1
|
Meshry N, Carneiro KMM. DNA as a promising biomaterial for bone regeneration and potential mechanisms of action. Acta Biomater 2025; 197:68-86. [PMID: 40090507 DOI: 10.1016/j.actbio.2025.03.024] [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: 11/29/2024] [Revised: 02/25/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
DNA nanotechnology has created new possibilities for the use of DNA in tissue regeneration - an important advance for DNA use beyond its paradigmatic role as the hereditary biomacromolecule. Biomaterials containing synthetic or natural DNA have been proposed for several applications including drug and gene delivery, and more recently, as osteoconductive biomaterials. This review provides an in-depth discussion of studies that have used DNA-based materials for biomineralization and/or bone repair, with expansion on the topic of DNA hydrogels specifically, and the advantages they offer for advancing the field of bone regeneration. Four mechanisms of action for the osteoconductive capabilities of DNA-based materials are discussed, and a proposed model for degradation of these materials and its link to their osteoconductive properties is later presented. Finally, the review considers current limitations of DNA-based materials and summarizes important aspects that need to be addressed for future application of DNA nanotechnology in tissue repair. STATEMENT OF SIGNIFICANCE: Herein we summarize the developing field of DNA-based materials for biomineralization and bone repair, with a focus on DNA hydrogels. We first provide a comprehensive review of different forms of DNA-based materials described thus far which have been shown to enhance bone repair and mineralization (namely DNA coatings, DNA-containing pastes, DNA nanostructures and DNA hydrogels). Next, we describe four different mechanisms by which DNA-based materials could be exerting their osteogenic effect. Then, we propose a novel model that links DNA degradation and osteoconductivity. Lastly, we suggest possible research directions to enhance DNA-based materials for future clinical application. The suggested mechanisms and the proposed model can guide future research to better understand how DNA functions as a mineral- and bone-promoting molecule.
Collapse
Affiliation(s)
- Nadeen Meshry
- Faculty of Dentistry, University of Toronto, Toronto, Canada, 124 Edward Street, Toronto, ON M5G 1G6, Canada
| | - Karina M M Carneiro
- Faculty of Dentistry, University of Toronto, Toronto, Canada, 124 Edward Street, Toronto, ON M5G 1G6, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Canada, 164 College St, Toronto, ON M5S 3G9, Canada.
| |
Collapse
|
2
|
Tursi NJ, Tiwari S, Bedanova N, Kannan T, Parzych E, Okba N, Liaw K, Sárközy A, Livingston C, Trullen MI, Gary EN, Vadovics M, Laenger N, Londregan J, Khan MS, Omo-Lamai S, Muramatsu H, Blatney K, Hojecki C, Machado V, Maricic I, Smith TRF, Humeau LM, Patel A, Kossenkov A, Brenner JS, Allman D, Krammer F, Pardi N, Weiner DB. Modulation of lipid nanoparticle-formulated plasmid DNA drives innate immune activation promoting adaptive immunity. Cell Rep Med 2025; 6:102035. [PMID: 40120578 PMCID: PMC12047470 DOI: 10.1016/j.xcrm.2025.102035] [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: 07/09/2024] [Revised: 11/20/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Nucleic acid vaccines have grown in importance over the past several years, with the development of new approaches remaining a focus. We describe a lipid nanoparticle-formulated DNA (DNA-LNP) formulation which induces robust innate and adaptive immunity with similar serological potency to mRNA-LNPs and adjuvanted protein. Using an influenza hemagglutinin (HA)-encoding construct, we show that priming with our HA DNA-LNP demonstrated stimulator of interferon genes (STING)-dependent upregulation and activation of migratory dendritic cell (DC) subpopulations. HA DNA-LNP induced superior antigen-specific CD8+ T cell responses relative to mRNA-LNPs or adjuvanted protein, with memory responses persisting beyond one year. In rabbits immunized with HA DNA-LNP, we observed immune responses comparable or superior to mRNA-LNPs at the same dose. In an additional model, a SARS-CoV-2 spike-encoding DNA-LNP elicited protective efficacy comparable to spike mRNA-LNPs. Our study identifies a platform-specific priming mechanism for DNA-LNPs divergent from mRNA-LNPs or adjuvanted protein, suggesting avenues for this approach in prophylactic and therapeutic vaccine development.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sachchidanand Tiwari
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Bedanova
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Toshitha Kannan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Elizabeth Parzych
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nisreen Okba
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kevin Liaw
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - András Sárközy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cory Livingston
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Maria Ibanez Trullen
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ebony N Gary
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Máté Vadovics
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Niklas Laenger
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Biology Department, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer Londregan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohammad Suhail Khan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Serena Omo-Lamai
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kerry Blatney
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Casey Hojecki
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Igor Maricic
- Inovio Pharmaceuticals, Plymouth Meeting, PA 19462, USA
| | | | | | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew Kossenkov
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Allman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Anurogo D, Chen CY, Lin CC, Pawitan JA, Qiu DW, Qiu JT. Codon optimized influenza H1 HA sequence but not CTLA-4 targeting of HA antigen to enhance the efficacy of DNA vaccines in an animal model. J Immunotoxicol 2024; 21:2400624. [PMID: 39319829 DOI: 10.1080/1547691x.2024.2400624] [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: 06/26/2024] [Revised: 06/05/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Infections caused by the influenza virus lead to both epidemic and pandemic outbreaks in humans and animals. Owing to their rapid production, safety, and stability, DNA vaccines represent a promising avenue for eliciting immunity and thwarting viral infections. While DNA vaccines have demonstrated substantial efficacy in murine models, their effectiveness in larger animals remains subdued. This limitation may be addressed by augmenting the immunogenicity of DNA-based vaccines. In the investigation here, protein expression was enhanced via codon optimization and then mouse cytotoxic T-lymphocyte antigen 4 (CTLA-4) was harnessed as a modulatory adjunct to bind directly to antigen-presenting cells. Further, the study evaluated the immunogenicity of two variants of the hemagglutinin (HA) antigen, i.e. the full-length and the C-terminal deletion versions. The study findings revealed that the codon-optimized HA gene (pcHA) led to increased protein synthesis, as evidenced by elevated mRNA levels. Codon optimization also significantly bolstered both cellular and humoral immune responses. In cytokine assays, all plasmid constructs, particularly pCTLA4-cHA, induced robust interferon (IFN)-γ production, while interleukin (IL)-4 levels remained uniformly non-significant. Mice immunized with pcHA displayed an augmented presence of IFNγ+ T-cells, underscoring the enhanced potency of the codon-optimized HA vaccine. Contrarily, CTLA-4-fused DNA vaccines did not significantly amplify the immune response.
Collapse
MESH Headings
- Animals
- Vaccines, DNA/immunology
- Vaccines, DNA/genetics
- Mice
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Codon/genetics
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Humans
- Female
- Mice, Inbred BALB C
- Disease Models, Animal
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza A Virus, H1N1 Subtype/immunology
Collapse
Affiliation(s)
- Dito Anurogo
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Makassar, Makassar City, Indonesia
| | - Chia-Yuan Chen
- Department of Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Chu-Chi Lin
- Department of Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Universitas Indonesia, Jakarta, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Universitas Indonesia, Jakarta, Indonesia
- Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Daniel W Qiu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - J Timothy Qiu
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
4
|
Pavlin M, Škorja Milić N, Kandušer M, Pirkmajer S. Importance of the electrophoresis and pulse energy for siRNA-mediated gene silencing by electroporation in differentiated primary human myotubes. Biomed Eng Online 2024; 23:47. [PMID: 38750477 PMCID: PMC11097476 DOI: 10.1186/s12938-024-01239-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Electrotransfection is based on application of high-voltage pulses that transiently increase membrane permeability, which enables delivery of DNA and RNA in vitro and in vivo. Its advantage in applications such as gene therapy and vaccination is that it does not use viral vectors. Skeletal muscles are among the most commonly used target tissues. While siRNA delivery into undifferentiated myoblasts is very efficient, electrotransfection of siRNA into differentiated myotubes presents a challenge. Our aim was to develop efficient protocol for electroporation-based siRNA delivery in cultured primary human myotubes and to identify crucial mechanisms and parameters that would enable faster optimization of electrotransfection in various cell lines. RESULTS We established optimal electroporation parameters for efficient siRNA delivery in cultured myotubes and achieved efficient knock-down of HIF-1α while preserving cells viability. The results show that electropermeabilization is a crucial step for siRNA electrotransfection in myotubes. Decrease in viability was observed for higher electric energy of the pulses, conversely lower pulse energy enabled higher electrotransfection silencing yield. Experimental data together with the theoretical analysis demonstrate that siRNA electrotransfer is a complex process where electropermeabilization, electrophoresis, siRNA translocation, and viability are all functions of pulsing parameters. However, despite this complexity, we demonstrated that pulse parameters for efficient delivery of small molecule such as PI, can be used as a starting point for optimization of electroporation parameters for siRNA delivery into cells in vitro if viability is preserved. CONCLUSIONS The optimized experimental protocol provides the basis for application of electrotransfer for silencing of various target genes in cultured human myotubes and more broadly for electrotransfection of various primary cell and cell lines. Together with the theoretical analysis our data offer new insights into mechanisms that underlie electroporation-based delivery of short RNA molecules, which can aid to faster optimisation of the pulse parameters in vitro and in vivo.
Collapse
Affiliation(s)
- Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000, Ljubljana, Slovenia.
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia.
| | - Nives Škorja Milić
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia
| | - Maša Kandušer
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
- Pharmacy Institute, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia.
| |
Collapse
|
5
|
Patel A. Shaping immunity against infectious diseases with multivalent DNA vaccines. VACCINE INSIGHTS 2024; 3:29-33. [PMID: 38694840 PMCID: PMC11062630 DOI: 10.18609/vac.2024.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2024]
Abstract
Immunization has dramatically transformed human and animal health. Since its earliest days, vaccination has served as a fundamental strategy for infectious disease prevention, providing population-level coverage for childhood diseases and seasonal infections, and serving as a rapid response to pandemic pathogens. Yet, there is continued circulation of endemic, emerging, and reemerging pathogens for which there are no licensed prophylactic measures. The successes of nucleic acid technologies during the COVID-19 pandemic, exemplified in the first two licensed mRNA vaccines [1] and the first DNA vaccine receiving emergency use authorization for human use [2], are reinvigorating vaccine development to tackle this urgent unmet need. The inherent stability of DNA offers advantageous features such as thermostability and extended shelf life. These characteristics are pivotal for transport and storage in resource-constrained environments, like low and middle-income countries. Furthermore, the ability to encode large transgenes and well-established modular assembly pipelines are key attributes of DNA-based platforms. This versatility extends to combination strategies of individual DNA vaccines as a multivalent drug product. Multivalent synthetic DNA vaccines are therefore emerging as part of the exciting nucleic acid-based vaccine landscape as a strategy to induce robust and durable immunity in diverse global populations.
Collapse
Affiliation(s)
- Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA, 19104
| |
Collapse
|
6
|
Tandel N, Patel D, Thakkar M, Shah J, Tyagi RK, Dalai SK. Poly(I:C) and R848 ligands show better adjuvanticity to induce B and T cell responses against the antigen(s). Heliyon 2024; 10:e26887. [PMID: 38455541 PMCID: PMC10918150 DOI: 10.1016/j.heliyon.2024.e26887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Poly(I:C) and R848, synthetic ligands that activate Toll-like receptor 3 (TLR3) and TLR7/8 respectively, have been well-established for their ability to stimulate the immune system and induce antigen-specific immune responses. These ligands are capable of inducing the production of cytokines and chemokines, and hence support the activation and differentiation of B and T cells. We saw the long-lasting and perdurable immune responses by these adjuvants essentially required for an efficacious subunit vaccine. In this study, we investigated the potential of poly(I:C) and R848 to elicit B and T cell responses to the OVA antigen. We assessed the stimulatory effects of these ligands on the immune system, their impact on B and T cell activation, and their ability to enhanced generation of B and T cells. Collectively, our findings contribute to the understanding how poly(I:C) and R848 can be utilized as an adjuvant system to enhance immune responses to protein-based subunit vaccines. In the end, this work provides insights for the development of novel vaccination strategies and improving the vaccine efficacy. Present work shall help formulate newer strategies for subunit vaccines to address the infectious diseases.
Collapse
Affiliation(s)
- Nikunj Tandel
- Institute of Science, Nirma University, SG highway, Ahmedabad, Gujarat, India
| | - Digna Patel
- Institute of Science, Nirma University, SG highway, Ahmedabad, Gujarat, India
| | - Mansi Thakkar
- Institute of Science, Nirma University, SG highway, Ahmedabad, Gujarat, India
| | - Jagrut Shah
- Institute of Science, Nirma University, SG highway, Ahmedabad, Gujarat, India
| | - Rajeev K. Tyagi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
| | - Sarat K. Dalai
- Institute of Science, Nirma University, SG highway, Ahmedabad, Gujarat, India
| |
Collapse
|
7
|
Kozak M, Hu J. DNA Vaccines: Their Formulations, Engineering and Delivery. Vaccines (Basel) 2024; 12:71. [PMID: 38250884 PMCID: PMC10820593 DOI: 10.3390/vaccines12010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
The concept of DNA vaccination was introduced in the early 1990s. Since then, advancements in the augmentation of the immunogenicity of DNA vaccines have brought this technology to the market, especially in veterinary medicine, to prevent many diseases. Along with the successful COVID mRNA vaccines, the first DNA vaccine for human use, the Indian ZyCovD vaccine against SARS-CoV-2, was approved in 2021. In the current review, we first give an overview of the DNA vaccine focusing on the science, including adjuvants and delivery methods. We then cover some of the emerging science in the field of DNA vaccines, notably efforts to optimize delivery systems, better engineer delivery apparatuses, identify optimal delivery sites, personalize cancer immunotherapy through DNA vaccination, enhance adjuvant science through gene adjuvants, enhance off-target and heritable immunity through epigenetic modification, and predict epitopes with bioinformatic approaches. We also discuss the major limitations of DNA vaccines and we aim to address many theoretical concerns.
Collapse
Affiliation(s)
- Michael Kozak
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
- The Department of Pathology and Laboratory Medicine, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Jiafen Hu
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
- The Department of Pathology and Laboratory Medicine, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
8
|
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: 13] [Impact Index Per Article: 6.5] [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
|
9
|
Borgo GM, Rutishauser RL. Generating and measuring effective vaccine-elicited HIV-specific CD8 + T cell responses. Curr Opin HIV AIDS 2023; 18:331-341. [PMID: 37751362 PMCID: PMC10552829 DOI: 10.1097/coh.0000000000000824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW There is growing consensus that eliciting CD8 + T cells in addition to antibodies may be required for an effective HIV vaccine for both prevention and cure. Here, we review key qualities of vaccine-elicited CD8 + T cells as well as major CD8 + T cell-based delivery platforms used in recent HIV vaccine clinical trials. RECENT FINDINGS Much progress has been made in improving HIV immunogen design and delivery platforms to optimize CD8 + T cell responses. With regards to viral vectors, recent trials have tested newer chimp and human adenovirus vectors as well as a CMV vector. DNA vaccine immunogenicity has been increased by delivering the vaccines by electroporation and together with adjuvants as well as administering them as part of a heterologous regimen. In preclinical models, self-amplifying RNA vaccines can generate durable tissue-based CD8 + T cells. While it may be beneficial for HIV vaccines to recapitulate the functional and phenotypic features of HIV-specific CD8 + T cells isolated from elite controllers, most of these features are not routinely measured in HIV vaccine clinical trials. SUMMARY Identifying a vaccine capable of generating durable T cell responses that target mutationally vulnerable epitopes and that can rapidly intercept infecting or rebounding virus remains a challenge for HIV. Comprehensive assessment of HIV vaccine-elicited CD8 + T cells, as well as comparisons between different vaccine platforms, will be critical to advance our understanding of how to design better CD8 + T cell-based vaccines for HIV.
Collapse
Affiliation(s)
- Gina M Borgo
- Department of Medicine, University of California, San Francisco, California, USA
| | | |
Collapse
|
10
|
Kamensek U, Cemazar M, Kranjc Brezar S, Jesenko T, Kos S, Znidar K, Markelc B, Modic Z, Komel T, Gorse T, Rebersek E, Jakopic H, Sersa G. What We Learned about the Feasibility of Gene Electrotransfer for Vaccination on a Model of COVID-19 Vaccine. Pharmaceutics 2023; 15:1981. [PMID: 37514166 PMCID: PMC10385748 DOI: 10.3390/pharmaceutics15071981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
DNA vaccination is one of the emerging approaches for a wide range of applications, including prophylactic vaccination against infectious diseases and therapeutic vaccination against cancer. The aim of this study was to evaluate the feasibility of our previously optimized protocols for gene electrotransfer (GET)-mediated delivery of plasmid DNA into skin and muscle tissues on a model of COVID-19 vaccine. Plasmids encoding the SARS-CoV-2 proteins spike (S) and nucleocapsid (N) were used as the antigen source, and a plasmid encoding interleukin 12 (IL-12) was used as an adjuvant. Vaccination was performed in the skin or muscle tissue of C57BL/6J mice on days 0 and 14 (boost). Two weeks after the boost, blood, spleen, and transfected tissues were collected to determine the expression of S, N, IL-12, serum interferon-γ, the induction of antigen-specific IgG antibodies, and cytotoxic T-cells. In accordance with prior in vitro experiments that indicated problems with proper expression of the S protein, vaccination with S did not induce S-specific antibodies, whereas significant induction of N-specific antibodies was detected after vaccination with N. Intramuscular vaccination outperformed skin vaccination and resulted in significant induction of humoral and cell-mediated immunity. Moreover, both boost and adjuvant were found to be redundant for the induction of an immune response. Overall, the study confirmed the feasibility of the GET for DNA vaccination and provided valuable insights into this approach.
Collapse
Affiliation(s)
- Urska Kamensek
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Maja Cemazar
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia
| | | | - Tanja Jesenko
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia
| | - Spela Kos
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
| | - Katarina Znidar
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
| | - Bostjan Markelc
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena Pot 5, SI-1000 Ljubljana, Slovenia
| | - Ziva Modic
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia
| | - Tilen Komel
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia
| | - Tim Gorse
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Eva Rebersek
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Helena Jakopic
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Gregor Sersa
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena Pot 5, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
11
|
Coirada FC, Fernandes ER, Mello LRD, Schuch V, Soares Campos G, Braconi CT, Boscardin SB, Santoro Rosa D. Heterologous DNA Prime- Subunit Protein Boost with Chikungunya Virus E2 Induces Neutralizing Antibodies and Cellular-Mediated Immunity. Int J Mol Sci 2023; 24:10517. [PMID: 37445695 DOI: 10.3390/ijms241310517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Chikungunya virus (CHIKV) has become a significant public health concern due to the increasing number of outbreaks worldwide and the associated comorbidities. Despite substantial efforts, there is no specific treatment or licensed vaccine against CHIKV to date. The E2 glycoprotein of CHIKV is a promising vaccine candidate as it is a major target of neutralizing antibodies during infection. In this study, we evaluated the immunogenicity of two DNA vaccines (a non-targeted and a dendritic cell-targeted vaccine) encoding a consensus sequence of E2CHIKV and a recombinant protein (E2*CHIKV). Mice were immunized with different homologous and heterologous DNAprime-E2* protein boost strategies, and the specific humoral and cellular immune responses were accessed. We found that mice immunized with heterologous non-targeted DNA prime- E2*CHIKV protein boost developed high levels of neutralizing antibodies, as well as specific IFN-γ producing cells and polyfunctional CD4+ and CD8+ T cells. We also identified 14 potential epitopes along the E2CHIKV protein. Furthermore, immunization with recombinant E2*CHIKV combined with the adjuvant AS03 presented the highest humoral response with neutralizing capacity. Finally, we show that the heterologous prime-boost strategy with the non-targeted pVAX-E2 DNA vaccine as the prime followed by E2* protein + AS03 boost is a promising combination to elicit a broad humoral and cellular immune response. Together, our data highlights the importance of E2CHIKV for the development of a CHIKV vaccine.
Collapse
Affiliation(s)
- Fernanda Caroline Coirada
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Edgar Ruz Fernandes
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Lucas Rodrigues de Mello
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04044-020, Brazil
| | - Viviane Schuch
- Departamento de Análises Clínicas e Toxicológicas, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
| | - Gúbio Soares Campos
- Laboratório de Virologia, Universidade Federal da Bahia (UFBA), Salvador 40110-909, Brazil
| | - Carla Torres Braconi
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Silvia Beatriz Boscardin
- Departamento de Parasitologia, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| | - Daniela Santoro Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| |
Collapse
|
12
|
Martins M, do Nascimento GM, Conforti A, Noll JCG, Impellizeri JA, Sanchez E, Wagner B, Lione L, Salvatori E, Pinto E, Compagnone M, Viscount B, Hayward J, Shorrock C, Aurisicchio L, Diel DG. A linear SARS-CoV-2 DNA vaccine candidate reduces virus shedding in ferrets. Arch Virol 2023; 168:124. [PMID: 36988739 PMCID: PMC10052258 DOI: 10.1007/s00705-023-05746-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/03/2023] [Indexed: 03/30/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has caused more than 760 million cases and over 6.8 million deaths as of March 2023. Vaccination has been the main strategy used to contain the spread of the virus and to prevent hospitalizations and deaths. Currently, two mRNA-based vaccines and one adenovirus-vectored vaccine have been approved and are available for use in the U.S. population. The versatility, low cost, and rapid production of DNA vaccines provide important advantages over other platforms. Additionally, DNA vaccines efficiently induce both B- and T-cell responses by expressing the antigen within transfected host cells, and the antigen, after being processed into peptides, can associate with MHC class I or II of antigen-presenting cells (APCs) to stimulate different T cell responses. However, the efficiency of DNA vaccination needs to be improved for use in humans. Importantly, in vivo DNA delivery combined with electroporation (EP) has been used successfully in the field of veterinary oncology, resulting in high rates of response after electrochemotherapy. Here, we evaluate the safety, immunogenicity, and protective efficacy of a novel linear SARS-CoV-2 DNA vaccine candidate delivered by intramuscular injection followed by electroporation (Vet-ePorator™) in ferrets. The linear SARS-CoV-2 DNA vaccine candidate did not cause unexpected side effects. Additionally, the vaccine elicited neutralizing antibodies and T cell responses on day 42 post-immunization using a low dose of the linear DNA construct in a prime-boost regimen. Most importantly, vaccination significantly reduced shedding of infectious SARS-CoV-2 through oral and nasal secretions in a ferret model.
Collapse
Affiliation(s)
- Mathias Martins
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Gabriela M do Nascimento
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | - Jessica C G Noll
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | - Bettina Wagner
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | | | | | - Brian Viscount
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - James Hayward
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - Clay Shorrock
- Applied DNA Sciences, Inc., New York, NY, USA
- LineaRx, Inc. , New York, NY, USA
| | - Luigi Aurisicchio
- Takis Biotech, Rome, Italy
- Evvivax Biotech, Rome, Italy
- Neomatrix Biotech, Rome, Italy
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
13
|
Murwanti R, Denda-Nagai K, Sugiura D, Mogushi K, Gendler SJ, Irimura T. Prevention of Inflammation-Driven Colon Carcinogenesis in Human MUC1 Transgenic Mice by Vaccination with MUC1 DNA and Dendritic Cells. Cancers (Basel) 2023; 15:cancers15061920. [PMID: 36980805 PMCID: PMC10047104 DOI: 10.3390/cancers15061920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The preventive efficacy of MUC1-specific DNA immunization on inflammation-driven colon carcinogenesis in human MUC1 transgenic (MUC1.Tg) mice was investigated. Mice were vaccinated with MUC1 DNA mixed with autologous bone-marrow-derived dendritic cells (BMDCs), and then colonic tumors were induced by azoxymethane (AOM) injection and oral administration of dextran sulfate sodium (DSS). Two types of tumors, squamous metaplasia and tubular adenoma, were observed. Both expressed high levels of MUC1 as indicated by the binding of anti-MUC1 antibodies with different specificities, whereas MUC1 expression was not detected in normal colonic mucosa. When mice were immunized with MUC1 DNA + BMDCs, tumor incidence, tumor number, and tumor size were significantly reduced. In contrast, vaccination with MUC1 DNA alone or BMDCs alone was ineffective in reducing tumor burden. Inflammation caused by DSS was not suppressed by the MUC1 DNA + BMDCs vaccination. Furthermore, MUC1 protein expression levels, as judged by anti-MUC1 antibody binding in tumors grown after vaccination, did not significantly differ from the control. In conclusion, an inflammation-driven carcinogenesis model was established in MUC1.Tg mice, closely resembling human colon carcinogenesis. In this model, vaccination with MUC1 DNA + BMDCs was effective in overriding MUC1 tolerance and reducing the tumor burden by a mechanism not affecting the level of colonic inflammation.
Collapse
Affiliation(s)
- Retno Murwanti
- Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55283, Indonesia
| | - Kaori Denda-Nagai
- Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Daisuke Sugiura
- Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Kaoru Mogushi
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Sandra J Gendler
- Department of Immunology, Mayo Clinic Arizona, 13400 E. Shea Blvd., Scottsdale, AZ 85259, USA
| | - Tatsuro Irimura
- Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Division of Glycobiologics, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
14
|
Zhao G, Zhang Z, Ding Y, Hou J, Liu Y, Zhang M, Sui C, Wang L, Xu X, Gao X, Kou Z. A DNA Vaccine Encoding the Full-Length Spike Protein of Beta Variant (B.1.351) Elicited Broader Cross-Reactive Immune Responses against Other SARS-CoV-2 Variants. Vaccines (Basel) 2023; 11:513. [PMID: 36992097 PMCID: PMC10054764 DOI: 10.3390/vaccines11030513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The SARS-CoV-2 pandemic remains an ongoing threat to global health with emerging variants, especially the Omicron variant and its sub-lineages. Although large-scale vaccination worldwide has delivered outstanding achievements for COVID-19 prevention, a declining effectiveness to a different extent in emerging SARS-CoV-2 variants was observed in the vaccinated population. Vaccines eliciting broader spectrum neutralizing antibodies and cellular immune responses are urgently needed and important. To achieve this goal, rational vaccine design, including antigen modeling, screening and combination, vaccine pipelines, and delivery, are keys to developing a next-generation COVID-19 vaccine. In this study, we designed several DNA constructs based on codon-optimized spike coding regions of several SARS-CoV-2 variants and analyzed their cross-reactive antibodies, including neutralizing antibodies, and cellular immune responses against several VOCs in C57BL/6 mice. The results revealed that different SARS-CoV-2 VOCs induced different cross-reactivity; pBeta, a DNA vaccine encoding the spike protein of the Beta variant, elicited broader cross-reactive neutralizing antibodies against other variants including the Omicron variants BA.1 and BA.4/5. This result demonstrates that the spike antigen from the Beta variant potentially serves as one of the antigens for multivalent vaccine design and development against variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Gan Zhao
- Advaccine Biopharmaceutics (Suzhou) Co., Ltd., Suzhou 215000, China
| | | | | | | | | | | | | | | | | | | | - Zhihua Kou
- Advaccine Biopharmaceutics (Suzhou) Co., Ltd., Suzhou 215000, China
| |
Collapse
|
15
|
Hebel C, Thomsen AR. A survey of mechanisms underlying current and potential COVID-19 vaccines. APMIS 2023; 131:37-60. [PMID: 36394112 DOI: 10.1111/apm.13284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
The emergence of SARS-CoV-2 caught the world off guard resulting in a global health crisis. Even though COVID-19 have caused the death of millions of people and many countries are still battling waves of infections, the odds of the pandemic ending soon have turned significantly in our favor. The key has been the development and distribution of a broad range of vaccines in record time. In this survey, we summarize the immunology required to understand the mechanisms underlying current and potential COVID-19 vaccines. Furthermore, we provide an up to date (according to data from WHO May 27, 2022) overview of the vaccine landscape consisting of 11 approved vaccines in phase 4, and a pipeline consisting of 161 vaccine candidates in clinical development and 198 in preclinical development (World Health Organization, Draft landscape and tracker of COVID-19 candidate vaccines [Internet], WHO, 2022). Our focus is to provide an understanding of the underlying biological mode of action of different vaccine platform designs, their advantages and disadvantages, rather than a deep dive into safety and efficacy data. We further present arguments concerning why a broad range of vaccines are needed and discuss future challenges.
Collapse
Affiliation(s)
- Christian Hebel
- Department of Immunology and Microbiology, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
New vector and vaccine platforms: mRNA, DNA, viral vectors. Curr Opin HIV AIDS 2022; 17:338-344. [DOI: 10.1097/coh.0000000000000763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Kisakov DN, Kisakova LA, Borgoyakova MB, Starostina EV, Taranov OS, Ivleva EK, Pyankov OV, Zaykovskaya AV, Shcherbakov DN, Rudometov AP, Rudometova NB, Volkova NV, Gureev VN, Ilyichev AA, Karpenko LI. Optimization of In Vivo Electroporation Conditions and Delivery of DNA Vaccine Encoding SARS-CoV-2 RBD Using the Determined Protocol. Pharmaceutics 2022; 14:pharmaceutics14112259. [PMID: 36365078 PMCID: PMC9693113 DOI: 10.3390/pharmaceutics14112259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 12/02/2022] Open
Abstract
Vaccination against SARS-CoV-2 and other viral infections requires safe, effective, and inexpensive vaccines that can be rapidly developed. DNA vaccines are candidates that meet these criteria, but one of their drawbacks is their relatively weak immunogenicity. Electroporation (EP) is an effective way to enhance the immunogenicity of DNA vaccines, but because of the different configurations of the devices that are used for EP, it is necessary to carefully select the conditions of the procedure, including characteristics such as voltage, current strength, number of pulses, etc. In this study, we determined the optimal parameters for delivery DNA vaccine by electroporation using the BEX CO device. BALB/c mice were used as a model. Plasmid DNA phMGFP was intramuscular (I/M) injected into the quadriceps muscle of the left hind leg of animals using insulin syringes, followed by EP. As a result of the experiments, the following EP parameters were determined: direct and reverse polarity rectangular DC current in three pulses, 12 V voltage for 30 ms and 950 ms intervals, with a current limit of 45 mA. The selected protocol induced a low level of injury and provided a high level of GFP expression. The chosen protocol was used to evaluate the immunogenicity of the DNA vaccine encoding the receptor-binding domain (RBD) of the SARS-CoV-2 protein (pVAXrbd) injected by EP. It was shown that the delivery of pVAXrbd via EP significantly enhanced both specific humoral and cellular immune responses compared to the intramuscular injection of the DNA vaccine.
Collapse
|
18
|
Li Z, Liu S, Li F, Li Y, Li Y, Peng P, Li S, He L, Liu T. Efficacy, immunogenicity and safety of COVID-19 vaccines in older adults: a systematic review and meta-analysis. Front Immunol 2022; 13:965971. [PMID: 36177017 PMCID: PMC9513208 DOI: 10.3389/fimmu.2022.965971] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/24/2022] [Indexed: 01/08/2023] Open
Abstract
BackgroundOlder adults are more susceptible to severe health outcomes for coronavirus disease 2019 (COVID-19). Universal vaccination has become a trend, but there are still doubts and research gaps regarding the COVID-19 vaccination in the elderly. This study aimed to investigate the efficacy, immunogenicity, and safety of COVID-19 vaccines in older people aged ≥ 55 years and their influencing factors.MethodsRandomized controlled trials from inception to April 9, 2022, were systematically searched in PubMed, EMBASE, the Cochrane Library, and Web of Science. We estimated summary relative risk (RR), rates, or standardized mean difference (SMD) with 95% confidence interval (CI) using random-effects meta-analysis. This study was registered with PROSPERO (CRD42022314456).ResultsOf the 32 eligible studies, 9, 21, and 25 were analyzed for efficacy, immunogenicity, and safety, respectively. In older adults, vaccination was efficacious against COVID-19 (79.49%, 95% CI: 60.55−89.34), with excellent seroconversion rate (92.64%, 95% CI: 86.77−96.91) and geometric mean titer (GMT) (SMD 3.56, 95% CI: 2.80−4.31) of neutralizing antibodies, and provided a significant protection rate against severe disease (87.01%, 50.80−96.57). Subgroup and meta-regression analyses consistently found vaccine types and the number of doses to be primary influencing factors for efficacy and immunogenicity. Specifically, mRNA vaccines showed the best efficacy (90.72%, 95% CI: 86.82−93.46), consistent with its highest seroconversion rate (98.52%, 95% CI: 93.45−99.98) and GMT (SMD 6.20, 95% CI: 2.02−10.39). Compared to the control groups, vaccination significantly increased the incidence of total adverse events (AEs) (RR 1.59, 95% CI: 1.38−1.83), including most local and systemic AEs, such as pain, fever, chill, etc. For inactivated and DNA vaccines, the incidence of any AEs was similar between vaccination and control groups (p > 0.1), while mRNA vaccines had the highest risk of most AEs (RR range from 1.74 to 7.22).ConclusionCOVID-19 vaccines showed acceptable efficacy, immunogenicity and safety in older people, especially providing a high protection rate against severe disease. The mRNA vaccine was the most efficacious, but it is worth surveillance for some AEs it caused. Increased booster coverage in older adults is warranted, and additional studies are urgently required for longer follow-up periods and variant strains.
Collapse
Affiliation(s)
- Zejun Li
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shouhuan Liu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fengming Li
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Yifeng Li
- College of Pediatrics, Chongqing Medical University, Chongqing, China
| | - Yilin Li
- College of Pediatrics, Chongqing Medical University, Chongqing, China
| | - Pu Peng
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Sai Li
- College of Pediatrics, Chongqing Medical University, Chongqing, China
| | - Li He
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Tieqiao Liu, ; Li He,
| | - Tieqiao Liu
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Tieqiao Liu, ; Li He,
| |
Collapse
|
19
|
Mokhtary P, Pourhashem Z, Mehrizi AA, Sala C, Rappuoli R. Recent Progress in the Discovery and Development of Monoclonal Antibodies against Viral Infections. Biomedicines 2022; 10:biomedicines10081861. [PMID: 36009408 PMCID: PMC9405509 DOI: 10.3390/biomedicines10081861] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 01/09/2023] Open
Abstract
Monoclonal antibodies (mAbs), the new revolutionary class of medications, are fast becoming tools against various diseases thanks to a unique structure and function that allow them to bind highly specific targets or receptors. These specialized proteins can be produced in large quantities via the hybridoma technique introduced in 1975 or by means of modern technologies. Additional methods have been developed to generate mAbs with new biological properties such as humanized, chimeric, or murine. The inclusion of mAbs in therapeutic regimens is a major medical advance and will hopefully lead to significant improvements in infectious disease management. Since the first therapeutic mAb, muromonab-CD3, was approved by the U.S. Food and Drug Administration (FDA) in 1986, the list of approved mAbs and their clinical indications and applications have been proliferating. New technologies have been developed to modify the structure of mAbs, thereby increasing efficacy and improving delivery routes. Gene delivery technologies, such as non-viral synthetic plasmid DNA and messenger RNA vectors (DMabs or mRNA-encoded mAbs), built to express tailored mAb genes, might help overcome some of the challenges of mAb therapy, including production restrictions, cold-chain storage, transportation requirements, and expensive manufacturing and distribution processes. This paper reviews some of the recent developments in mAb discovery against viral infections and illustrates how mAbs can help to combat viral diseases and outbreaks.
Collapse
Affiliation(s)
- Pardis Mokhtary
- Monoclonal Antibody Discovery Laboratory, Fondazione Toscana Life Sciences, 53100 Siena, Italy;
- Department of Biochemistry and Molecular Biology, University of Siena, 53100 Siena, Italy
| | - Zeinab Pourhashem
- Student Research Committee, Pasteur Institute of Iran, Tehran 1316943551, Iran;
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Akram Abouei Mehrizi
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Claudia Sala
- Monoclonal Antibody Discovery Laboratory, Fondazione Toscana Life Sciences, 53100 Siena, Italy;
- Correspondence: (C.S.); (R.R.)
| | - Rino Rappuoli
- Monoclonal Antibody Discovery Laboratory, Fondazione Toscana Life Sciences, 53100 Siena, Italy;
- Correspondence: (C.S.); (R.R.)
| |
Collapse
|
20
|
Perspective Technologies of Vaccination: Do We Still Need Old Vaccines? Vaccines (Basel) 2022; 10:vaccines10060891. [PMID: 35746498 PMCID: PMC9230923 DOI: 10.3390/vaccines10060891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/19/2022] [Indexed: 12/10/2022] Open
|
21
|
Abstract
Infectious diseases are a leading cause of death worldwide, and vaccines are the cheapest and efficient approach to preventing diseases. Use of conventional vaccination strategies such as live, attenuated, and subunit has limitations as it does not fully provide protection against many infectious diseases. Hence, there was a need for the development of a new vaccination strategy. Use of nucleic acids-DNA and RNA-has emerged as promising alternative to conventional vaccine approaches. Knowledge of mRNA biology, chemistry, and delivery systems in recent years have enabled mRNA to become a promising vaccine candidate. One of the advantages of a mRNA vaccine is that clinical batches can be generated after the availability of a sequence encoding the immunogen. The process is cell-free and scalable. mRNA is a noninfectious, nonintegrating molecule and there is no potential risk of infection or mutagenesis. mRNA is degraded by normal cellular processes, and its in vivo half-life can be regulated by different modifications and delivery methods. The efficacy can be increased by modifications of the nucleosides that can make mRNA more stable and highly translatable. Efficient in vivo delivery can be achieved by formulating mRNA into carrier molecules, allowing rapid uptake and expression in the cytoplasm. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in late 2019 and spread globally, prompting an international effort to accelerate development of a vaccine. The spike (S) glycoprotein mediates host cell attachment and is required for viral entry; it is the primary vaccine target for many candidate SARS-CoV-2 vaccines. Development of a lipid nanoparticle encapsulated mRNA vaccine that encodes the SARS-CoV-2 S glycoprotein stabilized in its prefusion conformation conferred 95% protection against Covid-19.
Collapse
Affiliation(s)
- Sunil Thomas
- Lankenau Institute for Medical Research, Wynnewood, PA, USA.
| | - Ann Abraham
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| |
Collapse
|
22
|
Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine Delivery Approaches and Advanced Delivery Systems for the Prevention of Viral Infections: From Development to Clinical Application. Pharmaceutics 2021; 13:2091. [PMID: 34959372 PMCID: PMC8707864 DOI: 10.3390/pharmaceutics13122091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections causing pandemics and chronic diseases are the main culprits implicated in devastating global clinical and socioeconomic impacts, as clearly manifested during the current COVID-19 pandemic. Immunoprophylaxis via mass immunisation with vaccines has been shown to be an efficient strategy to control such viral infections, with the successful and recently accelerated development of different types of vaccines, thanks to the advanced biotechnological techniques involved in the upstream and downstream processing of these products. However, there is still much work to be done for the improvement of efficacy and safety when it comes to the choice of delivery systems, formulations, dosage form and route of administration, which are not only crucial for immunisation effectiveness, but also for vaccine stability, dose frequency, patient convenience and logistics for mass immunisation. In this review, we discuss the main vaccine delivery systems and associated challenges, as well as the recent success in developing nanomaterials-based and advanced delivery systems to tackle these challenges. Manufacturing and regulatory requirements for the development of these systems for successful clinical and marketing authorisation were also considered. Here, we comprehensively review nanovaccines from development to clinical application, which will be relevant to vaccine developers, regulators, and clinicians.
Collapse
Affiliation(s)
- Ana Sara Cordeiro
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Yogita Patil-Sen
- Wrightington, Wigan and Leigh Teaching Hospitals NHS Foundation Trust, National Health Service, Wigan WN6 0SZ, UK;
| | - Maitreyi Shivkumar
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Ronak Patel
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Abdulwahhab Khedr
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A. Elsawy
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| |
Collapse
|
23
|
Creytens S, Pascha MN, Ballegeer M, Saelens X, de Haan CAM. Influenza Neuraminidase Characteristics and Potential as a Vaccine Target. Front Immunol 2021; 12:786617. [PMID: 34868073 PMCID: PMC8635103 DOI: 10.3389/fimmu.2021.786617] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/29/2021] [Indexed: 12/28/2022] Open
Abstract
Neuraminidase of influenza A and B viruses plays a critical role in the virus life cycle and is an important target of the host immune system. Here, we highlight the current understanding of influenza neuraminidase structure, function, antigenicity, immunogenicity, and immune protective potential. Neuraminidase inhibiting antibodies have been recognized as correlates of protection against disease caused by natural or experimental influenza A virus infection in humans. In the past years, we have witnessed an increasing interest in the use of influenza neuraminidase to improve the protective potential of currently used influenza vaccines. A number of well-characterized influenza neuraminidase-specific monoclonal antibodies have been described recently, most of which can protect in experimental challenge models by inhibiting the neuraminidase activity or by Fc receptor-dependent mechanisms. The relative instability of the neuraminidase poses a challenge for protein-based antigen design. We critically review the different solutions that have been proposed to solve this problem, ranging from the inclusion of stabilizing heterologous tetramerizing zippers to the introduction of inter-protomer stabilizing mutations. Computationally engineered neuraminidase antigens have been generated that offer broad, within subtype protection in animal challenge models. We also provide an overview of modern vaccine technology platforms that are compatible with the induction of robust neuraminidase-specific immune responses. In the near future, we will likely see the implementation of influenza vaccines that confront the influenza virus with a double punch: targeting both the hemagglutinin and the neuraminidase.
Collapse
MESH Headings
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antigenic Drift and Shift
- Antigens, Viral/immunology
- Antigens, Viral/ultrastructure
- Catalytic Domain/genetics
- Catalytic Domain/immunology
- Cross Protection
- Evolution, Molecular
- Humans
- Immunogenicity, Vaccine
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Alphainfluenzavirus/enzymology
- Alphainfluenzavirus/genetics
- Alphainfluenzavirus/immunology
- Betainfluenzavirus/enzymology
- Betainfluenzavirus/genetics
- Betainfluenzavirus/immunology
- Mutation
- Nanoparticles
- Neuraminidase/administration & dosage
- Neuraminidase/genetics
- Neuraminidase/immunology
- Neuraminidase/ultrastructure
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/ultrastructure
- Viral Proteins/administration & dosage
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Proteins/ultrastructure
Collapse
Affiliation(s)
- Sarah Creytens
- Vlaams Instituut voor Biotechnologie (VIB)-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Mirte N. Pascha
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Marlies Ballegeer
- Vlaams Instituut voor Biotechnologie (VIB)-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- Vlaams Instituut voor Biotechnologie (VIB)-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Cornelis A. M. de Haan
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
24
|
Pagant S, Liberatore RA. In Vivo Electroporation of Plasmid DNA: A Promising Strategy for Rapid, Inexpensive, and Flexible Delivery of Anti-Viral Monoclonal Antibodies. Pharmaceutics 2021; 13:1882. [PMID: 34834297 PMCID: PMC8618954 DOI: 10.3390/pharmaceutics13111882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Since the first approval of monoclonal antibodies by the United States Food and Drug Administration (FDA) in 1986, therapeutic antibodies have become one of the predominant classes of drugs in oncology and immunology. Despite their natural function in contributing to antiviral immunity, antibodies as drugs have only more recently been thought of as tools for combating infectious diseases. Passive immunization, or the delivery of the products of an immune response, offers near-immediate protection, unlike the active immune processes triggered by traditional vaccines, which rely on the time it takes for the host's immune system to develop an effective defense. This rapid onset of protection is particularly well suited to containing outbreaks of emerging viral diseases. Despite these positive attributes, the high cost associated with antibody manufacture and the need for a cold chain for storage and transport limit their deployment on a global scale, especially in areas with limited resources. The in vivo transfer of nucleic acid-based technologies encoding optimized therapeutic antibodies transform the body into a bioreactor for rapid and sustained production of biologics and hold great promise for circumventing the obstacles faced by the traditional delivery of antibodies. In this review, we provide an overview of the different antibody delivery strategies that are currently being developed, with particular emphasis on in vivo transfection of naked plasmid DNA facilitated by electroporation.
Collapse
|
25
|
Reeder SM, Bah MA, Tursi NJ, Brooks RC, Patel A, Esquivel R, Eaton A, Jhun H, Chu J, Kim K, Xu Z, Zavala F, Weiner DB. Strategic Variants of CSP Delivered as SynDNA Vaccines Demonstrate Heterogeneity of Immunogenicity and Protection from Plasmodium Infection in a Murine Model. Infect Immun 2021; 89:e0072820. [PMID: 34152830 PMCID: PMC8445182 DOI: 10.1128/iai.00728-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/27/2021] [Indexed: 11/20/2022] Open
Abstract
Malaria infects millions of people every year, and despite recent advances in controlling disease spread, such as vaccination, it remains a global health concern. The circumsporozoite protein (CSP) has long been acknowledged as a key target in antimalarial immunity. Leveraging the DNA vaccine platform against this formidable pathogen, the following five synthetic DNA vaccines encoding variations of CSP were designed and studied: 3D7, GPI1, ΔGPI, TM, and DD2. Among the single CSP antigen constructs, a range of immunogenicity was observed with ΔGPI generating the most robust immunity. In an intravenous (i.v.) sporozoite challenge, the best protection among vaccinated mice was achieved by ΔGPI, which performed almost as well as the monoclonal antibody 311 (MAb 311) antibody control. Further analyses revealed that ΔGPI develops high-molecular-weight multimers in addition to monomeric CSP. We then compared the immunity generated by ΔGPI versus synDNA mimics for the antimalaria vaccines RTS,S and R21. The anti-CSP antibody responses induced were similar among these three immunogens. T cell responses demonstrated that ΔGPI induced a more focused anti-CSP response. In an infectious mosquito challenge, all three of these constructs generated inhibition of liver-stage infection as well as immunity from blood-stage parasitemia. This study demonstrates that synDNA mimics of complex malaria immunogens can provide substantial protection as can a novel synDNA vaccine ΔGPI.
Collapse
Affiliation(s)
- Sophia M. Reeder
- The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Mamadou A. Bah
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Nicholas J. Tursi
- The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Rebekah C. Brooks
- The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Ami Patel
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Rianne Esquivel
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Alison Eaton
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Hugo Jhun
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jacqueline Chu
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kevin Kim
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Ziyang Xu
- The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - David B. Weiner
- The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Vaccine Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Alamri SS, Alluhaybi KA, Alhabbab RY, Basabrain M, Algaissi A, Almahboub S, Alfaleh MA, Abujamel TS, Abdulaal WH, ElAssouli MZ, Alharbi RH, Hassanain M, Hashem AM. Synthetic SARS-CoV-2 Spike-Based DNA Vaccine Elicits Robust and Long-Lasting Th1 Humoral and Cellular Immunity in Mice. Front Microbiol 2021; 12:727455. [PMID: 34557174 PMCID: PMC8454412 DOI: 10.3389/fmicb.2021.727455] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/30/2021] [Indexed: 12/30/2022] Open
Abstract
The ongoing global pandemic of coronavirus disease 2019 (COVID-19) calls for an urgent development of effective and safe prophylactic and therapeutic measures. The spike (S) glycoprotein of severe acute respiratory syndrome-coronavirus (SARS-CoV-2) is a major immunogenic and protective protein and plays a crucial role in viral pathogenesis. In this study, we successfully constructed a synthetic codon-optimized DNA-based vaccine as a countermeasure against SARS-CoV-2, denoted VIU-1005. The design was based on a codon-optimized coding sequence of a consensus full-length S glycoprotein. The immunogenicity of the vaccine was tested in two mouse models (BALB/c and C57BL/6J). Th1-skewed systemic S-specific IgG antibodies and neutralizing antibodies (nAbs) were significantly induced in both models 4 weeks after three injections with 100 μg of the VIU-1005 vaccine via intramuscular needle injection but not intradermal or subcutaneous routes. Such immunization induced long-lasting IgG and memory T cell responses in mice that lasted for at least 6 months. Interestingly, using a needle-free system, we showed an enhanced immunogenicity of VIU-1005 in which lower or fewer doses were able to elicit significantly high levels of Th1-biased systemic S-specific immune responses, as demonstrated by the significant levels of binding IgG antibodies, nAbs and IFN-γ, TNF and IL-2 cytokine production from memory CD8+ and CD4+ T cells in BALB/c mice. Furthermore, compared to intradermal needle injection, which failed to induce any significant immune response, intradermal needle-free immunization elicited a robust Th1-biased humoral response similar to that observed with intramuscular immunization. Together, our results demonstrate that the synthetic VIU-1005 candidate DNA vaccine is highly immunogenic and capable of inducing long-lasting Th1-skewed humoral and cellular immunity in mice. Furthermore, we show that the use of a needle-free system could enhance the immunogenicity and minimize doses needed to induce protective immunity in mice, supporting further preclinical and clinical testing of this candidate vaccine.
Collapse
Affiliation(s)
- Sawsan S. Alamri
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalid A. Alluhaybi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rowa Y. Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Basabrain
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia,Medical Research Center, Jazan University, Jazan, Saudi Arabia
| | - Sarah Almahboub
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,SaudiVax Ltd., Thuwal, Saudi Arabia
| | - Mohamed A. Alfaleh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki S. Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wesam H. Abdulaal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - M-Zaki ElAssouli
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rahaf H. Alharbi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mazen Hassanain
- SaudiVax Ltd., Thuwal, Saudi Arabia,Department of Surgery, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Anwar M. Hashem,
| |
Collapse
|
27
|
Naszályi Nagy L, Dhaene E, Van Zele M, Mihály J, Klébert S, Varga Z, Kövér KE, De Buysser K, Van Driessche I, Martins JC, Fehér K. Silica@zirconia Core@shell Nanoparticles for Nucleic Acid Building Block Sorption. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2166. [PMID: 34578482 PMCID: PMC8468278 DOI: 10.3390/nano11092166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 12/27/2022]
Abstract
The development of delivery systems for the immobilization of nucleic acid cargo molecules is of prime importance due to the need for safe administration of DNA or RNA type of antigens and adjuvants in vaccines. Nanoparticles (NP) in the size range of 20-200 nm have attractive properties as vaccine carriers because they achieve passive targeting of immune cells and can enhance the immune response of a weakly immunogenic antigen via their size. We prepared high capacity 50 nm diameter silica@zirconia NPs with monoclinic/cubic zirconia shell by a green, cheap and up-scalable sol-gel method. We studied the behavior of the particles upon water dialysis and found that the ageing of the zirconia shell is a major determinant of the colloidal stability after transfer into the water due to physisorption of the zirconia starting material on the surface. We determined the optimum conditions for adsorption of DNA building blocks, deoxynucleoside monophosphates (dNMP), the colloidal stability of the resulting NPs and its time dependence. The ligand adsorption was favored by acidic pH, while colloidal stability required neutral-alkaline pH; thus, the optimal pH for the preparation of nucleic acid-modified particles is between 7.0-7.5. The developed silica@zirconia NPs bind as high as 207 mg dNMPs on 1 g of nanocarrier at neutral-physiological pH while maintaining good colloidal stability. We studied the influence of biological buffers and found that while phosphate buffers decrease the loading dramatically, other commonly used buffers, such as HEPES, are compatible with the nanoplatform. We propose the prepared silica@zirconia NPs as promising carriers for nucleic acid-type drug cargos.
Collapse
Affiliation(s)
- Livia Naszályi Nagy
- NMR and Structure Analysis Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium; (L.N.N.); (J.C.M.)
| | - Evert Dhaene
- Sol-Gel Centre for Research on Inorganic Powders and Thin Films Synthesis, Department of Chemistry, Ghent University, Krijgslaan 281 S3, B-9000 Ghent, Belgium; (E.D.); (M.V.Z.); (K.D.B.); (I.V.D.)
| | - Matthias Van Zele
- Sol-Gel Centre for Research on Inorganic Powders and Thin Films Synthesis, Department of Chemistry, Ghent University, Krijgslaan 281 S3, B-9000 Ghent, Belgium; (E.D.); (M.V.Z.); (K.D.B.); (I.V.D.)
| | - Judith Mihály
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network (IMEC RCNS ELKH), Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; (J.M.); (S.K.); (Z.V.)
| | - Szilvia Klébert
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network (IMEC RCNS ELKH), Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; (J.M.); (S.K.); (Z.V.)
| | - Zoltán Varga
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network (IMEC RCNS ELKH), Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; (J.M.); (S.K.); (Z.V.)
| | - Katalin E. Kövér
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary;
| | - Klaartje De Buysser
- Sol-Gel Centre for Research on Inorganic Powders and Thin Films Synthesis, Department of Chemistry, Ghent University, Krijgslaan 281 S3, B-9000 Ghent, Belgium; (E.D.); (M.V.Z.); (K.D.B.); (I.V.D.)
| | - Isabel Van Driessche
- Sol-Gel Centre for Research on Inorganic Powders and Thin Films Synthesis, Department of Chemistry, Ghent University, Krijgslaan 281 S3, B-9000 Ghent, Belgium; (E.D.); (M.V.Z.); (K.D.B.); (I.V.D.)
| | - José C. Martins
- NMR and Structure Analysis Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium; (L.N.N.); (J.C.M.)
| | - Krisztina Fehér
- Molecular Recognition and Interaction Research Group, Hungarian Academy of Sciences-Eötvös Loránd Research Network at University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| |
Collapse
|
28
|
Sánchez-Arribas N, Martínez-Negro M, Aicart-Ramos C, Tros de Ilarduya C, Aicart E, Guerrero-Martínez A, Junquera E. Gemini Cationic Lipid-Type Nanovectors Suitable for the Transfection of Therapeutic Plasmid DNA Encoding for Pro-Inflammatory Cytokine Interleukin-12. Pharmaceutics 2021; 13:729. [PMID: 34063469 PMCID: PMC8156092 DOI: 10.3390/pharmaceutics13050729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/26/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Ample evidence exists on the role of interleukin-12 (IL-12) in the response against many pathogens, as well as on its remarkable antitumor properties. However, the unexpected toxicity and disappointing results in some clinical trials are prompting the design of new strategies and/or vectors for IL-12 delivery. This study was conceived to further endorse the use of gemini cationic lipids (GCLs) in combination with zwitterionic helper lipid DOPE (1,2-dioleoyl-sn-glycero-3-phosphatidyl ethanol amine) as nanovectors for the insertion of plasmid DNA encoding for IL-12 (pCMV-IL12) into cells. Optimal GCL formulations previously reported by us were selected for IL-12-based biophysical experiments. In vitro studies demonstrated efficient pCMV-IL12 transfection by GCLs with comparable or superior cytokine levels than those obtained with commercial control Lipofectamine2000*. Furthermore, the nanovectors did not present significant toxicity, showing high cell viability values. The proteins adsorbed on the nanovector surface were found to be mostly lipoproteins and serum albumin, which are both beneficial to increase the blood circulation time. These outstanding results are accompanied by an initial physicochemical characterization to confirm DNA compaction and protection by the lipid mixture. Although further studies would be necessary, the present GCLs exhibit promising characteristics as candidates for pCMV-IL12 transfection in future in vivo applications.
Collapse
Affiliation(s)
- Natalia Sánchez-Arribas
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (N.S.-A.); (M.M.-N.); (E.A.); (A.G.-M.)
| | - María Martínez-Negro
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (N.S.-A.); (M.M.-N.); (E.A.); (A.G.-M.)
| | - Clara Aicart-Ramos
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain;
| | - Conchita Tros de Ilarduya
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, 31080 Pamplona, Spain;
| | - Emilio Aicart
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (N.S.-A.); (M.M.-N.); (E.A.); (A.G.-M.)
| | - Andrés Guerrero-Martínez
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (N.S.-A.); (M.M.-N.); (E.A.); (A.G.-M.)
| | - Elena Junquera
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain; (N.S.-A.); (M.M.-N.); (E.A.); (A.G.-M.)
| |
Collapse
|
29
|
Chakraborty S, Mallajosyula V, Tato CM, Tan GS, Wang TT. SARS-CoV-2 vaccines in advanced clinical trials: Where do we stand? Adv Drug Deliv Rev 2021; 172:314-338. [PMID: 33482248 PMCID: PMC7816567 DOI: 10.1016/j.addr.2021.01.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
The ongoing SARS-CoV-2 pandemic has led to the focused application of resources and scientific expertise toward the goal of developing investigational vaccines to prevent COVID-19. The highly collaborative global efforts by private industry, governments and non-governmental organizations have resulted in a number of SARS-CoV-2 vaccine candidates moving to Phase III trials in a period of only months since the start of the pandemic. In this review, we provide an overview of the preclinical and clinical data on SARS-CoV-2 vaccines that are currently in Phase III clinical trials and in few cases authorized for emergency use. We further discuss relevant vaccine platforms and provide a discussion of SARS-CoV-2 antigens that may be targeted to increase the breadth and durability of vaccine responses.
Collapse
Affiliation(s)
- Saborni Chakraborty
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA, USA
| | - Vamsee Mallajosyula
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Cristina M Tato
- Infectious Disease Initiative, Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Gene S Tan
- J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA 92037, USA; Department of Infectious Diseases, University of California San Diego, La Jolla, CA 92037, USA
| | - Taia T Wang
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
30
|
Sakr MM, Elsayed NS, El-Housseiny GS. Latest updates on SARS-CoV-2 genomic characterization, drug, and vaccine development; a comprehensive bioinformatics review. Microb Pathog 2021; 154:104809. [PMID: 33647446 PMCID: PMC7910145 DOI: 10.1016/j.micpath.2021.104809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/18/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023]
Abstract
Amid the COVID-19 outbreak, several bioinformatic analyses have been conducted on SARS-CoV-2 virus genome. Numerous studies rushed to fill the gap about this novel virus. Comparison with other related sequences, structural predictions of the produced proteins, determination of variations in amino acid residues and depiction of possible drug and vaccine targets have been the focus of scientific research from the beginning of this year. In addition to discussing the viral taxonomy, clinical features, life cycle, and genome organization, this review will focus on the recent updates in genome and viral proteins characterization and potential therapeutic and vaccine candidates developed so far. Comparative studies with related genomes and proteins provide understanding for the viral molecular mechanisms and suggest targets for therapeutics and vaccinology trials to stop the escalation of this new virus. This pandemic, with its resulting social and economic afflictions, will definitely have significant marks on our lives in the following years.
Collapse
Affiliation(s)
- Masarra M Sakr
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St., 11566, Abbassia, Cairo, Egypt
| | - Noha S Elsayed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St., 11566, Abbassia, Cairo, Egypt.
| | - Ghadir S El-Housseiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St., 11566, Abbassia, Cairo, Egypt
| |
Collapse
|
31
|
Alves AMB, Costa SM, Pinto PBA. Dengue Virus and Vaccines: How Can DNA Immunization Contribute to This Challenge? FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:640964. [PMID: 35047911 PMCID: PMC8757892 DOI: 10.3389/fmedt.2021.640964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/17/2021] [Indexed: 01/02/2023] Open
Abstract
Dengue infections still have a tremendous impact on public health systems in most countries in tropical and subtropical regions. The disease is systemic and dynamic with broad range of manifestations, varying from mild symptoms to severe dengue (Dengue Hemorrhagic Fever and Dengue Shock Syndrome). The only licensed tetravalent dengue vaccine, Dengvaxia, is a chimeric yellow fever virus with prM and E genes from the different dengue serotypes. However, recent results indicated that seronegative individuals became more susceptible to develop severe dengue when infected after vaccination, and now WHO recommends vaccination only to dengue seropositive people. One possibility to explain these data is the lack of robust T-cell responses and antibody-dependent enhancement of virus replication in vaccinated people. On the other hand, DNA vaccines are excellent inducers of T-cell responses in experimental animals and it can also elicit antibody production. Clinical trials with DNA vaccines have improved and shown promising results regarding the use of this approach for human vaccination. Therefore, in this paper we review preclinical and clinical tests with DNA vaccines against the dengue virus. Most of the studies are based on the E protein since this antigen is the main target for neutralizing antibody production. Yet, there are other reports with DNA vaccines based on non-structural dengue proteins with protective results, as well. Combining structural and non-structural genes may be a solution for inducing immune responses aging in different infection moments. Furthermore, DNA immunizations are also a very good approach in combining strategies for vaccines against dengue, in heterologous prime/boost regimen or even administering different vaccines at the same time, in order to induce efficient humoral and cellular immune responses.
Collapse
Affiliation(s)
- Ada Maria Barcelos Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | | |
Collapse
|