1
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024:10.1038/s41573-024-01041-z. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
2
|
He Y, Orlet J, Cunio O, Thompson L, Jones MT. Sensitive and rapid analysis of plasmid DNA topology isoforms by capillary gel electrophoresis with laser induced fluorescence in uncoated capillary. Electrophoresis 2024; 45:1389-1397. [PMID: 38415778 DOI: 10.1002/elps.202300212] [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: 09/23/2023] [Revised: 01/28/2024] [Accepted: 02/11/2024] [Indexed: 02/29/2024]
Abstract
The work describes the use of SYBR Gold to improve the detection sensitivity of plasmid DNA topoisomers by capillary gel electrophoresis with laser induced fluorescence in an uncoated capillary. The impact of different dyes, including ethidium bromide, SYBR Green and SYBR Gold, was compared based on detection and separation of DNA plasmid topoisomers. Use of SYBR Gold enabled improvement of detection sensitivity by 15-fold while maintaining good separation resolution of the different topoisomers. The baseline dropped with the use SYBR Gold but was overcome by the employment of a capillary with longer ineffective length (40 vs. 20 cm). Separation resolution and reproducibility were impacted by the concentration of SYBR Gold and hydroxypropyl methylcellulose. With the use of a short capillary (10 cm effective length and 50 cm total length), fast separations of supercoiled, linear, open circular, and other isoforms were accomplished within 8 min. Appropriate capillary cleaning with 0.1 M sodium hydroxide/0.1 M hydrochloric acid and capillary storage with 0.1 M hydrochloric acid ensured good separation reproducibility of 217 runs during an extended period of use.
Collapse
Affiliation(s)
- Yan He
- Analytical Research and Development, Pfizer Inc, Chesterfield, Missouri, USA
| | - John Orlet
- Analytical Research and Development, Pfizer Inc, Chesterfield, Missouri, USA
| | - Olivia Cunio
- Analytical Research and Development, Pfizer Inc, Chesterfield, Missouri, USA
| | - Lawrence Thompson
- Analytical Research and Development, Pfizer Inc, Chesterfield, Missouri, USA
| | - Michael T Jones
- Analytical Research and Development, Pfizer Inc, Chesterfield, Missouri, USA
| |
Collapse
|
3
|
Mobasher M, Ansari R, Castejon AM, Barar J, Omidi Y. Advanced nanoscale delivery systems for mRNA-based vaccines. Biochim Biophys Acta Gen Subj 2024; 1868:130558. [PMID: 38185238 DOI: 10.1016/j.bbagen.2024.130558] [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: 11/07/2023] [Revised: 12/24/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
The effectiveness of messenger RNA (mRNA) vaccines, especially those designed for COVID-19, relies heavily on sophisticated delivery systems that ensure efficient delivery of mRNA to target cells. A variety of nanoscale vaccine delivery systems (VDSs) have been explored for this purpose, including lipid nanoparticles (LNPs), liposomes, and polymeric nanoparticles made from biocompatible polymers such as poly(lactic-co-glycolic acid), as well as viral vectors and lipid-polymer hybrid complexes. Among these, LNPs are particularly notable for their efficiency in encapsulating and protecting mRNA. These nanoscale VDSs can be engineered to enhance stability and facilitate uptake by cells. The choice of delivery system depends on factors like the specific mRNA vaccine, target cell types, stability requirements, and desired immune response. In this review, we shed light on recent advances in delivery mechanisms for self-amplifying RNA (saRNA) vaccines, emphasizing groundbreaking studies on nanoscale delivery systems aimed at improving the efficacy and safety of mRNA/saRNA vaccines.
Collapse
Affiliation(s)
- Maha Mobasher
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Rais Ansari
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Ana M Castejon
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
4
|
Nepveu-Traversy ME, Fausther-Bovendo H, Babuadze G(G. Human Tick-Borne Diseases and Advances in Anti-Tick Vaccine Approaches: A Comprehensive Review. Vaccines (Basel) 2024; 12:141. [PMID: 38400125 PMCID: PMC10891567 DOI: 10.3390/vaccines12020141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
This comprehensive review explores the field of anti-tick vaccines, addressing their significance in combating tick-borne diseases of public health concern. The main objectives are to provide a brief epidemiology of diseases affecting humans and a thorough understanding of tick biology, traditional tick control methods, the development and mechanisms of anti-tick vaccines, their efficacy in field applications, associated challenges, and future prospects. Tick-borne diseases (TBDs) pose a significant and escalating threat to global health and the livestock industries due to the widespread distribution of ticks and the multitude of pathogens they transmit. Traditional tick control methods, such as acaricides and repellents, have limitations, including environmental concerns and the emergence of tick resistance. Anti-tick vaccines offer a promising alternative by targeting specific tick proteins crucial for feeding and pathogen transmission. Developing vaccines with antigens based on these essential proteins is likely to disrupt these processes. Indeed, anti-tick vaccines have shown efficacy in laboratory and field trials successfully implemented in livestock, reducing the prevalence of TBDs. However, some challenges still remain, including vaccine efficacy on different hosts, polymorphisms in ticks of the same species, and the economic considerations of adopting large-scale vaccine strategies. Emerging technologies and approaches hold promise for improving anti-tick vaccine development and expanding their impact on public health and agriculture.
Collapse
Affiliation(s)
| | - Hugues Fausther-Bovendo
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 75550, USA;
| | - George (Giorgi) Babuadze
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 75550, USA;
| |
Collapse
|
5
|
Cunliffe RF, Stirling DC, Razzano I, Murugaiah V, Montomoli E, Kim S, Wane M, Horton H, Caproni LJ, Tregoning JS. Optimizing a linear 'Doggybone' DNA vaccine for influenza virus through the incorporation of DNA targeting sequences and neuraminidase antigen. DISCOVERY IMMUNOLOGY 2024; 3:kyad030. [PMID: 38567290 PMCID: PMC10917164 DOI: 10.1093/discim/kyad030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/08/2023] [Accepted: 01/02/2024] [Indexed: 04/04/2024]
Abstract
Influenza virus represents a challenge for traditional vaccine approaches due to its seasonal changes and potential for zoonotic transmission. Nucleic acid vaccines can overcome some of these challenges, especially through the inclusion of multiple antigens to increase the breadth of response. RNA vaccines were an important part of the response to the COVID-19 pandemic, but for future outbreaks DNA vaccines may have some advantages in terms of stability and manufacturing cost that warrant continuing investigation to fully realize their potential. Here, we investigate influenza virus vaccines made using a closed linear DNA platform, Doggybone™ DNA (dbDNA), produced by a rapid and scalable cell-free method. Influenza vaccines have mostly focussed on Haemagglutinin (HA), but the inclusion of Neuraminidase (NA) may provide additional protection. Here, we explored the potential of including NA in a dbDNA vaccine, looking at DNA optimization, mechanism and breadth of protection. We showed that DNA targeting sequences (DTS) improved immune responses against HA but not NA. We explored whether NA vaccine-induced protection against influenza virus infection was cell-mediated, but depletion of CD8 and NK cells made no impact, suggesting it was antibody-mediated. This is reflected in the restriction of protection to homologous strains of influenza virus. Importantly, we saw that including both HA and NA in a single combined vaccine did not dampen the immune response to either one. Overall, we show that linear dbDNA can induce an immune response against NA, which may offer increased protection in instances of HA mismatch where NA remains more conserved.
Collapse
Affiliation(s)
- Robert F Cunliffe
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - David C Stirling
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Ilaria Razzano
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
- VisMederi srl, Siena, 53100, Italia
| | | | - Emanuele Montomoli
- VisMederi srl, Siena, 53100, Italia
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Sungwon Kim
- Touchlight Genetics Ltd, Hampton, TW12 2ER, UK
| | - Madina Wane
- Touchlight Genetics Ltd, Hampton, TW12 2ER, UK
| | | | | | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| |
Collapse
|
6
|
Pagliari S, Dema B, Sanchez-Martinez A, Montalvo Zurbia-Flores G, Rollier CS. DNA Vaccines: History, Molecular Mechanisms and Future Perspectives. J Mol Biol 2023; 435:168297. [PMID: 37797831 DOI: 10.1016/j.jmb.2023.168297] [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: 07/28/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/07/2023]
Abstract
The history of DNA vaccine began as early as the 1960s with the discovery that naked DNA can transfect mammalian cells in vivo. In 1992, the evidence that such transfection could lead to the generation of antigen-specific antibody responses was obtained and supported the development of this technology as a novel vaccine platform. The technology then attracted immense interest and high hopes in vaccinology, as evidence of high immunogenicity and protection against virulent challenges accumulated from several animal models for several diseases. In particular, the capacity to induce T-cell responses was unprecedented in non-live vaccines. However, the technology suffered its major knock when the success in animals failed to translate to humans, where DNA vaccine candidates were shown to be safe but remained poorly immunogenic, or not associated with clinical benefit. Thanks to a thorough exploration of the molecular mechanisms of action of these vaccines, an impressive range of approaches have been and are currently being explored to overcome this major challenge. Despite limited success so far in humans as compared with later genetic vaccine technologies such as viral vectors and mRNA, DNA vaccines are not yet optimised for human use and may still realise their potential.
Collapse
Affiliation(s)
- Sthefany Pagliari
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Barbara Dema
- Pandemic Science Institute, Institute of Developmental and Regenerative Medicine (IDRM), University of Oxford, Oxford, UK
| | | | | | - Christine S Rollier
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.
| |
Collapse
|
7
|
Abbas MN, Jmel MA, Mekki I, Dijkgraaf I, Kotsyfakis M. Recent Advances in Tick Antigen Discovery and Anti-Tick Vaccine Development. Int J Mol Sci 2023; 24:4969. [PMID: 36902400 PMCID: PMC10003026 DOI: 10.3390/ijms24054969] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Ticks can seriously affect human and animal health around the globe, causing significant economic losses each year. Chemical acaricides are widely used to control ticks, which negatively impact the environment and result in the emergence of acaricide-resistant tick populations. A vaccine is considered as one of the best alternative approaches to control ticks and tick-borne diseases, as it is less expensive and more effective than chemical controls. Many antigen-based vaccines have been developed as a result of current advances in transcriptomics, genomics, and proteomic techniques. A few of these (e.g., Gavac® and TickGARD®) are commercially available and are commonly used in different countries. Furthermore, a significant number of novel antigens are being investigated with the perspective of developing new anti-tick vaccines. However, more research is required to develop new and more efficient antigen-based vaccines, including on assessing the efficiency of various epitopes against different tick species to confirm their cross-reactivity and their high immunogenicity. In this review, we discuss the recent advancements in the development of antigen-based vaccines (traditional and RNA-based) and provide a brief overview of recent discoveries of novel antigens, along with their sources, characteristics, and the methods used to test their efficiency.
Collapse
Affiliation(s)
- Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Mohamed Amine Jmel
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Imen Mekki
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Ingrid Dijkgraaf
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Michail Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| |
Collapse
|
8
|
mRNA-From COVID-19 Treatment to Cancer Immunotherapy. Biomedicines 2023; 11:biomedicines11020308. [PMID: 36830845 PMCID: PMC9953480 DOI: 10.3390/biomedicines11020308] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
This review provides an overview covering mRNA from its use in the COVID-19 pandemic to cancer immunotherapy, starting from the selection of appropriate antigens, tumor-associated and tumor-specific antigens, neoantigens, the basics of optimizing the mRNA molecule in terms of stability, efficacy, and tolerability, choosing the best formulation and the optimal route of administration, to summarizing current clinical trials of mRNA vaccines in tumor therapy.
Collapse
|
9
|
Hayashi H, Sun J, Yanagida Y, Otera T, Kubota-Koketsu R, Shioda T, Ono C, Matsuura Y, Arase H, Yoshida S, Nakamaru R, Ju N, Ide R, Tenma A, Kawabata S, Ehara T, Sakaguchi M, Tomioka H, Shimamura M, Okamoto S, Amaishi Y, Chono H, Mineno J, Komatsuno T, Saito Y, Rakugi H, Morishita R, Nakagami H. Preclinical study of a DNA vaccine targeting SARS-CoV-2. Curr Res Transl Med 2022; 70:103348. [PMID: 35489099 PMCID: PMC9020527 DOI: 10.1016/j.retram.2022.103348] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/09/2022] [Accepted: 04/16/2022] [Indexed: 01/31/2023]
Abstract
To fight against the worldwide COVID-19 pandemic, the development of an effective and safe vaccine against SARS-CoV-2 is required. As potential pandemic vaccines, DNA/RNA vaccines, viral vector vaccines and protein-based vaccines have been rapidly developed to prevent pandemic spread worldwide. In this study, we designed plasmid DNA vaccine targeting the SARS-CoV-2 Spike glycoprotein (S protein) as pandemic vaccine, and the humoral, cellular, and functional immune responses were characterized to support proceeding to initial human clinical trials. After intramuscular injection of DNA vaccine encoding S protein with alum adjuvant (three times at 2-week intervals), the humoral immunoreaction, as assessed by anti-S protein or anti-receptor-binding domain (RBD) antibody titers, and the cellular immunoreaction, as assessed by antigen-induced IFNγ expression, were up-regulated. In IgG subclass analysis, IgG2b was induced as the main subclass. Based on these analyses, DNA vaccine with alum adjuvant preferentially induced Th1-type T cell polarization. We confirmed the neutralizing action of DNA vaccine-induced antibodies by a binding assay of RBD recombinant protein with angiotensin-converting enzyme 2 (ACE2), a receptor of SARS-CoV-2, and neutralization assays using pseudo-virus, and live SARS-CoV-2. Further B cell epitope mapping analysis using a peptide array showed that most vaccine-induced antibodies recognized the S2 and RBD subunits. Finally, DNA vaccine protected hamsters from SARS-CoV-2 infection. In conclusion, DNA vaccine targeting the spike glycoprotein of SARS-CoV-2 might be an effective and safe approach to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Hiroki Hayashi
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan
| | - Jiao Sun
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan
| | - Yuka Yanagida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan
| | - Takako Otera
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan; Anges Inc, Japan
| | - Ritsuko Kubota-Koketsu
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Japan; Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Tatsuo Shioda
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Japan
| | - Chikako Ono
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Japan; Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Japan; Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Japan; Laboratory of Immunochemistry, WPI Immunology Frontier Research Centre, Osaka University, Japan
| | - Shota Yoshida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan; Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Japan
| | - Ryo Nakamaru
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan; Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Japan
| | - Nan Ju
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan; Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | - Munehisa Shimamura
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | - Hiromi Rakugi
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Japan; Lead contact, Japan.
| |
Collapse
|
10
|
Volokhov DV, Furtak V, Allen C, Pulle G, Zajac MD, Levin Y, Kochba E, Moore SM. Robust humoral immune response against rabies virus in rabbits and guinea pigs immunized with plasmid DNA vectors encoding rabies virus glycoproteins - An approach to the production of polyclonal antibody reagents. Mol Cell Probes 2022; 64:101833. [PMID: 35691598 DOI: 10.1016/j.mcp.2022.101833] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Abstract
DNA-based immunization has been previously shown to be an efficient approach to induce robust immunity against infectious diseases in animals and humans. The advantages of DNA vaccines are simplicity of their construction and production, low cost, high stability, and ability to elicit a full spectrum of immune responses to target antigens. The goals of this study were (i) to assess the antibody immune response to rabies virus glycoproteins (rGPs) in rabbits and guinea pigs after intramuscular immunization with pTargeT and pVAC2-mcs mammalian expression vectors encoding either the wild-type (WT) or codon-optimized (cOPT) rGP genes; and (ii) to prepare in-house rabbit anti-rGP polyclonal antibody reagents suitable for in Single Radial Immunodiffusion (SRID) and Indirect Fluorescent Antibody (IFA) assays. The maximum antibody responses against rabies virus in rabbits and guinea pigs were observed after immunization series with 500 μg/dose of pVAC2-mcs vector encoding either the WT or cOPT rGP genes adjuvanted with Emulsigen-D. No significant difference in the anti-rabies virus neutralizing antibody titers was observed in rabbits immunized with the WT and cOPT rGPs. The in-house rabbit anti-rGP polyclonal antibody reagents reacted comparable to the current reference reagents in SRID and IFA assays. The results of the study demonstrated that the DNA immunization of animals with the WT or cOPT rGPs is a promising approach to either induction of high anti-rabies virus neutralizing antibody titers in vivo or for production of polyclonal antibody reagents against rabies.
Collapse
Affiliation(s)
- Dmitriy V Volokhov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD, 20993, USA.
| | - Vyacheslav Furtak
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD, 20993, USA
| | - Cynthia Allen
- Vaccine Quality Division, Center for Biologics Evaluation (CBE), Biologic and Radiopharmaceutical Drugs Directorate (BRDD), Health Canada, 100 Eglantine Driveway, Ottawa, ON K1A 0K9, Canada
| | - Gayle Pulle
- Vaccine Quality Division, Center for Biologics Evaluation (CBE), Biologic and Radiopharmaceutical Drugs Directorate (BRDD), Health Canada, 100 Eglantine Driveway, Ottawa, ON K1A 0K9, Canada
| | - Michelle D Zajac
- Kansas State University, College of Veterinary Medicine, Department of Diagnostic Medicine and Pathobiology, 1800 Denison Ave., Manhattan, KS, 66506, USA
| | - Yotam Levin
- NanoPass Technologies Ltd., 3 Golda Meir St., Nes Ziona, 7403648, Israel
| | - Efrat Kochba
- NanoPass Technologies Ltd., 3 Golda Meir St., Nes Ziona, 7403648, Israel
| | - Susan M Moore
- The University of Missouri Veterinary Medical Diagnostic Laboratory (VMDL), One Health Laboratory, 1509 Rollins St., Columbia, MO, 65211, USA
| |
Collapse
|
11
|
Wang M, Liu J, Gao T, Xu L, Zhang X, Nie J, Li Y, Chen H. A platform method for plasmid isoforms analysis by capillary gel electrophoresis. Electrophoresis 2022; 43:1174-1182. [DOI: 10.1002/elps.202100343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/07/2022]
Affiliation(s)
- Meng Wang
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines Institute for Biological Product Control National Institutes for Food and Drug Control (NIFDC) NHC Key Laboratory of Research on Quality and Standardization of Biotech Products National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Biological Products Beijing P. R. China
| | - Jun‐Kai Liu
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines Institute for Biological Product Control National Institutes for Food and Drug Control (NIFDC) NHC Key Laboratory of Research on Quality and Standardization of Biotech Products National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Biological Products Beijing P. R. China
| | | | | | | | - Jian‐Hui Nie
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines Institute for Biological Product Control National Institutes for Food and Drug Control (NIFDC) NHC Key Laboratory of Research on Quality and Standardization of Biotech Products National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Biological Products Beijing P. R. China
| | - Yan Li
- National Medical Products Administration (NMPA) Key Laboratory for Quality Control and Evaluation of Vaccines and Biological Products SiChuan Institute for Drug Control Sichuan P. R. China
| | | |
Collapse
|
12
|
Mann JFS, McKay PF, Klein K, Pankrac J, Tregoning JS, Shattock RJ. Blocking T-cell egress with FTY720 extends DNA vaccine expression but reduces immunogenicity. Immunology 2022; 165:301-311. [PMID: 34775601 PMCID: PMC9426614 DOI: 10.1111/imm.13429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/01/2022] Open
Abstract
Optimal immunogenicity from nucleic acid vaccines requires a balance of antigen expression that effectively engages the host immune system without generating a cellular response that rapidly destroys cells producing the antigen and thereby limiting vaccine antigen expression. We investigated the role of the cellular response on the expression and antigenicity of DNA vaccines using a plasmid DNA construct expressing luciferase. Repeated intramuscular administration led to diminished luciferase expression, suggesting a role for immune-mediated clearance of expression. To investigate the role of cell trafficking, we used the sphingosine 1-phosphate receptor (S1PR) modulator, FTY720 (Fingolimod), which traps lymphocytes within the lymphoid tissues. When lymphocyte trafficking was blocked with FTY720, DNA transgene expression was maintained at a constant level for a significantly extended time period. Both continuous and staggered administration of FTY720 prolonged transgene expression. However, blocking lymphocyte egress during primary transgene administration did not result in an increase of transgene expression during secondary administration. Interestingly, there was a disconnect between transgene expression and immunogenicity, as increasing expression by this approach did not enhance the overall immune response. Furthermore, when FTY720 was administered alongside a DNA vaccine expressing the HIV gp140 envelope antigen, there was a significant reduction in both antigen-specific antibody and T-cell responses. This indicates that the developing antigen-specific cellular response clears DNA vaccine expression but requires access to the site of expression in order to develop an effective immune response.
Collapse
Affiliation(s)
- Jamie F. S. Mann
- Department of Infectious DiseasesImperial College LondonSt Mary’s CampusLondonUK
- Bristol Veterinary SchoolUniversity of BristolBristolUK
| | - Paul F. McKay
- Department of Infectious DiseasesImperial College LondonSt Mary’s CampusLondonUK
| | - Katja Klein
- Department of Infectious DiseasesImperial College LondonSt Mary’s CampusLondonUK
- School of Cellular and Molecular MedicineUniversity of BristolBristolUK
| | - Joshua Pankrac
- Department of Microbiology and ImmunologyUniversity of Western OntarioLondonOntarioCanada
| | - John S. Tregoning
- Department of Infectious DiseasesImperial College LondonSt Mary’s CampusLondonUK
| | - Robin J. Shattock
- Department of Infectious DiseasesImperial College LondonSt Mary’s CampusLondonUK
| |
Collapse
|
13
|
Naik R, Peden K. Regulatory Considerations on the Development of mRNA Vaccines. Curr Top Microbiol Immunol 2022; 440:187-205. [PMID: 32638114 DOI: 10.1007/82_2020_220] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Developing traditional viral vaccines for infectious diseases usually takes years, as these are usually produced either by chemical inactivation of the virus or attenuation of the pathogen, processes that can take considerable time to validate and also require the live pathogen. With the advent of nucleic-acid vaccines (DNA and mRNA), the time to vaccine design and production is considerably shortened, since once the platform has been established, all that is required is the sequence of the antigen gene, its synthesis and insertion into an appropriate expression vector; importantly, no infectious virus is required. mRNA vaccines have some advantages over DNA vaccines, such as expression in non-dividing cells and the absence of the perceived risk of integration into host genome. Also, generally lower doses are required to induce the immune response. Based on experience in recent clinical trials, mRNA-based vaccines are a promising novel platform that might be useful for the development of vaccines against emerging pandemic infectious diseases. This chapter discusses some of the specific issues that mRNA vaccines raise with respect to production, quality, safety and efficacy, and how they have been addressed so as to allow their evaluation in clinical trials.
Collapse
Affiliation(s)
- Ramachandra Naik
- Division of Vaccines and Related Products Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Building 71, Room 3045, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Keith Peden
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Building 52/72, Room 1220, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA.
| |
Collapse
|
14
|
Abdullahi IN, Emeribe AU, Adekola HA, Abubakar SD, Dangana A, Shuwa HA, Nwoba ST, Mustapha JO, Haruna MT, Olowookere KA, Animasaun OS, Ugwu CE, Onoja SO, Gadama AS, Mohammed M, Daneji IM, Amadu DO, Ghamba PE, Onukegbe NB, Shehu MS, Isomah C, Babayo A, Ahmad AEF. Leveraging on the genomics and immunopathology of SARS-CoV-2 for vaccines development: prospects and challenges. Hum Vaccin Immunother 2021; 17:620-637. [PMID: 32936732 PMCID: PMC7993231 DOI: 10.1080/21645515.2020.1812313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence and case-fatality rates (CFRs) of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection, the etiological agent for Coronavirus Disease 2019 (COVID-19), have been rising unabated. Even though the entire world has been implementing infection prevention and control measures, the pandemic continues to spread. It has been widely accepted that preventive vaccination strategies are the public health measures for countering this pandemic. This study critically reviews the latest scientific advancement in genomics, replication pattern, pathogenesis, and immunopathology of SARS-CoV-2 infection and how these concepts could be used in the development of vaccines. We also offer a detailed discussion on the anticipated potency, efficacy, safety, and pharmaco-economic issues that are and will be associated with candidate COVID-19 vaccines.
Collapse
Affiliation(s)
- Idris Nasir Abdullahi
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Anthony Uchenna Emeribe
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, University of Calabar, Calabar, Nigeria
| | | | - Sharafudeen Dahiru Abubakar
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Amos Dangana
- Department of Medical Laboratory Services, University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - Halima Ali Shuwa
- Lydia Becker Institute of Immunology, Manchester Collaborative Center for Inflammation Research, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | | | - Jelili Olaide Mustapha
- Biological Sciences Department, Faculty of Science, University of Alberta, Edmonton, Canada
| | | | - Kafayat Adepeju Olowookere
- Department of Medical Laboratory Services, Ladoke Akintola University of Technology Teaching Hospital, Ogbomoso, Nigeria
| | - Olawale Sunday Animasaun
- Nigeria Field Epidemiology and Laboratory Training Programme, African Field Epidemiology Network, Abuja, Nigeria
| | - Charles Egede Ugwu
- Department of Medical Laboratory Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Abdullahi Sani Gadama
- Department of Medical Microbiology and Parasitology, Faculty of Clinical Sciences, Bayero University, Kano, Nigeria
| | - Musa Mohammed
- Department of Medicine, Immunology Unit, Ahmadu Bello University, Zaria, Nigeria
| | - Isa Muhammad Daneji
- Department of Medical Microbiology and Parasitology, Faculty of Clinical Sciences, Bayero University, Kano, Nigeria
| | - Dele Ohinoyi Amadu
- Department of Medical Microbiology and Parasitology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Peter Elisha Ghamba
- WHO National Polio Reference Laboratory, University of Maiduguri Teaching Hospital, Maiduguri, Nigeria
| | | | - Muhammad Sagir Shehu
- Medical Laboratory Department, College of Health Technology, Ningi, Bauchi State, Nigeria
| | - Chiladi Isomah
- Medical Laboratory Science Department, Rivers State University, Port Harcourt, Nigeria
| | - Adamu Babayo
- Department of Medical Microbiology and Parasitology, Faculty of Clinical Sciences, Bayero University, Kano, Nigeria
| | - Abdurrahman El-Fulaty Ahmad
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
15
|
Affiliation(s)
- Rajesh Bhatia
- Former Director, Communicable Diseases, World Health Organization South-East Asia Regional Office, New Delhi 110 002, India
| |
Collapse
|
16
|
Tregoning JS, Brown ES, Cheeseman HM, Flight KE, Higham SL, Lemm N, Pierce BF, Stirling DC, Wang Z, Pollock KM. Vaccines for COVID-19. Clin Exp Immunol 2020; 202:162-192. [PMID: 32935331 PMCID: PMC7597597 DOI: 10.1111/cei.13517] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Since the emergence of COVID-19, caused by the SARS-CoV-2 virus at the end of 2019, there has been an explosion of vaccine development. By 24 September 2020, a staggering number of vaccines (more than 200) had started preclinical development, of which 43 had entered clinical trials, including some approaches that have not previously been licensed for human vaccines. Vaccines have been widely considered as part of the exit strategy to enable the return to previous patterns of working, schooling and socializing. Importantly, to effectively control the COVID-19 pandemic, production needs to be scaled-up from a small number of preclinical doses to enough filled vials to immunize the world's population, which requires close engagement with manufacturers and regulators. It will require a global effort to control the virus, necessitating equitable access for all countries to effective vaccines. This review explores the immune responses required to protect against SARS-CoV-2 and the potential for vaccine-induced immunopathology. We describe the profile of the different platforms and the advantages and disadvantages of each approach. The review also addresses the critical steps between promising preclinical leads and manufacturing at scale. The issues faced during this pandemic and the platforms being developed to address it will be invaluable for future outbreak control. Nine months after the outbreak began we are at a point where preclinical and early clinical data are being generated for the vaccines; an overview of this important area will help our understanding of the next phases.
Collapse
Affiliation(s)
- J. S. Tregoning
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - E. S. Brown
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - H. M. Cheeseman
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - K. E. Flight
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - S. L. Higham
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - N.‐M. Lemm
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - B. F. Pierce
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - D. C. Stirling
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - Z. Wang
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - K. M. Pollock
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| |
Collapse
|
17
|
Maeta M, Miura N, Tanaka H, Nakamura T, Kawanishi R, Nishikawa Y, Asano K, Tanaka M, Tamagawa S, Nakai Y, Tange K, Yoshioka H, Harashima H, Akita H. Vitamin E Scaffolds of pH-Responsive Lipid Nanoparticles as DNA Vaccines in Cancer and Protozoan Infection. Mol Pharm 2020; 17:1237-1247. [PMID: 32129629 DOI: 10.1021/acs.molpharmaceut.9b01262] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
DNA vaccinations are promising strategies for treating diseases that require cellular immunity (i.e., cancer and protozoan infection). Here, we report on the use of a liposomal nanocarrier (lipid nanoparticles (LNPs)) composed of an SS-cleavable and pH-activated lipidlike material (ssPalm) as an in vivo DNA vaccine. After subcutaneous administration, the LNPs containing an ssPalmE, an ssPalm with vitamin E scaffolds, elicited a higher gene expression activity in comparison with the other LNPs composed of the ssPalms with different hydrophobic scaffolds. Immunization with the ssPalmE-LNPs encapsulating plasmid DNA that encodes ovalbumin (OVA, a model tumor antigen) or profilin (TgPF, a potent antigen of Toxoplasma gondii) induced substantial antitumor or antiprotozoan effects, respectively. Flow cytometry analysis of the cells that had taken up the LNPs in draining lymph nodes (dLNs) showed that the ssPalmE-LNPs were largely taken up by macrophages and a small number of dendritic cells. We found that the transient deletion of CD169+ macrophages, a subpopulation of macrophages that play a key role in cancer immunity, unexpectedly enhanced the activity of the DNA vaccine. These data suggest that the ssPalmE-LNPs are effective DNA vaccine carriers, and a strategy for avoiding their being trapped by CD169+ macrophages will be a promising approach for developing next-generation DNA vaccines.
Collapse
Affiliation(s)
- Mio Maeta
- Department of Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo City, Hokkaido 060-0812, Japan
| | - Naoya Miura
- Department of Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo City, Hokkaido 060-0812, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-0856, Japan
| | - Takashi Nakamura
- Department of Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo City, Hokkaido 060-0812, Japan
| | - Ryo Kawanishi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro City, Hokkaido 080-8555, Japan
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro City, Hokkaido 080-8555, Japan
| | - Kenichi Asano
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji City, Tokyo 192-0392, Japan
| | - Masato Tanaka
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji City, Tokyo 192-0392, Japan
| | - Shinya Tamagawa
- DDS Research Laboratory, NOF CORPORATION, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Yuta Nakai
- DDS Research Laboratory, NOF CORPORATION, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Kota Tange
- DDS Research Laboratory, NOF CORPORATION, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Hiroki Yoshioka
- DDS Research Laboratory, NOF CORPORATION, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Hideyoshi Harashima
- Department of Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo City, Hokkaido 060-0812, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-0856, Japan
| |
Collapse
|
18
|
Safety Profile of a Multi-Antigenic DNA Vaccine Against Hepatitis C Virus. Vaccines (Basel) 2020; 8:vaccines8010053. [PMID: 32013228 PMCID: PMC7158683 DOI: 10.3390/vaccines8010053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 12/17/2022] Open
Abstract
Despite direct acting antivirals (DAAs) curing >95% of individuals infected with hepatitis C (HCV), in order to achieve the World Health Organization HCV Global Elimination Goals by 2030 there are still major challenges that need to be overcome. DAAs alone are unlikely to eliminate HCV in the absence of a vaccine that can limit viral transmission. Consequently, a prophylactic HCV vaccine is necessary to relieve the worldwide burden of HCV disease. DNA vaccines are a promising vaccine platform due to their commercial viability and ability to elicit robust T-cell-mediated immunity (CMI). We have developed a novel cytolytic DNA vaccine that encodes non-structural HCV proteins and a truncated mouse perforin (PRF), which is more immunogenic than the respective canonical DNA vaccine lacking PRF. Initially we assessed the ability of the HCV pNS3-PRF and pNS4/5-PRF DNA vaccines to elicit robust long-term CMI without any adverse side-effects in mice. Interferon-γ (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) assay was used to evaluate CMI against NS3, NS4 and NS5B in a dose-dependent manner. This analysis showed a dose-dependent bell-curve of HCV-specific responses in vaccinated animals. We then thoroughly examined the effects associated with reactogenicity of cytolytic DNA vaccination with the multi-antigenic HCV DNA vaccine (pNS3/4/5B). Hematological, biochemical and histological studies were performed in male Sprague Dawley rats with a relative vaccine dose 10–20-fold higher than the proposed dose in Phase I clinical studies. The vaccine was well tolerated, and no toxicity was observed. Thus, the cytolytic multi-antigenic DNA vaccine is safe and elicits broad memory CMI.
Collapse
|
19
|
Kim NY, Son WR, Choi JY, Yu CH, Hur GH, Jeong ST, Shin YK, Hong SY, Shin S. Immunogenicity and Biodistribution of Anthrax DNA Vaccine Delivered by Intradermal Electroporation. Curr Drug Deliv 2020; 17:414-421. [PMID: 32286944 DOI: 10.2174/1567201817666200414144550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/17/2020] [Accepted: 03/07/2020] [Indexed: 01/21/2023]
Abstract
PURPOSE Anthrax is a lethal bacterial disease caused by gram-positive bacterium Bacillus anthracis and vaccination is a desirable method to prevent anthrax infections. In the present study, DNA vaccine encoding a protective antigen of Bacillus anthracis was prepared and we investigated the influence of DNA electrotransfer in the skin on the induced immune response and biodistribution. METHODS AND RESULTS The tdTomato reporter gene for the whole animal in vivo imaging was used to assess gene transfer efficiency into the skin as a function of electrical parameters. Compared to that with 25 V, the transgene expression of red fluorescent protein increased significantly when a voltage of 90 V was used. Delivery of DNA vaccines expressing Bacillus anthracis protective antigen domain 4 (PAD4) with an applied voltage of 90 V induced robust PA-D4-specific antibody responses. In addition, the in vivo fate of anthrax DNA vaccine was studied after intradermal administration into the mouse. DNA plasmids remained at the skin injection site for an appropriate period of time after immunization. Intradermal administration of DNA vaccine resulted in detection in various organs (viz., lung, heart, kidney, spleen, brain, and liver), although the levels were significantly reduced. CONCLUSION Our results offer important insights into how anthrax DNA vaccine delivery by intradermal electroporation affects the immune response and biodistribution of DNA vaccine. Therefore, it may provide valuable information for the development of effective DNA vaccines against anthrax infection.
Collapse
Affiliation(s)
| | | | | | - Chi Ho Yu
- The 4th R & D Institute Directorate, Agency for Defense Development, Daejon, Korea
| | - Gyeung Haeng Hur
- The 4th R & D Institute Directorate, Agency for Defense Development, Daejon, Korea
| | - Seong Tae Jeong
- The 4th R & D Institute Directorate, Agency for Defense Development, Daejon, Korea
| | - Young Kee Shin
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Sung Youl Hong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Sungho Shin
- Bio-MAX/N-Bio, Seoul National University, Seoul, Korea
| |
Collapse
|
20
|
Rosa MT, Loreto ELS. The Catenulida flatworm can express genes from its microbiome or from the DNA it ingests. Sci Rep 2019; 9:19045. [PMID: 31836792 PMCID: PMC6910973 DOI: 10.1038/s41598-019-55659-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022] Open
Abstract
Stenostomum are tiny planarians of the phylum Platyhelminthes that reproduce asexually. We transfected these worms using plasmids containing a gfp reporter gene. Here we show that they can express genes present in plasmids carried by bacteria and those that are encoded by naked DNA, such as plasmids or PCR fragments, transfected by electroporation; they can also express genes taken up during feeding. The microbiome associated with worm maintenance was evaluated, and the results indicated that when a plasmid is maintained in the microbiome, gfp gene expression is stable. When genes originate from naked DNA or bacteria not maintained in the microbiome, GFP expression is transient. Therefore, changes in the microbiome can modify the ability of worms to express foreign genes. In stable GFP-expressing worms, NSG showed that the gfp gene was maintained in the plasmid and was not integrated into the chromosome. These results suggest that, at least for some organisms such as flatworms, the expression of genes provided by the microbiome or the environment can be considered among the potential sources of phenotypic plasticity, which can have implications for evolvability.
Collapse
Affiliation(s)
| | - Elgion L S Loreto
- Department of Biochemistry and Molecular Biology, CCNE, Univ. Fed. de Santa Maria, Av. Roraima 1000, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
21
|
Gallagher TB, Mellado-Sanchez G, Jorgensen AL, Moore S, Nataro JP, Pasetti MF, Baillie LW. Development of a multiple-antigen protein fusion vaccine candidate that confers protection against Bacillus anthracis and Yersinia pestis. PLoS Negl Trop Dis 2019; 13:e0007644. [PMID: 31430284 PMCID: PMC6716679 DOI: 10.1371/journal.pntd.0007644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 08/30/2019] [Accepted: 07/19/2019] [Indexed: 12/25/2022] Open
Abstract
Bacillus anthracis and Yersinia pestis are zoonotic bacteria capable of causing severe and sometimes fatal infections in animals and humans. Although considered as diseases of antiquity in industrialized countries due to animal and public health improvements, they remain endemic in vast regions of the world disproportionally affecting the poor. These pathogens also remain a serious threat if deployed in biological warfare. A single vaccine capable of stimulating rapid protection against both pathogens would be an extremely advantageous public health tool. We produced multiple-antigen fusion proteins (MaF1 and MaF2) containing protective regions from B. anthracis protective antigen (PA) and lethal factor (LF), and from Y. pestis V antigen (LcrV) and fraction 1 (F1) capsule. The MaF2 sequence was also expressed from a plasmid construct (pDNA-MaF2). Immunogenicity and protective efficacy were investigated in mice following homologous and heterologous prime-boost immunization. Antibody responses were determined by ELISA and anthrax toxin neutralization assay. Vaccine efficacy was determined against lethal challenge with either anthrax toxin or Y. pestis. Both constructs elicited LcrV and LF-specific serum IgG, and MaF2 elicited toxin-neutralizing antibodies. Immunizations with MaF2 conferred 100% and 88% protection against Y. pestis and anthrax toxin, respectively. In contrast, pDNA-MaF2 conferred only 63% protection against Y. pestis and no protection against anthrax toxin challenge. pDNA-MaF2-prime MaF2-boost induced 75% protection against Y. pestis and 25% protection against anthrax toxin. Protection was increased by the molecular adjuvant CARDif. In conclusion, MaF2 is a promising multi-antigen vaccine candidate against anthrax and plague that warrants further investigation. Anthrax and plague are ancient infectious diseases that continue to affect people living in poor, endemic regions and to threaten industrialized nations due to their potential use in biowarfare. Candidate vaccines need improvement to minimize non-desirable effects and increase their efficacy. The purpose of this work was to develop and evaluate a single subunit vaccine capable of conferring protection against Bacillus anthracis and Yersinia pestis. To this end, specific regions from their genome or key protective protein sequences from both microorganisms were combined to obtain either recombinant plasmids or recombinant proteins and tested as vaccine candidates in mice. The recombinant protein MaF2 induced specific antibody responses and afforded full and partial protection against Y. pestis and B. anthracis, respectively. Meanwhile, the DNA vaccine equivalent to MaF2 conferred only partial protection against Y. pestis, which increased when combined with an MaF2 protein boost. MaF2 emerged as a promising dual pathogen recombinant vaccine that warrants further investigation.
Collapse
Affiliation(s)
- Theresa B. Gallagher
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Gabriela Mellado-Sanchez
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Ana L. Jorgensen
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Stephen Moore
- BIOMET, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - James P. Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Box, Charlottesville, VA, United States of America
| | - Marcela F. Pasetti
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
- * E-mail: (MFP); (LWB)
| | - Les W. Baillie
- The Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, United Kingdom
- * E-mail: (MFP); (LWB)
| |
Collapse
|
22
|
Bernelin-Cottet C, Urien C, McCaffrey J, Collins D, Donadei A, McDaid D, Jakob V, Barnier-Quer C, Collin N, Bouguyon E, Bordet E, Barc C, Boulesteix O, Leplat JJ, Blanc F, Contreras V, Bertho N, Moore AC, Schwartz-Cornil I. Electroporation of a nanoparticle-associated DNA vaccine induces higher inflammation and immunity compared to its delivery with microneedle patches in pigs. J Control Release 2019; 308:14-28. [PMID: 31265882 DOI: 10.1016/j.jconrel.2019.06.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/18/2022]
Abstract
DNA vaccination is an attractive technology, based on its well-established manufacturing process, safety profile, adaptability to rapidly combat pandemic pathogens, and stability at ambient temperature; however an optimal delivery method of DNA remains to be determined. As pigs are a relevant model for humans, we comparatively evaluated the efficiency of vaccine DNA delivery in vivo to pigs using dissolvable microneedle patches, intradermal inoculation with needle (ID), surface electroporation (EP), with DNA associated or not to cationic poly-lactic-co-glycolic acid nanoparticles (NPs). We used a luciferase encoding plasmid (pLuc) as a reporter and vaccine plasmids encoding antigens from the Porcine Reproductive and Respiratory Syndrome Virus (PRRSV), a clinically-significant swine arterivirus. Patches were successful at inducing luciferase expression in skin although at lower level than EP. EP induced the cutaneaous recruitment of granulocytes, of MHC2posCD172Apos myeloid cells and type 1 conventional dendritic cells, in association with local production of IL-1β, IL-8 and IL-17; these local responses were more limited with ID and undetectable with patches. The addition of NP to EP especially promoted the recruitment of the MHC2posCD172Apos CD163int and CD163neg myeloid subsets. Notably we obtained the strongest and broadest IFNγ T-cell response against a panel of PRRSV antigens with DNA + NPs delivered by EP, whereas patches and ID were ineffective. The anti-PRRSV IgG responses were the highest with EP administration independently of NPs, mild with ID, and undetectable with patches. These results contrast with the immunogenicity and efficacy previously induced in mice with patches. This study concludes that successful DNA vaccine administration in skin can be achieved in pigs with electroporation and patches, but only the former induces local inflammation, humoral and cellular immunity, with the highest potency when NPs were used. This finding shows the importance of evaluating the delivery and immunogenicity of DNA vaccines beyond the mouse model in a preclinical model relevant to human such as pig and reveals that EP with DNA combined to NP induces strong immunogenicity.
Collapse
Affiliation(s)
| | - Céline Urien
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Joanne McCaffrey
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; Xeolas Pharmaceuticals Ltd., Dublin, Ireland
| | - Damien Collins
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; Xeolas Pharmaceuticals Ltd., Dublin, Ireland
| | - Agnese Donadei
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; Xeolas Pharmaceuticals Ltd., Dublin, Ireland
| | | | - Virginie Jakob
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Christophe Barnier-Quer
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Nicolas Collin
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Edwige Bouguyon
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Elise Bordet
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | | | | | - Jean-Jacques Leplat
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Fany Blanc
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Vanessa Contreras
- Immunology of viral infections and autoimmune diseases, IDMIT Department, IBFJ, INSERM U1184-CEA - Université Paris Sud 11, Fontenay-Aux-Roses et Le Kremlin-Bicêtre, France
| | - Nicolas Bertho
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France; BIOEPAR, Oniris, INRA, 44307 Nantes, France
| | - Anne C Moore
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | | |
Collapse
|
23
|
Bernelin-Cottet C, Urien C, Fretaud M, Langevin C, Trus I, Jouneau L, Blanc F, Leplat JJ, Barc C, Boulesteix O, Riou M, Dysart M, Mahé S, Studsrub E, Nauwynck H, Bertho N, Bourry O, Schwartz-Cornil I. A DNA Prime Immuno-Potentiates a Modified Live Vaccine against the Porcine Reproductive and Respiratory Syndrome Virus but Does Not Improve Heterologous Protection. Viruses 2019; 11:E576. [PMID: 31242645 PMCID: PMC6631340 DOI: 10.3390/v11060576] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV), an RNA virus inducing abortion in sows and respiratory disease in young pigs, is a leading infectious cause of economic losses in the swine industry. Modified live vaccines (MLVs) help in controlling the disease, but their efficacy is often compromised by the high genetic diversity of circulating viruses, leading to vaccine escape variants in the field. In this study, we hypothesized that a DNA prime with naked plasmids encoding PRRSV antigens containing conserved T-cell epitopes may improve the protection of MLV against a heterologous challenge. Plasmids were delivered with surface electroporation or needle-free jet injection and European strain-derived PRRSV antigens were targeted or not to the dendritic cell receptor XCR1. Compared to MLV-alone, the DNA-MLV prime- boost regimen slightly improved the IFNγ T-cell response, and substantially increased the antibody response against envelope motives and the nucleoprotein N. The XCR1-targeting of N significantly improved the anti-N specific antibody response. Despite this immuno-potentiation, the DNA-MLV regimen did not further decrease the serum viral load or the nasal viral shedding of the challenge strain over MLV-alone. Finally, the heterologous protection, achieved in absence of detectable effective neutralizing antibodies, was not correlated to the measured antibody or to the IFNγ T-cell response. Therefore, immune correlates of protection remain to be identified and represent an important gap of knowledge in PRRSV vaccinology. This study importantly shows that a naked DNA prime immuno-potentiates an MLV, more on the B than on the IFNγ T-cell response side, and has to be further improved to reach cross-protection.
Collapse
Affiliation(s)
- Cindy Bernelin-Cottet
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Céline Urien
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Maxence Fretaud
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Christelle Langevin
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
- VIM, EMERG'IN-Plateforme d'Infectiologie Expérimentale IERP, INRA, Domaine de Vilvert, 78352 Jouy-en-Josas, France.
| | - Ivan Trus
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| | - Luc Jouneau
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Fany Blanc
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Jean-Jacques Leplat
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Céline Barc
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, Centre Val de Loire, INRA, 37380 Nouzilly, France.
| | - Olivier Boulesteix
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, Centre Val de Loire, INRA, 37380 Nouzilly, France.
| | - Mickaël Riou
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, Centre Val de Loire, INRA, 37380 Nouzilly, France.
| | - Marilyn Dysart
- Pharmajet, 400 Corporate Circle Suite N, Golden, CO 80401, USA.
| | - Sophie Mahé
- Unité Virologie et Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, Anses, BP 53, 22440 Ploufragan, France.
| | | | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| | - Nicolas Bertho
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Olivier Bourry
- Unité Virologie et Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, Anses, BP 53, 22440 Ploufragan, France.
| | | |
Collapse
|
24
|
Bernelin-Cottet C, Urien C, Stubsrud E, Jakob V, Bouguyon E, Bordet E, Barc C, Boulesteix O, Contreras V, Barnier-Quer C, Collin N, Trus I, Nauwynck H, Bertho N, Schwartz-Cornil I. A DNA-Modified Live Vaccine Prime-Boost Strategy Broadens the T-Cell Response and Enhances the Antibody Response against the Porcine Reproductive and Respiratory Syndrome Virus. Viruses 2019; 11:E551. [PMID: 31207934 PMCID: PMC6630347 DOI: 10.3390/v11060551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023] Open
Abstract
The Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) induces reproductive disorders in sows and respiratory illnesses in growing pigs and is considered as one of the main pathogenic agents responsible for economic losses in the porcine industry worldwide. Modified live PRRSV vaccines (MLVs) are very effective vaccine types against homologous strains but they present only partial protection against heterologous viral variants. With the goal to induce broad and cross-protective immunity, we generated DNA vaccines encoding B and T antigens derived from a European subtype 1 strain that include T-cell epitope sequences known to be conserved across strains. These antigens were expressed either in a native form or in the form of vaccibodies targeted to the endocytic receptor XCR1 and CD11c expressed by different types of antigen-presenting cells (APCs). When delivered in skin with cationic nanoparticles and surface electroporation, multiple DNA vaccinations as a stand-alone regimen induced substantial antibody and T-cell responses, which were not promoted by targeting antigens to APCs. Interestingly, a DNA-MLV prime-boost strategy strongly enhanced the antibody response and broadened the T-cell responses over the one induced by MLV or DNA-only. The anti-nucleoprotein antibody response induced by the DNA-MLV prime-boost was clearly promoted by targeting the antigen to CD11c and XCR1, indicating a benefit of APC-targeting on the B-cell response. In conclusion, a DNA-MLV prime-boost strategy, by enhancing the potency and breadth of MLV vaccines, stands as a promising vaccine strategy to improve the control of PRRSV in infected herds.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Antibody Formation
- Immunity, Cellular
- Immunization Schedule
- Organisms, Genetically Modified/genetics
- Organisms, Genetically Modified/immunology
- Porcine Reproductive and Respiratory Syndrome/prevention & control
- Porcine respiratory and reproductive syndrome virus/genetics
- Porcine respiratory and reproductive syndrome virus/immunology
- Swine
- T-Lymphocytes/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Cindy Bernelin-Cottet
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Céline Urien
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | | | - Virginie Jakob
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Edwige Bouguyon
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Elise Bordet
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Céline Barc
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, INRA, 37380 Nouzilly, France.
| | - Olivier Boulesteix
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, INRA, 37380 Nouzilly, France.
| | - Vanessa Contreras
- Immunology of viral infections and autoimmune diseases, IDMIT Department, IBFJ, INSERM U1184-CEA-Université Paris Sud 11, 92260 Fontenay-Aux-Roses et 94270 Le Kremlin-Bicêtre, France.
| | - Christophe Barnier-Quer
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Ivan Trus
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| | - Nicolas Bertho
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | | |
Collapse
|
25
|
Chrun T, Lacôte S, Urien C, Richard CA, Tenbusch M, Aubrey N, Pulido C, Lakhdar L, Marianneau P, Schwartz-Cornil I. A DNA Vaccine Encoding the Gn Ectodomain of Rift Valley Fever Virus Protects Mice via a Humoral Response Decreased by DEC205 Targeting. Front Immunol 2019; 10:860. [PMID: 31105695 PMCID: PMC6494931 DOI: 10.3389/fimmu.2019.00860] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 04/03/2019] [Indexed: 12/13/2022] Open
Abstract
The Rift Valley fever virus (RVFV) is responsible for a serious mosquito-borne viral disease in humans and ruminants. The development of a new and safer vaccine is urgently needed due to the risk of introduction of this arbovirus into RVFV-free continents. We recently showed that a DNA vaccine encoding eGn, the ectodomain of the RVFV Gn glycoprotein, conferred a substantial protection in the sheep natural host and that the anti-eGn IgG levels correlated to protection. Addressing eGn to DEC205 reduced the protective efficacy while decreasing the antibody and increasing the IFNγ T cell responses in sheep. In order to get further insight into the involved mechanisms, we evaluated our eGn-encoding DNA vaccine strategy in the reference mouse species. A DNA vaccine encoding eGn induced full clinical protection in mice and the passive transfer of immune serum was protective. This further supports that antibodies, although non-neutralizing in vitro, are instrumental in the protection against RVFV. Addressing eGn to DEC205 was also detrimental to protection in mice, and in this species, both the antibody and the IFNγ T cell responses were strongly decreased. Conversely when using a plasmid encoding a different antigen, i.e., mCherry, DEC205 targeting promoted the antibody response. Altogether our results show that the outcome of targeting antigens to DEC205 depends on the species and on the fused antigen and is not favorable in the case of eGn. In addition, we bring evidences that eGn in itself is a pertinent antigen to be included in a DNA vaccine and that next developments should aim at promoting the anti-eGn antibody response.
Collapse
Affiliation(s)
- Tiphany Chrun
- VIM-INRA-Université Paris-Saclay, Jouy-en-Josas, France.,ANSES-Laboratoire de Lyon, Unité Virologie, Lyon, France
| | - Sandra Lacôte
- ANSES-Laboratoire de Lyon, Unité Virologie, Lyon, France
| | - Céline Urien
- VIM-INRA-Université Paris-Saclay, Jouy-en-Josas, France
| | | | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Nicolas Aubrey
- ISP, INRA, Université de Tours, UMR 1282 Team BioMAP, Nouzilly, France
| | - Coralie Pulido
- ANSES-Laboratoire de Lyon, Plateforme d'Expérimentation Animale, Lyon, France
| | - Latifa Lakhdar
- ANSES-Laboratoire de Lyon, Plateforme d'Expérimentation Animale, Lyon, France
| | | | | |
Collapse
|
26
|
Collins C, Lorenzen N, Collet B. DNA vaccination for finfish aquaculture. FISH & SHELLFISH IMMUNOLOGY 2019; 85:106-125. [PMID: 30017931 DOI: 10.1016/j.fsi.2018.07.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 06/08/2023]
Abstract
In fish, DNA vaccines have been shown to give very high protection in experimental facilities against a number of viral diseases, particularly diseases caused by rhabdoviruses. However, their efficacy in generating protection against other families of fish viral pathogens is less clear. One DNA vaccine is currently in use commercially in fish farms in Canada and the commercialisation of another was authorised in Europe in 2017. The mechanism of action of DNA vaccines, including the role of the innate immune responses induced shortly after DNA vaccination in the activation of the adaptive immunity providing longer term specific protection, is still not fully understood. In Europe the procedure for the commercialisation of a veterinary DNA vaccine requires the resolution of certain concerns particularly about safety for the host vaccinated fish, the consumer and the environment. Relating to consumer acceptance and particularly environmental safety, a key question is whether a DNA vaccinated fish is considered a Genetically Modified Organism (GMO). In the present opinion paper these key aspects relating to the mechanisms of action, and to the development and the use of DNA vaccines in farmed fish are reviewed and discussed.
Collapse
Affiliation(s)
| | | | - Bertrand Collet
- Marine Scotland, Aberdeen, United Kingdom; Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique (INRA), Université Paris-Saclay, Jouy-en-Josas, France.
| |
Collapse
|
27
|
Tejeda-Mansir A, García-Rendón A, Guerrero-Germán P. Plasmid-DNA lipid and polymeric nanovaccines: a new strategic in vaccines development. Biotechnol Genet Eng Rev 2018; 35:46-68. [DOI: 10.1080/02648725.2018.1560552] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Armando Tejeda-Mansir
- Department of Scientific and Technological Research, University of Sonora, Sonora, México
| | | | | |
Collapse
|
28
|
Kim NY, Ahn HB, Yu CH, Song DH, Hur GH, Shin YK, Shin S. Intradermal immunization with botulinum neurotoxin serotype E DNA vaccine induces humoral and cellular immunity and protects against lethal toxin challenge. Hum Vaccin Immunother 2018; 15:412-419. [PMID: 30235058 DOI: 10.1080/21645515.2018.1526554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) produced by the spore-forming, gram-positive, anaerobic bacterium Clostridium botulinum are the most toxic substances known and cause botulism, flaccid paralysis, or death. Owing to their high lethality, BoNTs are classified as category A agents by the Centers for Disease Control (CDC). Currently, there are no vaccines available to protect against BoNTs, so the rapid development of a safe and effective vaccine is important. DNA-based vaccines have recently drawn great attention because they can be developed quickly and can be applied in mass vaccination strategies to prevent disease outbreaks. Here, we report on the immunogenic and protective efficacy of a DNA vaccine, encoding a 50-kDa carboxy-terminal fragment of the BoNT serotype E heavy chain, which is delivered via an intradermal route. This plasmid DNA vaccine induced robust humoral and cellular BoNT/E-specific immune responses and completely protected animals against lethal challenge with BoNT/E. These results not only indicate that DNA vaccines could be further developed as safe and effective candidates for vaccines against BoNTs but also suggest a possible approach for developing vaccines that protect against bio-threat toxins.
Collapse
Affiliation(s)
| | - Hye Bin Ahn
- b Graduate School of Convergence Science and Technology , Seoul National University , Seoul , Republic of Korea
| | - Chi Ho Yu
- c Agency for Defense Development , Daejon , Republic of Korea
| | - Dong Hyun Song
- c Agency for Defense Development , Daejon , Republic of Korea
| | | | - Young Kee Shin
- d College of Pharmacy , Seoul National University , Seoul , Republic of Korea
| | - Sungho Shin
- e Bio-MAX/N-Bio , Seoul National University , Seoul , Republic of Korea
| |
Collapse
|
29
|
Wang J, Jiang Y, Yang W, Shi C, Huang H, Sun H, Liu G, Wang C, Yang G, Cai Y. Vaccination with DNA encoding ES 43-kDa /45-kDa antigens significantly reduces Trichinella spiralis infection in mice. Res Vet Sci 2018; 120:4-10. [DOI: 10.1016/j.rvsc.2018.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/30/2018] [Accepted: 08/20/2018] [Indexed: 01/24/2023]
|
30
|
A Rift Valley fever virus Gn ectodomain-based DNA vaccine induces a partial protection not improved by APC targeting. NPJ Vaccines 2018; 3:14. [PMID: 29707242 PMCID: PMC5910381 DOI: 10.1038/s41541-018-0052-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/26/2018] [Accepted: 03/28/2018] [Indexed: 01/20/2023] Open
Abstract
Rift Valley fever virus, a phlebovirus endemic in Africa, causes serious diseases in ruminants and humans. Due to the high probability of new outbreaks and spread to other continents where competent vectors are present, vaccine development is an urgent priority as no licensed vaccines are available outside areas of endemicity. In this study, we evaluated in sheep the protective immunity induced by DNA vaccines encoding the extracellular portion of the Gn antigen which was either or not targeted to antigen-presenting cells. The DNA encoding untargeted antigen was the most potent at inducing IgG responses, although not neutralizing, and conferred a significant clinical and virological protection upon infectious challenge, superior to DNA vaccines encoding the targeted antigen. A statistical analysis of the challenge parameters supported that the anti-eGn IgG, rather than the T-cell response, was instrumental in protection. Altogether, this work shows that a DNA vaccine encoding the extracellular portion of the Gn antigen confers substantial—although incomplete—protective immunity in sheep, a natural host with high preclinical relevance, and provides some insights into key immune correlates useful for further vaccine improvements against the Rift Valley fever virus. A vaccine made from the genome of Rift Valley fever virus (RVFV) offers partial protection, but pieces of the puzzle are missing, say scientists. French and Spanish researchers, led by the French National Institute for Agricultural Research’s Isabelle Schwartz-Cornil, tested in sheep three slightly-differing vaccine candidates using RVFV genes. Such DNA vaccines are designed to generate proteins which a host’s immune system can use to arm itself against a genuine viral infection. Two of the candidates, designed to target cells that would present the viral proteins to the host’s immune system, provided some benefit to the vaccinated sheep. However, the third untargeted candidate, was the most efficient at protecting sheep, although not completely, and at boosting antibody levels despite not neutralizing the virus. These results provide hope for DNA vaccines against RVFV, and offer direction for future research effort.
Collapse
|
31
|
Abstract
Currently used vaccines have had major effects on eliminating common infections, largely by duplicating the immune responses induced by natural infections. Now vaccinology faces more complex problems, such as waning antibody, immunosenescence, evasion of immunity by the pathogen, deviation of immunity by the microbiome, induction of inhibitory responses, and complexity of the antigens required for protection. Fortunately, vaccine development is now incorporating knowledge from immunology, structural biology, systems biology and synthetic chemistry to meet these challenges. In addition, international organisations are developing new funding and licensing pathways for vaccines aimed at pathogens with epidemic potential that emerge from tropical areas.
Collapse
Affiliation(s)
| | - Marta V Pinto
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Manish Sadarangani
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver BC, Canada.
| | - Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
32
|
Abstract
In spite of current influenza vaccines being immunogenic, evolution of the influenza virus can reduce efficacy and so influenza remains a major threat to public health. One approach to improve influenza vaccines is to include adjuvants; substances that boost the immune response. Adjuvants are particularly beneficial for influenza vaccines administered during a pandemic when a rapid response is required or for use in patients with impaired immune responses, such as infants and the elderly. This review outlines the current use of adjuvants in human influenza vaccines, including what they are, why they are used and what is known of their mechanism of action. To date, six adjuvants have been used in licensed human vaccines: Alum, MF59, AS03, AF03, virosomes and heat labile enterotoxin (LT). In general these adjuvants are safe and well tolerated, but there have been some rare adverse events when adjuvanted vaccines are used at a population level that may discourage the inclusion of adjuvants in influenza vaccines, for example the association of LT with Bell's Palsy. Improved understanding about the mechanisms of the immune response to vaccination and infection has led to advances in adjuvant technology and we describe the experimental adjuvants that have been tested in clinical trials for influenza but have not yet progressed to licensure. Adjuvants alone are not sufficient to improve influenza vaccine efficacy because they do not address the underlying problem of mismatches between circulating virus and the vaccine. However, they may contribute to improved efficacy of next-generation influenza vaccines and will most likely play a role in the development of effective universal influenza vaccines, though what that role will be remains to be seen.
Collapse
Affiliation(s)
- John S Tregoning
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| | - Ryan F Russell
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| | - Ekaterina Kinnear
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| |
Collapse
|
33
|
Airway T cells protect against RSV infection in the absence of antibody. Mucosal Immunol 2018; 11:249-256. [PMID: 28537249 DOI: 10.1038/mi.2017.46] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 04/02/2017] [Indexed: 02/04/2023]
Abstract
Tissue resident memory T (Trm) cells act as sentinels and early responders to infection. Respiratory syncytial virus (RSV)-specific Trm cells have been detected in the lungs after human RSV infection, but whether they have a protective role is unknown. To dissect the protective function of Trm cells, BALB/c mice were infected with RSV; infected mice developed antigen-specific CD8+ Trm cells (CD103+/CD69+) in the lungs and airways. Intranasally transferring cells from the airways of previously infected animals to naïve animals reduced weight loss on infection in the recipient mice. Transfer of airway CD8 cells led to reduced disease and viral load and increased interferon-γ in the airways of recipient mice, while CD4 transfer reduced tumor necrosis factor-α in the airways. Because DNA vaccines induce a systemic T-cell response, we compared vaccination with infection for the effect of memory CD8 cells generated in different compartments. Intramuscular DNA immunization induced RSV-specific CD8 T cells, but they were immunopathogenic and not protective. Notably, there was a marked difference in the induction of Trm cells; infection but not immunization induced antigen-specific Trm cells in a range of tissues. These findings demonstrate a protective role for airway CD8 against RSV and support the need for vaccines to induce antigen-specific airway cells.
Collapse
|
34
|
Vogel AB, Lambert L, Kinnear E, Busse D, Erbar S, Reuter KC, Wicke L, Perkovic M, Beissert T, Haas H, Reece ST, Sahin U, Tregoning JS. Self-Amplifying RNA Vaccines Give Equivalent Protection against Influenza to mRNA Vaccines but at Much Lower Doses. Mol Ther 2017; 26:446-455. [PMID: 29275847 PMCID: PMC5835025 DOI: 10.1016/j.ymthe.2017.11.017] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022] Open
Abstract
New vaccine platforms are needed to address the time gap between pathogen emergence and vaccine licensure. RNA-based vaccines are an attractive candidate for this role: they are safe, are produced cell free, and can be rapidly generated in response to pathogen emergence. Two RNA vaccine platforms are available: synthetic mRNA molecules encoding only the antigen of interest and self-amplifying RNA (sa-RNA). sa-RNA is virally derived and encodes both the antigen of interest and proteins enabling RNA vaccine replication. Both platforms have been shown to induce an immune response, but it is not clear which approach is optimal. In the current studies, we compared synthetic mRNA and sa-RNA expressing influenza virus hemagglutinin. Both platforms were protective, but equivalent levels of protection were achieved using 1.25 μg sa-RNA compared to 80 μg mRNA (64-fold less material). Having determined that sa-RNA was more effective than mRNA, we tested hemagglutinin from three strains of influenza H1N1, H3N2 (X31), and B (Massachusetts) as sa-RNA vaccines, and all protected against challenge infection. When sa-RNA was combined in a trivalent formulation, it protected against sequential H1N1 and H3N2 challenges. From this we conclude that sa-RNA is a promising platform for vaccines against viral diseases.
Collapse
Affiliation(s)
- Annette B Vogel
- BioNTech RNA Pharmaceuticals GmbH, An der Goldgrube 12, 55131 Mainz, Germany.
| | - Laura Lambert
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London W2 1PG, UK
| | - Ekaterina Kinnear
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London W2 1PG, UK
| | - David Busse
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London W2 1PG, UK
| | - Stephanie Erbar
- BioNTech RNA Pharmaceuticals GmbH, An der Goldgrube 12, 55131 Mainz, Germany
| | | | - Lena Wicke
- BioNTech RNA Pharmaceuticals GmbH, An der Goldgrube 12, 55131 Mainz, Germany
| | | | - Tim Beissert
- TRON GmbH, Freiligrathstraße 12, 55131 Mainz, Germany
| | - Heinrich Haas
- BioNTech RNA Pharmaceuticals GmbH, An der Goldgrube 12, 55131 Mainz, Germany
| | - Stephen T Reece
- BioNTech RNA Pharmaceuticals GmbH, An der Goldgrube 12, 55131 Mainz, Germany
| | - Ugur Sahin
- BioNTech AG, An der Goldgrube 12, 55131 Mainz, Germany
| | - John S Tregoning
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London W2 1PG, UK.
| |
Collapse
|
35
|
Dong LL, Tang R, Zhai YJ, Malla T, Hu K. DNA vaccine expressing herpes simplex virus 1 glycoprotein C and D protects mice against herpes simplex keratitis. Int J Ophthalmol 2017; 10:1633-1639. [PMID: 29181304 DOI: 10.18240/ijo.2017.11.01] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/05/2017] [Indexed: 02/08/2023] Open
Abstract
AIM To investigate whether DNA vaccine encoding herpes simplex virus 1 (HSV-1) glycoprotein C (gC) and glycoprotein D (gD) will achieve better protective effect against herpes simplex keratitis (HSK) than DNA vaccine encoding gD alone. METHODS DNA vaccine expressing gD or gC combined gD (gD.gC) were constructed and carried by chitosan nanoparticle. The expression of fusion protein gD and gC were detected in DNA/nanoparticle transfected 293T cells by Western-blot. For immunization, mice were inoculated with DNA/nanoparticle for 3 times with 2wk interval, and two weeks after the final immunization, the specific immune responses and clinical degrees of primary HSK were evaluated. RESULTS Fusion protein gD.gC could be expressed successfully in cultured 293T cells. And, pRSC-gC.gD-IL21 DNA/chitosan nanoparticle could effectively elicit strongest humoral and cellular immune response in primary HSK mice evidenced by higher levels of specific neutralizing antibody and sIgA production, enhanced cytotoxicities of splenocytes and nature killer cells (NK), when compared with those of gD alone or mocked vaccine immunized mice. As a result, gC-based vaccine immunized mice showed least HSK disease. CONCLUSION gC-based DNA vaccine could effectively prevent the progress of primary HSK, suggesting that this DNA vaccine could be a promising vaccine for HSK treatment in the future.
Collapse
Affiliation(s)
- Li-Li Dong
- Department of Ophthalmology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China.,Department of Ophthalmology, Jiangsu Taizhou People's Hospital, Taizhou 225300, Jiangsu Province, China.,Medical School of Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Ru Tang
- Medical School of Southeast University, Nanjing 210009, Jiangsu Province, China.,Department of Ophthalmology, the People's Hospital of Danyang, Zhenjiang 212300, Jiangsu Province, China
| | - Yu-Jia Zhai
- Medical School of Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Tejsu Malla
- Medical School of Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China.,Nanjing Ning Yi Eye Center, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
36
|
Dutton JL, Woo WP, Chandra J, Xu Y, Li B, Finlayson N, Griffin P, Frazer IH. An escalating dose study to assess the safety, tolerability and immunogenicity of a Herpes Simplex Virus DNA vaccine, COR-1. Hum Vaccin Immunother 2017; 12:3079-3088. [PMID: 27580249 PMCID: PMC5215501 DOI: 10.1080/21645515.2016.1221872] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This paper describes a single site, open-label Phase I clinical trial evaluating the safety, tolerability and immunogenicity in healthy volunteers of a herpes simplex polynucleotide vaccine that has previously been shown to enhance immunogenicity and protect against lethal herpes simplex virus type 2 (HSV-2) challenge in mice. Five escalating doses of the vaccine, COR-1, were given by intradermal injection to HSV-1 and 2 seronegative healthy individuals. COR-1 was found to be safe and well-tolerated; the only vaccine-related adverse events were mild. While vaccine-induced antibody responses were not detectable, cell-mediated immune responses to HSV-specific peptide groups were identified in 19 of the 20 subjects who completed the study, and local inflammation at the immunisation site was observed. This study indicates COR-1 has potential to be used as a therapeutic vaccine for HSV-2 infection.
Collapse
Affiliation(s)
- Julie L Dutton
- a Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd) , Translational Research Institute , Woolloongabba , QLD , Australia.,b University of Queensland , Diamantina Institute, Translational Research Institute , Woolloongabba , QLD, Australia
| | - Wai-Ping Woo
- a Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd) , Translational Research Institute , Woolloongabba , QLD , Australia.,b University of Queensland , Diamantina Institute, Translational Research Institute , Woolloongabba , QLD, Australia
| | - Janin Chandra
- a Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd) , Translational Research Institute , Woolloongabba , QLD , Australia.,b University of Queensland , Diamantina Institute, Translational Research Institute , Woolloongabba , QLD, Australia
| | - Yan Xu
- a Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd) , Translational Research Institute , Woolloongabba , QLD , Australia.,b University of Queensland , Diamantina Institute, Translational Research Institute , Woolloongabba , QLD, Australia
| | - Bo Li
- a Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd) , Translational Research Institute , Woolloongabba , QLD , Australia.,b University of Queensland , Diamantina Institute, Translational Research Institute , Woolloongabba , QLD, Australia
| | - Neil Finlayson
- a Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd) , Translational Research Institute , Woolloongabba , QLD , Australia
| | - Paul Griffin
- c Q-Pharm Pty Ltd, Brisbane, Australia; Department of Medicine and Infectious Diseases, Mater Hospital and Mater Medical Research Institute, Brisbane, Australia; The University of Queensland , Brisbane , Australia
| | - Ian H Frazer
- a Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd) , Translational Research Institute , Woolloongabba , QLD , Australia.,b University of Queensland , Diamantina Institute, Translational Research Institute , Woolloongabba , QLD, Australia
| |
Collapse
|
37
|
Cloning and Expression of a Secretory form of Truncated ORF2 (aa 112-607) from Hepatitis E Virus in the pVAX1 Vector. Jundishapur J Microbiol 2017. [DOI: 10.5812/jjm.13543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
38
|
Versiani AF, Astigarraga RG, Rocha ESO, Barboza APM, Kroon EG, Rachid MA, Souza DG, Ladeira LO, Barbosa-Stancioli EF, Jorio A, Da Fonseca FG. Multi-walled carbon nanotubes functionalized with recombinant Dengue virus 3 envelope proteins induce significant and specific immune responses in mice. J Nanobiotechnology 2017; 15:26. [PMID: 28376812 PMCID: PMC5379608 DOI: 10.1186/s12951-017-0259-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/20/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Dengue is the most prevalent arthropod-borne viral disease in the world. In this article we present results on the development, characterization and immunogenic evaluation of an alternative vaccine candidate against Dengue. METHODS The MWNT-DENV3E nanoconjugate was developed by covalent functionalization of carboxylated multi-walled carbon nanotubes (MWNT) with recombinant dengue envelope (DENV3E) proteins. The recombinant antigens were bound to the MWNT using a diimide-activated amidation process and the immunogen was characterized by TEM, AFM and Raman Spectroscopy. Furthermore, the immunogenicity of this vaccine candidate was evaluated in a murine model. RESULTS Immunization with MWNT-DENV3E induced comparable IgG responses in relation to the immunization with non-conjugated proteins; however, the inoculation of the nanoconjugate into mice generated higher titers of neutralizing antibodies. Cell-mediated responses were also evaluated, and higher dengue-specific splenocyte proliferation was observed in cell cultures derived from mice immunized with MWNT-DENV3E when compared to animals immunized with the non-conjugated DENV3E. CONCLUSIONS Despite the recent licensure of the CYD-TDV dengue vaccine in some countries, results from the vaccine's phase III trial have cast doubts about its overall efficacy and global applicability. While questions about the effectiveness of the CYD-TDV vaccine still lingers, it is wise to keep at hand an array of vaccine candidates, including alternative non-classical approaches like the one presented here.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Antibody Formation
- Antigens, Viral/immunology
- Cell Proliferation
- Cytokines/immunology
- Dengue/immunology
- Dengue/prevention & control
- Dengue Vaccines/immunology
- Dengue Vaccines/therapeutic use
- Dengue Virus/immunology
- Female
- Immunity, Cellular
- Immunoglobulin G/blood
- Mice
- Mice, Inbred BALB C
- Microscopy, Atomic Force
- Microscopy, Electron, Transmission
- Nanoconjugates/chemistry
- Nanomedicine
- Nanotubes, Carbon/chemistry
- Recombinant Proteins/chemistry
- Recombinant Proteins/immunology
- Spectrum Analysis, Raman
- Spleen/cytology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/therapeutic use
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Alice F. Versiani
- Laboratory of Basic and Applied Virology, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Ruiz G. Astigarraga
- Laboratory of Basic and Applied Virology, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Eliseu S. O. Rocha
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Ana Paula M. Barboza
- Laboratório de Nanoscopia, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Erna G. Kroon
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Milene A. Rachid
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Daniele G. Souza
- Laboratory of Microorganism-Host Interaction, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Luiz O. Ladeira
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Edel F. Barbosa-Stancioli
- Laboratory of Basic and Applied Virology, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Ado Jorio
- Laboratório de Nanoscopia, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Flávio G. Da Fonseca
- Laboratory of Basic and Applied Virology, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| |
Collapse
|
39
|
Clinical Use of DNA Vaccines. HANDBOOK OF ELECTROPORATION 2017. [PMCID: PMC7153459 DOI: 10.1007/978-3-319-32886-7_106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Owing to their unique advantages in simplicity, safety, scalability, and possibility of repeated administrations, DNA vaccines represent an appealing and competitive immunization approach for a wide array of conditions, including but not limited to infectious diseases and cancer immunotherapy. Despite the exciting efficacy observed in preclinical studies, DNA vaccines have faced challenges in inducing strong immune responses in humans. This unexpected poor immunogenicity has severely hampered the translation of DNA vaccines from investigational medications to licensed products. To overcome this obstacle, tremendous efforts have been made to improve antigen expression and enhance immunogenicity. Among these endeavors, in vivo DNA electroporation (EP) has proved to be a breakthrough technology capable of mediating efficient DNA uptake and resulting in enhanced antigen expression and vaccine immunogenicity. EP-mediated DNA delivery has become one of the major platforms used in clinical trials to evaluate DNA vaccines in humans. In this chapter, in addition to EP delivery, other progress made in DNA vaccine development including plasmid optimization, antigen design, and immunologic adjuvants is also reviewed. Finally, the use of DNA vaccines in the context of clinical trials for infectious diseases and cancer immunotherapy is summarized. Specifically, the strategies that allow DNA vaccines to overcome antigenic diversity for viral infection and break immune tolerance for cancer therapy are explored. Based on the advantages of DNA vaccines and the immense progress, led by the electroporation-mediated vaccine delivery, DNA vaccines appear to have the potential to fundamentally transform the vaccine field, providing important benefits for preventing and curing diseases.
Collapse
|
40
|
Cockrell AS, Baric RS. An effective DNA vaccine platform for Middle East respiratory syndrome coronavirus. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:499. [PMID: 28149861 DOI: 10.21037/atm.2016.11.40] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Adam S Cockrell
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA; Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
41
|
Strategies to enhance immunogenicity of cDNA vaccine encoded antigens by modulation of antigen processing. Vaccine 2016; 34:5132-5140. [PMID: 27593157 DOI: 10.1016/j.vaccine.2016.08.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/21/2016] [Accepted: 08/12/2016] [Indexed: 11/20/2022]
Abstract
Most vaccines are based on protective humoral responses while for intracellular pathogens CD8(+) T cells are regularly needed to provide protection. However, poor processing efficiency of antigens is often a limiting factor in CD8(+) T cell priming, hampering vaccine efficacy. The multistage cDNA vaccine H56, encoding three secreted Mycobacterium tuberculosis antigens, was used to test a complete strategy to enhance vaccine' immunogenicity. Potential CD8(+) T cell epitopes in H56 were predicted using the NetMHC3.4/ANN program. Mice were immunized with H56 cDNA using dermal DNA tattoo immunization and epitope candidates were tested for recognition by responding CD8(+) T cells in ex vivo assays. Seven novel CD8(+) T cell epitopes were identified. H56 immunogenicity could be substantially enhanced by two strategies: (i) fusion of the H56 sequence to cDNA of proteins that modify intracellular antigen processing or provide CD4(+) T cell help, (ii) by substitution of the epitope's hydrophobic C-terminal flanking residues for polar glutamic acid, which facilitated their proteasome-mediated generation. We conclude that this whole strategy of in silico prediction of potential CD8(+) T cell epitopes in novel antigens, followed by fusion to sequences with immunogenicity-enhancing properties or modification of epitope flanking sequences to improve proteasome-mediated processing, may be exploited to design novel vaccines against emerging or 'hard to treat' intracellular pathogens.
Collapse
|
42
|
Lambert L, Kinnear E, McDonald JU, Grodeland G, Bogen B, Stubsrud E, Lindeberg MM, Fredriksen AB, Tregoning JS. DNA Vaccines Encoding Antigen Targeted to MHC Class II Induce Influenza-Specific CD8(+) T Cell Responses, Enabling Faster Resolution of Influenza Disease. Front Immunol 2016; 7:321. [PMID: 27602032 PMCID: PMC4993793 DOI: 10.3389/fimmu.2016.00321] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/10/2016] [Indexed: 01/14/2023] Open
Abstract
Current influenza vaccines are effective but imperfect, failing to cover against emerging strains of virus and requiring seasonal administration to protect against new strains. A key step to improving influenza vaccines is to improve our understanding of vaccine-induced protection. While it is clear that antibodies play a protective role, vaccine-induced CD8+ T cells can improve protection. To further explore the role of CD8+ T cells, we used a DNA vaccine that encodes antigen dimerized to an immune cell targeting module. Immunizing CB6F1 mice with the DNA vaccine in a heterologous prime-boost regime with the seasonal protein vaccine improved the resolution of influenza disease compared with protein alone. This improved disease resolution was dependent on CD8+ T cells. However, DNA vaccine regimes that induced CD8+ T cells alone were not protective and did not boost the protection provided by protein. The MHC-targeting module used was an anti-I-Ed single chain antibody specific to the BALB/c strain of mice. To test the role of MHC targeting, we compared the response between BALB/c, C57BL/6 mice, and an F1 cross of the two strains (CB6F1). BALB/c mice were protected, C57BL/6 were not, and the F1 had an intermediate phenotype; showing that the targeting of antigen is important in the response. Based on these findings, and in agreement with other studies using different vaccines, we conclude that, in addition to antibody, inducing a protective CD8 response is important in future influenza vaccines.
Collapse
Affiliation(s)
- Laura Lambert
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London , London , UK
| | - Ekaterina Kinnear
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London , London , UK
| | - Jacqueline U McDonald
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London , London , UK
| | - Gunnveig Grodeland
- K. G. Jebsen Centre for Influenza Vaccine Research, Institute of Clinical Medicine, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Bjarne Bogen
- K. G. Jebsen Centre for Influenza Vaccine Research, Institute of Clinical Medicine, Oslo University Hospital, University of Oslo, Oslo, Norway; Centre for Immune Regulation, Institute for Immunology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | | | | | | | - John S Tregoning
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St. Mary's Campus, Imperial College London , London , UK
| |
Collapse
|
43
|
Lee HY, Nyon MP, Strych U. Vaccine Development Against Middle East Respiratory Syndrome. CURRENT TROPICAL MEDICINE REPORTS 2016; 3:80-86. [PMID: 32226714 PMCID: PMC7099997 DOI: 10.1007/s40475-016-0084-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Various types of vaccines are under pre-clinical and clinical development to address the recent appearance of Middle East respiratory syndrome or MERS, an emerging infectious disease that has already caused over 600 deaths and remains a threat to world health. The causative agent for this respiratory disease is a member of the betacoronavirus genus, phylogenetically closely related to the SARS coronavirus that caused an international health emergency in 2002. With lessons learned from the outbreak of severe acute respiratory syndrome, and with undeniable technological advances, vaccine development against MERS was initially fast-paced and has produced several DNA and protein vaccine candidates with promising results during early pre-clinical testing. At least one vaccine candidate has even entered first-in-humans clinical trials now. With the number of MERS cases declining though and other infectious diseases attracting increased attention, the question remains, whether, similar to the situation after the SARS pandemic, vaccine development is halted or remains the priority it rightfully should.
Collapse
Affiliation(s)
- Hai Yen Lee
- Tropical Infectious Diseases Research and Education Centre (TIDREC), University of Malaya, Kuala Lumpur, Malaysia
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Houston, TX USA
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX USA
| | - Mun Peak Nyon
- Tropical Infectious Diseases Research and Education Centre (TIDREC), University of Malaya, Kuala Lumpur, Malaysia
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Houston, TX USA
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX USA
| | - Ulrich Strych
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Houston, TX USA
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
44
|
Badamchi-Zadeh A, McKay PF, Korber BT, Barinaga G, Walters AA, Nunes A, Gomes JP, Follmann F, Tregoning JS, Shattock RJ. A Multi-Component Prime-Boost Vaccination Regimen with a Consensus MOMP Antigen Enhances Chlamydia trachomatis Clearance. Front Immunol 2016; 7:162. [PMID: 27199987 PMCID: PMC4848310 DOI: 10.3389/fimmu.2016.00162] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/15/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND A vaccine for Chlamydia trachomatis is of urgent medical need. We explored bioinformatic approaches to generate an immunogen against C. trachomatis that would induce cross-serovar T-cell responses as (i) CD4(+) T cells have been shown in animal models and human studies to be important in chlamydial protection and (ii) antibody responses may be restrictive and serovar specific. METHODS A consensus antigen based on over 1,500 major outer membrane protein (MOMP) sequences provided high epitope coverage against the most prevalent C. trachomatis strains in silico. Having designed the T-cell immunogen, we assessed it for immunogenicity in prime-boost regimens. This consensus MOMP transgene was delivered using plasmid DNA, Human Adenovirus 5 (HuAd5) or modified vaccinia Ankara (MVA) vectors with or without MF59(®) adjuvanted recombinant MOMP protein. RESULTS Different regimens induced distinct immune profiles. The DNA-HuAd5-MVA-Protein vaccine regimen induced a cellular response with a Th1-biased serum antibody response, alongside high serum and vaginal MOMP-specific antibodies. This regimen significantly enhanced clearance against intravaginal C. trachomatis serovar D infection in both BALB/c and B6C3F1 mouse strains. This enhanced clearance was shown to be CD4(+) T-cell dependent. Future studies will need to confirm the specificity and precise mechanisms of protection. CONCLUSION A C. trachomatis vaccine needs to induce a robust cellular response with broad cross-serovar coverage and a heterologous prime-boost regimen may be an approach to achieve this.
Collapse
Affiliation(s)
| | - Paul F McKay
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Bette T Korber
- Los Alamos National Laboratory, Theoretical Division , Los Alamos, NM , USA
| | - Guillermo Barinaga
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Adam A Walters
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Alexandra Nunes
- Department of Infectious Diseases, National Institute of Health , Lisbon , Portugal
| | - João Paulo Gomes
- Department of Infectious Diseases, National Institute of Health , Lisbon , Portugal
| | - Frank Follmann
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institute , Copenhagen , Denmark
| | - John S Tregoning
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Robin J Shattock
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| |
Collapse
|
45
|
Hu K, Malla T, Zhai Y, Dong L, Tang R. Topical Administration Is a Promising Inoculating Route versus Intramuscular Inoculation for the Nanoparticle-Carried DNA Vaccine to Prevent Corneal Infections. Ophthalmic Res 2015; 55:99-110. [DOI: 10.1159/000441898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/20/2015] [Indexed: 11/19/2022]
|
46
|
Kinnear E, Caproni LJ, Tregoning JS. A Comparison of Red Fluorescent Proteins to Model DNA Vaccine Expression by Whole Animal In Vivo Imaging. PLoS One 2015; 10:e0130375. [PMID: 26091084 PMCID: PMC4475043 DOI: 10.1371/journal.pone.0130375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/19/2015] [Indexed: 11/19/2022] Open
Abstract
DNA vaccines can be manufactured cheaply, easily and rapidly and have performed well in pre-clinical animal studies. However, clinical trials have so far been disappointing, failing to evoke a strong immune response, possibly due to poor antigen expression. To improve antigen expression, improved technology to monitor DNA vaccine transfection efficiency is required. In the current study, we compared plasmid encoded tdTomato, mCherry, Katushka, tdKatushka2 and luciferase as reporter proteins for whole animal in vivo imaging. The intramuscular, subcutaneous and tattooing routes were compared and electroporation was used to enhance expression. We observed that overall, fluorescent proteins were not a good tool to assess expression from DNA plasmids, with a highly heterogeneous response between animals. Of the proteins used, intramuscular delivery of DNA encoding either tdTomato or luciferase gave the clearest signal, with some Katushka and tdKatushka2 signal observed. Subcutaneous delivery was weakly visible and nothing was observed following DNA tattooing. DNA encoding haemagglutinin was used to determine whether immune responses mirrored visible expression levels. A protective immune response against H1N1 influenza was induced by all routes, even after a single dose of DNA, though qualitative differences were observed, with tattooing leading to high antibody responses and subcutaneous DNA leading to high CD8 responses. We conclude that of the reporter proteins used, expression from DNA plasmids can best be assessed using tdTomato or luciferase. But, the disconnect between visible expression level and immunogenicity suggests that in vivo whole animal imaging of fluorescent proteins has limited utility for predicting DNA vaccine efficacy.
Collapse
Affiliation(s)
- Ekaterina Kinnear
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Lisa J. Caproni
- Touchlight Genetics Ltd., Leatherhead, Surrey, United Kingdom
| | - John S. Tregoning
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
| |
Collapse
|