 subjects survived lower dose challenges (100 or 500 PFU). Conjunctival challenge resulted in a protracted time-to death compared to i.m. Asymptomatic infection was observed in survivors with limited detection of EBOV replication. Inconsistent seropositivity in survivors may suggest physical or natural immunological barriers are sufficient to prevent widespread viral dissemination.
Collapse
Affiliation(s)
- Robert W Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Abhishek N Prasad
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Courtney B Woolsey
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Krystle N Agans
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Viktoriya Borisevich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Natalie S Dobias
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Jason E Comer
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Daniel J Deer
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Joan B Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Angela L Rasmussen
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY, 10032, USA
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Walter Ian Lipkin
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY, 10032, USA
| | - Karla A Fenton
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA
| | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77550, USA.
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, 77550, USA.
| |
Collapse
|
6
|
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
|
7
|
Gomez AM, Babuadze G(G, Plourde-Campagna MA, Azizi H, Berger A, Kozak R, de La Vega MA, XIII A, Naghibosadat M, Nepveu-Traversy ME, Ruel J, Kobinger GP. A novel intradermal tattoo-based injection device enhances the immunogenicity of plasmid DNA vaccines. NPJ Vaccines 2022; 7:172. [PMID: 36543794 PMCID: PMC9771775 DOI: 10.1038/s41541-022-00581-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/24/2022] [Indexed: 12/24/2022] Open
Abstract
In recent years, tattooing technology has shown promising results toward evaluating vaccines in both animal models and humans. However, this technology has some limitations due to variability of experimental evaluations or operator procedures. The current study evaluated a device (intradermal oscillating needle array injection device: IONAID) capable of microinjecting a controlled dose of any aqueous vaccine into the intradermal space. IONAID-mediated administration of a DNA-based vaccine encoding the glycoprotein (GP) from the Ebola virus resulted in superior T- and B-cell responses with IONAID when compared to single intramuscular (IM) or intradermal (ID) injection in mice. Moreover, humoral immune responses, induced after IONAID vaccination, were significantly higher to those obtained with traditional passive DNA tattooing in guinea pigs and rabbits. This device was well tolerated and safe during HIV vaccine delivery in non-human primates (NHPs), while inducing robust immune responses. In summary, this study shows that the IONAID device improves vaccine performance, which could be beneficial to the animal and human health, and importantly, provide a dose-sparing approach (e.g., monkeypox vaccine).
Collapse
Affiliation(s)
- Alejandro M. Gomez
- grid.23856.3a0000 0004 1936 8390Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6 Canada
| | - George (Giorgi) Babuadze
- grid.17063.330000 0001 2157 2938Biological Sciences Platform, University Toronto, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | | | - Hiva Azizi
- grid.23856.3a0000 0004 1936 8390Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6 Canada
| | - Alice Berger
- grid.23856.3a0000 0004 1936 8390Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6 Canada
| | - Robert Kozak
- grid.17063.330000 0001 2157 2938Biological Sciences Platform, University Toronto, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | - Marc-Antoine de La Vega
- grid.176731.50000 0001 1547 9964Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555 USA
| | - Ara XIII
- grid.176731.50000 0001 1547 9964Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555 USA
| | - Maedeh Naghibosadat
- grid.17063.330000 0001 2157 2938Biological Sciences Platform, University Toronto, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | | | - Jean Ruel
- grid.23856.3a0000 0004 1936 8390Département de Génie Mécanique, Université Laval, Québec, QC G1V 0A6 Canada
| | - Gary P. Kobinger
- grid.176731.50000 0001 1547 9964Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555 USA
| |
Collapse
|
8
|
Evaluation of Zika virus DNA vaccines based on NS1 and domain III of E. Int Immunopharmacol 2022; 113:109308. [DOI: 10.1016/j.intimp.2022.109308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
|
9
|
Choi H, Kudchodkar SB, Xu Z, Ho M, Xiao P, Ramos S, Humeau L, Weiner DB, Muthumani K. Elicitation of immune responses against Nipah virus by an engineered synthetic DNA vaccine. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2022.968338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Nipah virus (NiV) is a re-emerging pathogen that causes severe disease in animals and humans. Current treatment measures for NiV infection are insufficient, and there is no approved vaccine against NiV for either humans or animals. Nipah virus is listed as a high-priority pathogen for vaccine and therapeutic research by the World Health Organization (WHO). In the present study, we employed synthetic enhanced DNA technologies developed to design and produce novel consensus NiV Fusion (NiV-F) and Glycoprotein (NiV-G) antigen sequences for inclusion in synthetic DNA vaccines for NiV. The expression of each vaccine antigen was confirmed in vitro using immune-binding assays. Electroporation-enhanced intramuscular injection of each NiV-F and NiV-G into mice induced potent cellular immune responses to multiple epitopes of NiV-G and NiV-F that included antigen-specific CD8+ T cells. Both vaccines elicited high antibody titers in mice, with a single immunization sufficient to seroconvert 100% of immunized animals. Additionally, the NiV-F vaccine also induced antibodies to neutralize NiV-F-pseudotyped virus particles. These data support further study of these novel synthetic enhanced NiV nucleic acid-based antigens as potential components of an effective vaccine against the Nipah virus.
Collapse
|
10
|
Liu Y, Sun B, Pan J, Feng Y, Ye W, Xu J, Lan M, Sun H, Zhang X, Sun Y, Yang S, Shi J, Zhang F, Cheng L, Jiang D, Yang K. Construction and evaluation of DNA vaccine encoding Ebola virus glycoprotein fused with lysosome-associated membrane protein. Antiviral Res 2021; 193:105141. [PMID: 34274417 DOI: 10.1016/j.antiviral.2021.105141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/20/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Ebola virus (EBOV) of the genus Ebolavirus belongs to the family Filoviridae, which cause disease in both humans and non-human primates. Zaire Ebola virus accounts for the highest fatality rate, reaching 90%. Considering that EBOV has a high infection and fatality rate, the development of a highly effective vaccine has become a top public health priority. Glycoprotein (GP) plays a critical role during infection and protective immune responses. Herein, we developed an EBOV GP recombinant DNA vaccine that targets the major histocompatibility complex (MHC) class II compartment by fusing with lysosomal-associated membrane protein 1 (LAMP1). Through lysosome trafficking and antigen presentation transferring, the LAMP1 targeting strategy successfully improved both humoral and cellular EBOV-GP-specific immune responses. After three consecutive immunizations, the serum antibody titers, especially the neutralizing activity of mice immunized with the pVAX-LAMP/GPEBO vaccine were significantly higher than those of the other groups. Antigen-specific T cells showed positive activity against three dominant peptides, EAAVSHLTTLATIST, IGEWAFWETKKNLTR, and ELRTFSILNRKAIDF, with high affinity for MHC class II molecules predicted by IEDB-recommended. Preliminary safety observation denied histological alterations. DNA vaccine candidate pVAX-LAMP/GPEBO shows promise against Ebola epidemic and further evaluation is guaranteed.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- BALB 3T3 Cells
- Ebola Vaccines/administration & dosage
- Ebola Vaccines/adverse effects
- Ebola Vaccines/genetics
- Ebola Vaccines/immunology
- Ebolavirus/genetics
- Ebolavirus/immunology
- Female
- Glycoproteins/genetics
- Glycoproteins/immunology
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Humans
- Lysosomal Membrane Proteins/genetics
- Lysosomal Membrane Proteins/immunology
- Mice
- Neutralization Tests
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/adverse effects
- Vaccines, DNA/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Yang Liu
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Baozeng Sun
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Jingyu Pan
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Yuancai Feng
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Wei Ye
- Department of Microbiology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Jiahao Xu
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Mingfu Lan
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Hao Sun
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Xiyang Zhang
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Yuanjie Sun
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Shuya Yang
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Jingqi Shi
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Fanglin Zhang
- Department of Microbiology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Linfeng Cheng
- Department of Microbiology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China
| | - Dongbo Jiang
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China.
| | - Kun Yang
- Department of Immunology, Basic Medicine School, Air-Force Medical University (the Fourth Military Medical University), Xi'an, Shaanxi, 86-710032, PR China.
| |
Collapse
|
11
|
Machhi J, Shahjin F, Das S, Patel M, Abdelmoaty MM, Cohen JD, Singh PA, Baldi A, Bajwa N, Kumar R, Vora LK, Patel TA, Oleynikov MD, Soni D, Yeapuri P, Mukadam I, Chakraborty R, Saksena CG, Herskovitz J, Hasan M, Oupicky D, Das S, Donnelly RF, Hettie KS, Chang L, Gendelman HE, Kevadiya BD. Nanocarrier vaccines for SARS-CoV-2. Adv Drug Deliv Rev 2021; 171:215-239. [PMID: 33428995 PMCID: PMC7794055 DOI: 10.1016/j.addr.2021.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/18/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 global pandemic has seen rapid spread, disease morbidities and death associated with substantive social, economic and societal impacts. Treatments rely on re-purposed antivirals and immune modulatory agents focusing on attenuating the acute respiratory distress syndrome. No curative therapies exist. Vaccines remain the best hope for disease control and the principal global effort to end the pandemic. Herein, we summarize those developments with a focus on the role played by nanocarrier delivery.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Srijanee Das
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA; Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt
| | - Jacob D Cohen
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Preet Amol Singh
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Ashish Baldi
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Neha Bajwa
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Raj Kumar
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lalit K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Tapan A Patel
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology (CHARUSAT), Changa, Anand 388421, Gujarat, India
| | - Maxim D Oleynikov
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Dhruvkumar Soni
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Rajashree Chakraborty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Caroline G Saksena
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Jonathan Herskovitz
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Suvarthi Das
- Department of Medicine, Stanford Medical School, Stanford University, Palo Alto, CA 94304, USA
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Kenneth S Hettie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Department of Otolaryngology - Head & Neck Surgery, Stanford University, Palo Alto, CA 94304, USA
| | - Linda Chang
- Departments of Diagnostic Radiology & Nuclear Medicine, and Neurology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA.
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| |
Collapse
|
12
|
Fan Y, Yen CW, Lin HC, Hou W, Estevez A, Sarode A, Goyon A, Bian J, Lin J, Koenig SG, Leung D, Nagapudi K, Zhang K. Automated high-throughput preparation and characterization of oligonucleotide-loaded lipid nanoparticles. Int J Pharm 2021; 599:120392. [PMID: 33639228 DOI: 10.1016/j.ijpharm.2021.120392] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/25/2021] [Accepted: 02/11/2021] [Indexed: 01/18/2023]
Abstract
Lipid nanoparticles (LNPs) are increasingly employed to improve delivery efficiency and therapeutic efficacy of nucleic acids. Various formulation parameters can affect the quality attributes of these nanoparticle formulations, but currently there is a lack of systemic screening approaches to address this challenge. Here, we developed an automated high-throughput screening (HTS) workflow for streamline preparation and analytical characterization of LNPs loaded with antisense oligonucleotides (ASOs) in a full 96-well plate within 3 hrs. ASO-loaded LNPs were formulated by an automated solvent-injection method using a robotic liquid handler, and assessed for particle size distribution, encapsulation efficiency, and stability with different formulation compositions and ASO loadings. Results indicated that the PEGylated lipid content significantly affected the particle size distribution, while the ionizable lipid / ASO charge ratio impacted the encapsulation efficiency of ASOs. Furthermore, results from our HTS approach correlated with those from the state-of-the-art scale-up method using a microfluidic formulator, therefore opening up a new avenue for robust formulation development and design of experiment methods, while reducing material usage by 10 folds, improving analytical outputs and accumulation of information by 100 folds.
Collapse
Affiliation(s)
- Yuchen Fan
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Chun-Wan Yen
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hsiu-Chao Lin
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Weijia Hou
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alberto Estevez
- Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Apoorva Sarode
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alexandre Goyon
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Juan Bian
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jessica Lin
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stefan G Koenig
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dennis Leung
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kelly Zhang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
13
|
Karpenko LI, Apartsin EK, Dudko SG, Starostina EV, Kaplina ON, Antonets DV, Volosnikova EA, Zaitsev BN, Bakulina AY, Venyaminova AG, Ilyichev AA, Bazhan SI. Cationic Polymers for the Delivery of the Ebola DNA Vaccine Encoding Artificial T-Cell Immunogen. Vaccines (Basel) 2020; 8:vaccines8040718. [PMID: 33271964 PMCID: PMC7760684 DOI: 10.3390/vaccines8040718] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 11/16/2022] Open
Abstract
Background: According to current data, an effective Ebola virus vaccine should induce both humoral and T-cell immunity. In this work, we focused our efforts on methods for delivering artificial T-cell immunogen in the form of a DNA vaccine, using generation 4 polyamidoamine dendrimers (PAMAM G4) and a polyglucin:spermidine conjugate (PG). Methods: Optimal conditions were selected for obtaining complexes of previously developed DNA vaccines with cationic polymers. The sizes, mobility and surface charge of the complexes with PG and PAMAM 4G have been determined. The immunogenicity of the obtained vaccine constructs was investigated in BALB/c mice. Results: It was shown that packaging of DNA vaccine constructs both in the PG envelope and the PAMAM 4G envelope results in an increase in their immunogenicity as compared with the group of mice immunized with the of vector plasmid pcDNA3.1 (a negative control). The highest T-cell responses were shown in mice immunized with complexes of DNA vaccines with PG and these responses significantly exceeded those in the groups of animals immunized with both the combination of naked DNAs and the combination DNAs coated with PAMAM 4G. In the group of animals immunized with complexes of the DNA vaccines with PAMAM 4G, no statistical differences were found in the ability to induce T-cell responses, as compared with the group of mice immunized with the combination of naked DNAs. Conclusions: The PG conjugate can be considered as a promising and safe means to deliver DNA-based vaccines. The use of PAMAM requires further optimization.
Collapse
Affiliation(s)
- Larisa I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
- Correspondence: (L.I.K.); (S.I.B.); Tel.: +7-383-363-47-00 (ext. 2001) (L.I.K. & S.I.B.)
| | - Evgeny K. Apartsin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.K.A.); (A.G.V.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
- Laboratoire de Chimie de Coordination, CNRS, 31077 Toulouse, France
| | - Sergei G. Dudko
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Ekaterina V. Starostina
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Olga N. Kaplina
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Denis V. Antonets
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Ekaterina A. Volosnikova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Boris N. Zaitsev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Anastasiya Yu. Bakulina
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Aliya G. Venyaminova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.K.A.); (A.G.V.)
| | - Alexander A. Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Sergei I. Bazhan
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
- Correspondence: (L.I.K.); (S.I.B.); Tel.: +7-383-363-47-00 (ext. 2001) (L.I.K. & S.I.B.)
| |
Collapse
|
14
|
A novel synthetic DNA vaccine elicits protective immune responses against Powassan virus. PLoS Negl Trop Dis 2020; 14:e0008788. [PMID: 33119599 PMCID: PMC7595275 DOI: 10.1371/journal.pntd.0008788] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/10/2020] [Indexed: 01/07/2023] Open
Abstract
Powassan virus (POWV) infection is a tick-borne emerging infectious disease in the United States and North America. Like Zika virus, POWV is a member of the family Flaviviridae. POWV causes severe neurological sequalae, meningitis, encephalitis, and can cause death. Although the risk of human POWV infection is low, its incidence in the U.S. in the past 16 years has increased over 300%, urging immediate attention. Despite the disease severity and its growing potential for threatening larger populations, currently there are no licensed vaccines which provide protection against POWV. We developed a novel synthetic DNA vaccine termed POWV-SEV by focusing on the conserved portions of POWV pre-membrane and envelope (prMEnv) genes. A single immunization of POWV-SEV elicited broad T and B cell immunity in mice with minimal cross-reactivity against other flaviviruses. Antibody epitope mapping demonstrated a similarity between POWV-SEV-induced immune responses and those elicited naturally in POWV-infected patients. Finally, POWV-SEV induced immunity provided protection against POWV disease in lethal challenge experiments.
Collapse
|
15
|
Xu Z, Patel A, Tursi NJ, Zhu X, Muthumani K, Kulp DW, Weiner DB. Harnessing Recent Advances in Synthetic DNA and Electroporation Technologies for Rapid Vaccine Development Against COVID-19 and Other Emerging Infectious Diseases. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:571030. [PMID: 35047878 PMCID: PMC8757735 DOI: 10.3389/fmedt.2020.571030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
DNA vaccines are considered as a third-generation vaccination approach in which antigenic materials are encoded as DNA plasmids for direct in vivo production to elicit adaptive immunity. As compared to other platforms, DNA vaccination is considered to have a strong safety profile, as DNA plasmids neither replicate nor elicit vector-directed immune responses in hosts. While earlier work found the immune responses induced by DNA vaccines to be sub-optimal in larger mammals and humans, recent developments in key synthetic DNA and electroporation delivery technologies have now allowed DNA vaccines to elicit significantly more potent and consistent responses in several clinical studies. This paper will review findings from the recent clinical and preclinical studies on DNA vaccines targeting emerging infectious diseases (EID) including COVID-19 caused by the SARS-CoV-2 virus, and the technological advancements pivotal to the improved responses-including the use of the advanced delivery technology, DNA-encoded cytokine/mucosal adjuvants, and innovative concepts in immunogen design. With continuous advancement over the past three decades, the DNA approach is now poised to develop vaccines against COVID-19, as well as other EIDs.
Collapse
Affiliation(s)
- Ziyang Xu
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ami Patel
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Nicholas J. Tursi
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Xizhou Zhu
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Kar Muthumani
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Daniel W. Kulp
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - David B. Weiner
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
16
|
Gary EN, Weiner DB. DNA vaccines: prime time is now. Curr Opin Immunol 2020; 65:21-27. [PMID: 32259744 PMCID: PMC7195337 DOI: 10.1016/j.coi.2020.01.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 01/07/2023]
Abstract
Recently newer synthetic DNA vaccines have been rapidly advanced to clinical study and have demonstrated an impressive degree of immune potency and tolerability. Improvements in DNA delivery over prior needle and syringe approaches include jet delivery, gene gun delivery, among others. Among the most effective of these new delivery methods, advanced electroporation (EP), combined with other advances, induces robust humoral and cellular immunity in both preventative as well as therapeutic studies. Advancements in the design of the DNA inserts include leader sequence changes, RNA and codon optimizations, improved insert designs, increased concentrations of DNA, and skin delivery, appear to complement newer delivery strategies. These advances also provide a framework for the in vivo production of synthetic DNA biologics. In this review, we focus on recent studies of synthetic DNA vaccines in the clinic for the prevention or treatment of infectious diseases with a focus on adaptive electroporation for delivery, and briefly summarize novel preclinical data advancing the in vivo delivery of DNA-encoded antibody-like biologics.
Collapse
|
17
|
Matz KM, Marzi A, Feldmann H. Ebola vaccine trials: progress in vaccine safety and immunogenicity. Expert Rev Vaccines 2020; 18:1229-1242. [PMID: 31779496 DOI: 10.1080/14760584.2019.1698952] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction: Ebolaviruses are non-segmented negative-strand RNA viruses in the Filoviridae family that cause a neglected infectious disease designated as Ebola virus disease (EVD). The most prominent member is the Ebola virus (EBOV), representing the Zaire ebolavirus species that has been responsible for the largest reported EVD outbreaks including the West African epidemic and the current outbreak in the Democratic Republic of the Congo. Today, the most advanced EVD vaccine approaches target EBOV and multiple phase 1-4 human trials have been performed over the past few years. The most advanced platforms include vectored vaccines based on vesicular stomatitis virus (VSV-EBOV), distinct human (Ad5 and Ad26) and chimpanzee (ChAd3) adenoviruses and modified vaccinia Ankara (MVA) as well as DNA-based vaccines administered as a prime-only or homologous or combined prime-boost immunization.Areas covered: Here, we review and discuss human trials with a focus on vaccine safety and immunogenicity.Expert opinion: Despite obvious progress and promising success in EBOV vaccine development, many shortcomings and challenges remain to be tackled in the future.
Collapse
Affiliation(s)
- Keesha M Matz
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| |
Collapse
|
18
|
Abstract
Since its discovery in 1976, Ebola virus (EBOV) has caused numerous outbreaks of fatal hemorrhagic disease in Africa. The biggest outbreak on record is the 2013-2016 epidemic in west Africa with almost 30,000 cases and over 11,000 fatalities, devastatingly affecting Guinea, Liberia, and Sierra Leone. The epidemic highlighted the need for licensed drugs or vaccines to quickly combat the disease. While at the beginning of the epidemic no licensed countermeasures were available, several experimental drugs with preclinical efficacy were accelerated into human clinical trials and used to treat patients with Ebola virus disease (EVD) toward the end of the epidemic. In the same manner, vaccines with preclinical efficacy were administered primarily to known contacts of EVD patients on clinical trial protocols using a ring-vaccination strategy. In this review, we describe the pathogenesis of EBOV and summarize the current status of EBOV vaccine development and treatment of EVD.
Collapse
Affiliation(s)
- Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
| |
Collapse
|
19
|
Smith TRF, Patel A, Ramos S, Elwood D, Zhu X, Yan J, Gary EN, Walker SN, Schultheis K, Purwar M, Xu Z, Walters J, Bhojnagarwala P, Yang M, Chokkalingam N, Pezzoli P, Parzych E, Reuschel EL, Doan A, Tursi N, Vasquez M, Choi J, Tello-Ruiz E, Maricic I, Bah MA, Wu Y, Amante D, Park DH, Dia Y, Ali AR, Zaidi FI, Generotti A, Kim KY, Herring TA, Reeder S, Andrade VM, Buttigieg K, Zhao G, Wu JM, Li D, Bao L, Liu J, Deng W, Qin C, Brown AS, Khoshnejad M, Wang N, Chu J, Wrapp D, McLellan JS, Muthumani K, Wang B, Carroll MW, Kim JJ, Boyer J, Kulp DW, Humeau LMPF, Weiner DB, Broderick KE. Immunogenicity of a DNA vaccine candidate for COVID-19. Nat Commun 2020; 11:2601. [PMID: 32433465 PMCID: PMC7239918 DOI: 10.1038/s41467-020-16505-0] [Citation(s) in RCA: 427] [Impact Index Per Article: 85.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/08/2020] [Indexed: 01/07/2023] Open
Abstract
The coronavirus family member, SARS-CoV-2 has been identified as the causal agent for the pandemic viral pneumonia disease, COVID-19. At this time, no vaccine is available to control further dissemination of the disease. We have previously engineered a synthetic DNA vaccine targeting the MERS coronavirus Spike (S) protein, the major surface antigen of coronaviruses, which is currently in clinical study. Here we build on this prior experience to generate a synthetic DNA-based vaccine candidate targeting SARS-CoV-2 S protein. The engineered construct, INO-4800, results in robust expression of the S protein in vitro. Following immunization of mice and guinea pigs with INO-4800 we measure antigen-specific T cell responses, functional antibodies which neutralize the SARS-CoV-2 infection and block Spike protein binding to the ACE2 receptor, and biodistribution of SARS-CoV-2 targeting antibodies to the lungs. This preliminary dataset identifies INO-4800 as a potential COVID-19 vaccine candidate, supporting further translational study.
Collapse
Affiliation(s)
- Trevor R. F. Smith
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Ami Patel
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Stephanie Ramos
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Dustin Elwood
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Xizhou Zhu
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Jian Yan
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Ebony N. Gary
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Susanne N. Walker
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Katherine Schultheis
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Mansi Purwar
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Ziyang Xu
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Jewell Walters
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Pratik Bhojnagarwala
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Maria Yang
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Neethu Chokkalingam
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Patrick Pezzoli
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Elizabeth Parzych
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Emma L. Reuschel
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Arthur Doan
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Nicholas Tursi
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Miguel Vasquez
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Jihae Choi
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Edgar Tello-Ruiz
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Igor Maricic
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Mamadou A. Bah
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Yuanhan Wu
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Dinah Amante
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Daniel H. Park
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Yaya Dia
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Ali Raza Ali
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Faraz I. Zaidi
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Alison Generotti
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Kevin Y. Kim
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Timothy A. Herring
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Sophia Reeder
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Viviane M. Andrade
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Karen Buttigieg
- 0000 0004 5909 016Xgrid.271308.fNational Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Gan Zhao
- Advaccine (Suzhou) Biopharmaceuticals Co., Ltd, Suzhou, China
| | - Jiun-Ming Wu
- Advaccine (Suzhou) Biopharmaceuticals Co., Ltd, Suzhou, China
| | - Dan Li
- 0000 0001 0125 2443grid.8547.eKey Laboratory of Medical Molecular Virology of MOH and MOE and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Linlin Bao
- 0000 0001 0125 2443grid.8547.eKey Laboratory of Medical Molecular Virology of MOH and MOE and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiangning Liu
- 0000 0001 0125 2443grid.8547.eKey Laboratory of Medical Molecular Virology of MOH and MOE and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Deng
- 0000 0001 0125 2443grid.8547.eKey Laboratory of Medical Molecular Virology of MOH and MOE and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chuan Qin
- 0000 0001 0125 2443grid.8547.eKey Laboratory of Medical Molecular Virology of MOH and MOE and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ami Shah Brown
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Makan Khoshnejad
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Nianshuang Wang
- 0000 0004 1936 9924grid.89336.37Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712 USA
| | - Jacqueline Chu
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Daniel Wrapp
- 0000 0004 1936 9924grid.89336.37Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712 USA
| | - Jason S. McLellan
- 0000 0004 1936 9924grid.89336.37Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712 USA
| | - Kar Muthumani
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Bin Wang
- 0000 0001 0125 2443grid.8547.eKey Laboratory of Medical Molecular Virology of MOH and MOE and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Miles W. Carroll
- 0000 0004 5909 016Xgrid.271308.fNational Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - J. Joseph Kim
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Jean Boyer
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - Daniel W. Kulp
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Laurent M. P. F. Humeau
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| | - David B. Weiner
- 0000 0001 1956 6678grid.251075.4Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104 USA
| | - Kate E. Broderick
- 0000 0004 0417 098Xgrid.421774.3Inovio Pharmaceuticals, Plymouth Meeting, Philadelphia, PA 19462 USA
| |
Collapse
|
20
|
O'Donnell K, Marzi A. The Ebola virus glycoprotein and its immune responses across multiple vaccine platforms. Expert Rev Vaccines 2020; 19:267-277. [PMID: 32129120 DOI: 10.1080/14760584.2020.1738225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: For over 40 years, ebolaviruses have been responsible for sporadic outbreaks of severe and often fatal hemorrhagic fever in humans and nonhuman primates across western and central Africa. In December 2013, an unprecedented Ebola virus (EBOV) epidemic began in West Africa and resulted in the largest outbreak to date. The past and current epidemics in West Africa and the Democratic Republic of the Congo has focused attention on the potential vaccine platforms developed over the past 20 years.Areas covered: This review summarizes the extraordinary progress using a variety of vaccination platforms including DNA, subunit, and several viral vector approaches, replicating and non-replicating, incorporating the primary antigen of EBOV, the glycoprotein. These vaccine constructs have shown varying degrees of protective efficacy in the 'gold-standard' nonhuman primate model for EBOV infections and were immunogenic in human clinical trials.Expert commentary: A number of these vaccine platforms have moved into phase III clinical trials over the past years and with the recent approval of the first EBOV vaccine in the European Union and the USA there is a strong potential to prevent future outbreaks/epidemics of EBOV infections on the scale of the West African epidemic.
Collapse
Affiliation(s)
- Kyle O'Donnell
- Laboratory of Virology, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
21
|
Yenkoidiok-Douti L, Jewell CM. Integrating Biomaterials and Immunology to Improve Vaccines Against Infectious Diseases. ACS Biomater Sci Eng 2020; 6:759-778. [PMID: 33313391 PMCID: PMC7725244 DOI: 10.1021/acsbiomaterials.9b01255] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite the success of vaccines in preventing many infectious diseases, effective vaccines against pathogens with ongoing challenges - such as HIV, malaria, and tuberculosis - remain unavailable. The emergence of new pathogen variants, the continued prevalence of existing pathogens, and the resurgence of yet other infectious agents motivate the need for new, interdisciplinary approaches to direct immune responses. Many current and candidate vaccines, for example, are poorly immunogenic, provide only transient protection, or create risks of regaining pathogenicity in certain immune-compromised conditions. Recent advances in biomaterials research are creating new potential to overcome these challenges through improved formulation, delivery, and control of immune signaling. At the same time, many of these materials systems - such as polymers, lipids, and self-assembly technologies - may achieve this goal while maintaining favorable safety profiles. This review highlights ways in which biomaterials can advance existing vaccines to safer, more efficacious technologies, and support new vaccines for pathogens that do not yet have vaccines. Biomaterials that have not yet been applied to vaccines for infectious disease are also discussed, and their potential in this area is highlighted.
Collapse
Affiliation(s)
- Lampouguin Yenkoidiok-Douti
- Fischell Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Drive, College Park, MD, 20742, United States
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Rockville, MD, 20852, United States
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Drive, College Park, MD, 20742, United States
- Department of Veterans Affairs, VA Maryland Health Care System, 10. N Green Street, Baltimore, MD 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD 20742, United States
- Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, 22 S. Greene Street, Suite N9E17, Baltimore, MD 21201, United States
| |
Collapse
|
22
|
Reeder SM, Reuschel EL, Bah MA, Yun K, Tursi NJ, Kim KY, Chu J, Zaidi FI, Yilmaz I, Hart RJ, Perrin B, Xu Z, Humeau L, Weiner DB, Aly ASI. Synthetic DNA Vaccines Adjuvanted with pIL-33 Drive Liver-Localized T Cells and Provide Protection from Plasmodium Challenge in a Mouse Model. Vaccines (Basel) 2020; 8:vaccines8010021. [PMID: 31936739 PMCID: PMC7157753 DOI: 10.3390/vaccines8010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
The need for a malaria vaccine is indisputable. A single vaccine for Plasmodium pre-erythrocytic stages targeting the major sporozoite antigen circumsporozoite protein (CSP) has had partial success. Additionally, CD8+ T cells targeting liver-stage (LS) antigens induced by live attenuated sporozoite vaccines were associated with protection in human challenge experiments. To further evaluate protection mediated by LS antigens, we focused on exported pre-erythrocytic proteins (exported protein 1 (EXP1), profilin (PFN), exported protein 2 (EXP2), inhibitor of cysteine proteases (ICP), transmembrane protein 21 (TMP21), and upregulated in infective sporozoites-3 (UIS3)) expressed in all Plasmodium species and designed optimized, synthetic DNA (synDNA) immunogens. SynDNA antigen cocktails were tested with and without the molecular adjuvant plasmid IL-33. Immunized animals developed robust T cell responses including induction of antigen-specific liver-localized CD8+ T cells, which were enhanced by the co-delivery of plasmid IL-33. In total, 100% of mice in adjuvanted groups and 71%–88% in non-adjuvanted groups were protected from blood-stage disease following Plasmodium yoelii sporozoite challenge. This study supports the potential of synDNA LS antigens as vaccine components for malaria parasite infection.
Collapse
Affiliation(s)
- Sophia M. Reeder
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emma L. Reuschel
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - Mamadou A. Bah
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - Kun Yun
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Kevin Y. Kim
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - Jacqueline Chu
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - Faraz I. Zaidi
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - Ilknur Yilmaz
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul 34820, Turkey
| | - Robert J. Hart
- Department of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Benjamin Perrin
- Department of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Ziyang Xu
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laurent Humeau
- Inovio Pharmaceuticals, Inc., Plymouth Meeting, PA 19462, USA
| | - David B. Weiner
- The Vaccine Center, Wistar Institute, Philadelphia, PA 19104, USA
- Correspondence: (D.B.W.); (A.S.I.A.)
| | - Ahmed S. I. Aly
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul 34820, Turkey
- Department of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
- Correspondence: (D.B.W.); (A.S.I.A.)
| |
Collapse
|
23
|
Suschak JJ, Schmaljohn CS. Vaccines against Ebola virus and Marburg virus: recent advances and promising candidates. Hum Vaccin Immunother 2019; 15:2359-2377. [PMID: 31589088 DOI: 10.1080/21645515.2019.1651140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The filoviruses Ebola virus and Marburg virus are among the most dangerous pathogens in the world. Both viruses cause viral hemorrhagic fever, with case fatality rates of up to 90%. Historically, filovirus outbreaks had been relatively small, with only a few hundred cases reported. However, the recent West African Ebola virus outbreak underscored the threat that filoviruses pose. The three year-long outbreak resulted in 28,646 Ebola virus infections and 11,323 deaths. The lack of Food and Drug Administration (FDA) licensed vaccines and antiviral drugs hindered early efforts to contain the outbreak. In response, the global scientific community has spurred the advanced development of many filovirus vaccine candidates. Novel vaccine platforms, such as viral vectors and DNA vaccines, have emerged, leading to the investigation of candidate vaccines that have demonstrated protective efficacy in small animal and nonhuman primate studies. Here, we will discuss several of these vaccine platforms with a particular focus on approaches that have advanced into clinical development.
Collapse
Affiliation(s)
- John J Suschak
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | - Connie S Schmaljohn
- Headquarters Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
24
|
Tebas P, Kraynyak KA, Patel A, Maslow JN, Morrow MP, Sylvester AJ, Knoblock D, Gillespie E, Amante D, Racine T, McMullan T, Jeong M, Roberts CC, Park YK, Boyer J, Broderick KE, Kobinger GP, Bagarazzi M, Weiner DB, Sardesai NY, White SM. Intradermal SynCon® Ebola GP DNA Vaccine Is Temperature Stable and Safely Demonstrates Cellular and Humoral Immunogenicity Advantages in Healthy Volunteers. J Infect Dis 2019; 220:400-410. [DOI: 10.1093/infdis/jiz132] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/18/2019] [Indexed: 11/13/2022] Open
Abstract
AbstractBackgroundNonlive vaccine approaches that are simple to deliver and stable at room temperature or 2–8°C could be advantageous in controlling future Ebola virus (EBOV) outbreaks. Using an immunopotent DNA vaccine that generates protection from lethal EBOV challenge in small animals and nonhuman primates, we performed a clinical study to evaluate both intramuscular (IM) and novel intradermal (ID) DNA delivery.MethodsTwo DNA vaccine candidates (INO-4201 and INO-4202) targeting the EBOV glycoprotein (GP) were evaluated for safety, tolerability, and immunogenicity in a phase 1 clinical trial. The candidates were evaluated alone, together, or in combination with plasmid-encoded human cytokine interleukin-12 followed by in vivo electroporation using either the CELLECTRA® IM or ID delivery devices.ResultsThe safety profile of all 5 regimens was shown to be benign, with the ID route being better tolerated. Antibodies to EBOV GP were generated by all 5 regimens with the fastest and steepest rise observed in the ID group. Cellular immune responses were generated with every regimen.ConclusionsID delivery of INO-4201 was well tolerated and resulted in 100% seroreactivity after 2 doses and elicited interferon-γ T-cell responses in over 70% of subjects, providing a new approach for EBOV prevention in diverse populations.Clinical Trials Registration. NCT02464670.
Collapse
Affiliation(s)
| | | | - Ami Patel
- The Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania
| | | | | | | | | | | | - Dinah Amante
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| | | | | | | | | | | | - Jean Boyer
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| | | | | | | | - David B Weiner
- The Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania
| | | | - Scott M White
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| |
Collapse
|