51
|
Yang W, Zhang D, Li Z, Zhang K. Predictors of poor serologic response to COVID-19 vaccine in patients with cancer: a systematic review and meta-analysis. Eur J Cancer 2022; 172:41-50. [PMID: 35752155 PMCID: PMC9160160 DOI: 10.1016/j.ejca.2022.05.031] [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: 04/04/2022] [Revised: 05/14/2022] [Accepted: 05/24/2022] [Indexed: 02/08/2023]
Abstract
Backgrounds Patients with cancer presented a lower probability to obtain seroconversion after a complete course of COVID-19 vaccination. However, little was known on the factors that predict poor seroconversion in this frail population. Methods We searched the PubMed, EMBASE, and China National Knowledge Infrastructure databases for all articles within a range of published years from 2019 to 2022 on the predictors of response to COVID-19 vaccine in patients with cancer (last search was updated on 2st March 2022). The odds ratio corresponding to the 95% confidence interval was used to assess the outcome. The statistical heterogeneity among studies was assessed with the Q-test and I2 statistics. The review was registered with PROSPERO (CRD42022315687) and reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Results Twenty cohort studies met the inclusion criteria for this study, with 5,499 patients with cancer. We found that advanced age, male patients, and metastatic disease increased negative seropositivity to COVID-19 vaccine. Immunoglobulin heavy chain variable mutation status, high concentration of Ig G, Ig M, and Ig A were correlated with seropositivity. Relating to cancer treatment strategy, anti-CD20 therapy within recent 12 months and chemotherapy were negatively correlated with seroconversion. Meta-analysis found no significant difference associated with targeted treatment, immunotherapy, and endocrine treatment. Conclusions Our meta-analysis assessed the factors that predict poor seroconversion in order to plan better prevention strategies in this frail population. The results proposed that enhanced vaccination strategies would be beneficial for the special patients such as advanced male, or patients receiving active chemotherapy, and carefully prevention should be emphasised even after a complete course of vaccination.
Collapse
Affiliation(s)
- Wenxing Yang
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Dongxue Zhang
- Equipment and Material Department, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Zhuo Li
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Kui Zhang
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.
| |
Collapse
|
52
|
Zhang C, Yang M. Newly Emerged Antiviral Strategies for SARS-CoV-2: From Deciphering Viral Protein Structural Function to the Development of Vaccines, Antibodies, and Small Molecules. Int J Mol Sci 2022; 23:6083. [PMID: 35682761 PMCID: PMC9181103 DOI: 10.3390/ijms23116083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 01/09/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become the most severe health crisis, causing extraordinary economic disruption worldwide. SARS-CoV-2 is a single-stranded RNA-enveloped virus. The process of viral replication and particle packaging is finished in host cells. Viral proteins, including both structural and nonstructural proteins, play important roles in the viral life cycle, which also provides the targets of treatment. Therefore, a better understanding of the structural function of virus proteins is crucial to speed up the development of vaccines and therapeutic strategies. Currently, the structure and function of proteins encoded by the SARS-CoV-2 genome are reviewed by several studies. However, most of them are based on the analysis of SARS-CoV-1 particles, lacking a systematic review update for SARS-CoV-2. Here, we specifically focus on the structure and function of proteins encoded by SARS-CoV-2. Viral proteins that contribute to COVID-19 infection and disease pathogenesis are reviewed according to the most recent research findings. The structure-function correlation of viral proteins provides a fundamental rationale for vaccine development and targeted therapy. Then, current antiviral vaccines are updated, such as inactive viral vaccines and protein-based vaccines and DNA, mRNA, and circular RNA vaccines. A summary of other therapeutic options is also reviewed, including monoclonal antibodies such as a cross-neutralizer antibody, a constructed cobinding antibody, a dual functional monoclonal antibody, an antibody cocktail, and an engineered bispecific antibody, as well as peptide-based inhibitors, chemical compounds, and clustered regularly interspaced short palindromic repeats (CRISPR) exploration. Overall, viral proteins and their functions provide the basis for targeted therapy and vaccine development.
Collapse
Affiliation(s)
- Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65212, USA;
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
53
|
Leishmania tarentolae as an Antigen Delivery Platform: Dendritic Cell Maturation after Infection with a Clone Engineered to Express the SARS-CoV-2 Spike Protein. Vaccines (Basel) 2022; 10:vaccines10050803. [PMID: 35632559 PMCID: PMC9144667 DOI: 10.3390/vaccines10050803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 01/18/2023] Open
Abstract
Background: Protozoa of the genus Leishmania are characterized by their capacity to target macrophages and Dendritic Cells (DCs). These microorganisms could thus be exploited for the delivery of antigens to immune cells. Leishmania tarentolae is regarded as a non-pathogenic species; it was previously used as a biofactory for protein production and has been considered as a candidate vaccine or as an antigen delivery platform. However, results on the type of immune polarization determined by L. tarentolae are still inconclusive. Methods: DCs were derived from human monocytes and exposed to live L. tarentolae, using both the non-engineered P10 strain, and the same strain engineered for expression of the spike protein from SARS-CoV-2. We then determined: (i) parasite internalization in the DCs; and (ii) the capacity of the assayed strains to activate DCs and the type of immune polarization. Results: Protozoan parasites from both strains were effectively engulfed by DCs, which displayed a full pattern of maturation, in terms of MHC class II and costimulatory molecule expression. In addition, after parasite infection, a limited release of Th1 cytokines was observed. Conclusions: Our results indicate that L. tarentolae could be used as a vehicle for antigen delivery to DCs and to induce the maturation of these cells. The limited cytokine release suggests L. tarentolae as a neutral vaccine vehicle that could be administered in association with appropriate immune-modulating molecules.
Collapse
|
54
|
An adjuvanted zoster vaccine elicits potent cellular immune responses in mice without QS21. NPJ Vaccines 2022; 7:45. [PMID: 35459225 PMCID: PMC9033770 DOI: 10.1038/s41541-022-00467-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Herpes zoster (HZ) is caused by reactivation of latent varicella-zoster virus (VZV) when VZV-specific cellular immunity is insufficient to control reactivation. Currently, Shingrix, which contains the VZV gE protein and GSK’s AS01B adjuvant composed of liposomes formulated with cholesterol, monophosphoryl lipid A (MPL) and QS21, is used for prevention of HZ. However, reactogenicity to Shingrix is common leading to poor patient compliance in receiving one or both shots. Here, we evaluated the immunogenicity of a newly formulated gE protein-based HZ vaccine containing Second-generation Lipid Adjuvant (SLA), a synthetic TLR4 ligand, formulated in an oil-in-water emulsion (SLA-SE) without QS21 (gE/SLA-SE). In VZV-primed mouse models, gE/SLA-SE-induced gE-specific humoral and cellular immune responses at comparable levels to those elicited by Shingrix in young mice, as both gE/SLA-SE and Shingrix induce polyfunctional CD4+ T-cell responses. In aged mice, gE/SLA-SE elicited more robust gE-specific T-cell responses than Shingrix. Furthermore, gE/SLA-SE-induced T-cell responses were sustained until 5 months after immunization. Thus, QS21-free, gE/SLA-SE is a promising candidate for development of gE-based HZ vaccines with high immunogenicity—particularly when targeting an older population.
Collapse
|
55
|
Vishweshwaraiah YL, Dokholyan NV. Toward rational vaccine engineering. Adv Drug Deliv Rev 2022; 183:114142. [PMID: 35150769 PMCID: PMC8931536 DOI: 10.1016/j.addr.2022.114142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/29/2022]
Abstract
Technological revolutions in several fields have pushed the boundaries of vaccine design and provided new avenues for vaccine development. Next-generation vaccine platforms have shown promise in targeting challenging antigens, for which traditional approaches have been ineffective. With advances in protein engineering, structural biology, computational biology and immunology, the structural vaccinology approach, which uses protein structure information to develop immunogens, holds promise for future vaccine design. In this review, we highlight various vaccine development strategies, along with their advantages and limitations. We discuss the rational vaccine design approach, which focuses on structure-based vaccine design. Finally, we discuss antigen engineering using the epitope-scaffold approach, gaps in structural vaccinology, and remaining challenges in vaccine design.
Collapse
Affiliation(s)
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033-0850, USA; Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033-0850, USA; Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
56
|
Methodological advances in the design of peptide-based vaccines. Drug Discov Today 2022; 27:1367-1380. [DOI: 10.1016/j.drudis.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/02/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022]
|
57
|
Qiao Y, Li S, Jin S, Pan Y, Shi Y, Kong W, Shan Y. A self-assembling nanoparticle vaccine targeting the conserved epitope of influenza virus hemagglutinin stem elicits a cross-protective immune response. NANOSCALE 2022; 14:3250-3260. [PMID: 35157751 DOI: 10.1039/d1nr08460g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Various vaccine strategies have been developed to provide broad protection against diverse influenza viruses. The hemagglutinin (HA) stem is the major potential target of these vaccines. Enhancing immunogenicity and eliciting cross-protective immune responses are critical for HA stem-based vaccine designs. In this study, the A helix (Ah) and CD helix (CDh) from the HA stem were fused with ferritin, individually, or in tandem, yielding Ah-f, CDh-f and (A + CD)h-f nanoparticles (NPs), respectively. These NPs were produced through a prokaryotic expression system. After three immunizations with AS03-adjuvanted NPs in BALB/c mice via the subcutaneous route, CDh-f and (A + CD)h-f induced robust humoral and cellular immune responses. Furthermore, CDh-f and (A + CD)h-f conferred complete protection against a lethal challenge of H3N2 virus, while no remarkable immune responses and protective effects were detected in the Ah-f group. These results indicate that the CDh-based nanovaccine represents a promising vaccine platform against influenza, and the epitope-conjugated ferritin NPs may be a potential vaccine platform against other infectious viruses, such as SARS-COV-2.
Collapse
Affiliation(s)
- Yongbo Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| | - Shuang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| | - Shenghui Jin
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| | - Yi Pan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
58
|
Abstract
This review discusses peptide epitopes used as antigens in the development of vaccines in clinical trials as well as future vaccine candidates. It covers peptides used in potential immunotherapies for infectious diseases including SARS-CoV-2, influenza, hepatitis B and C, HIV, malaria, and others. In addition, peptides for cancer vaccines that target examples of overexpressed proteins are summarized, including human epidermal growth factor receptor 2 (HER-2), mucin 1 (MUC1), folate receptor, and others. The uses of peptides to target cancers caused by infective agents, for example, cervical cancer caused by human papilloma virus (HPV), are also discussed. This review also provides an overview of model peptide epitopes used to stimulate non-specific immune responses, and of self-adjuvanting peptides, as well as the influence of other adjuvants on peptide formulations. As highlighted in this review, several peptide immunotherapies are in advanced clinical trials as vaccines, and there is great potential for future therapies due the specificity of the response that can be achieved using peptide epitopes.
Collapse
Affiliation(s)
- Ian W Hamley
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| |
Collapse
|
59
|
Alu A, Chen L, Lei H, Wei Y, Tian X, Wei X. Intranasal COVID-19 vaccines: From bench to bed. EBioMedicine 2022; 76:103841. [PMID: 35085851 PMCID: PMC8785603 DOI: 10.1016/j.ebiom.2022.103841] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 02/05/2023] Open
Abstract
Currently licensed COVID-19 vaccines are all designed for intramuscular (IM) immunization. However, vaccination today failed to prevent the virus infection through the upper respiratory tract, which is partially due to the absence of mucosal immunity activation. Despite the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, the next generation of COVID-19 vaccine is in demand and intranasal (IN) vaccination method has been demonstrated to be potent in inducing both mucosal and systemic immune responses. Presently, although not licensed, various IN vaccines against SARS-CoV-2 are under intensive investigation, with 12 candidates reaching clinical trials at different phases. In this review, we give a detailed description about current status of IN COVID-19 vaccines, including virus-vectored vaccines, recombinant subunit vaccines and live attenuated vaccines. The ongoing clinical trials for IN vaccines are highlighted. Additionally, the underlying mechanisms of mucosal immunity and potential mucosal adjuvants and nasal delivery devices are also summarized.
Collapse
Affiliation(s)
- Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Lei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
60
|
Côté-Cyr M, Zottig X, Gauthier L, Archambault D, Bourgault S. Self-Assembly of Flagellin into Immunostimulatory Ring-like Nanostructures as an Antigen Delivery System. ACS Biomater Sci Eng 2022; 8:694-707. [PMID: 35080372 DOI: 10.1021/acsbiomaterials.1c01332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Proteinaceous nanoparticles represent attractive antigen carriers for vaccination as their size and repetitive antigen displays that mimic most viral particles enable efficient immune processing. However, these nanocarriers are often unable to stimulate efficiently the innate immune system, requiring coadministration with adjuvants to promote long-lasting protective immunity. The protein flagellin, which constitutes the primary constituent of the bacterial flagellum, has been widely evaluated as an antigen carrier due to its intrinsic adjuvant properties involving activation of the innate immune receptor Toll-like receptor 5 (TLR5). Although flagellin is known for its ability to self-assemble into micron-scale length nanotubes, few studies have evaluated the potential usage of flagellin-based nanostructures as immunostimulatory antigen carriers. In this study, we reported for the first time a strategy to guide the self-assembly of a flagellin protein from Bacillus subtilis, Hag, into lower aspect ratio nanoparticles by hindering non-covalent interactions responsible for its elongation into nanotubes. We observed that addition of an antigenic sequence derived from the influenza A virus (3M2e) at the C-terminus of this flagellin, as opposed to positioning the epitope into mid-sequence, precluded filament elongation and resulted in low aspect ratio ring-like nanostructures upon salting-out-induced self-assembly. These nanostructures displayed the antigen at their surface and shared morphological and structural characteristics with flagellin nanotubes, with a diameter of approximately 12 nm, and an α-helix-rich secondary structure. Flagellin ring-like nanostructures were efficiently internalized by antigen-presenting cells, and avidly activated the TLR5 in vitro as well as the innate and adaptive immune responses. Intranasal immunization of mice with these nanostructures resulted in the potentiation of the antigen-specific antibody response and protection against a lethal infection with the influenza A virus, illustrating the potential of these intrinsically immunostimulatory nanostructures as antigen carriers.
Collapse
Affiliation(s)
- Mélanie Côté-Cyr
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Ximena Zottig
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Laurie Gauthier
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| |
Collapse
|
61
|
Wuethrich I, W. Pelzer B, Khodamoradi Y, Vehreschild MJGT. The role of the human gut microbiota in colonization and infection with multidrug-resistant bacteria. Gut Microbes 2022; 13:1-13. [PMID: 33870869 PMCID: PMC8078746 DOI: 10.1080/19490976.2021.1911279] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
About 100 years ago, the first antibiotic drug was introduced into health care. Since then, antibiotics have made an outstanding impact on human medicine. However, our society increasingly suffers from collateral damage exerted by these highly effective drugs. The rise of resistant pathogen strains, combined with a reduction of microbiota diversity upon antibiotic treatment, has become a significant obstacle in the fight against invasive infections worldwide.Alternative and complementary strategies to classical "Fleming antibiotics" comprise microbiota-based treatments such as fecal microbiota transfer and administration of probiotics, live-biotherapeutics, prebiotics, and postbiotics. Other promising interventions, whose efficacy may also be influenced by the human microbiota, are phages and vaccines. They will facilitate antimicrobial stewardship, to date the only globally applied antibiotic resistance mitigation strategy.In this review, we present the available evidence on these nontraditional interventions, highlight their interaction with the human microbiota, and discuss their clinical applicability.
Collapse
Affiliation(s)
- Irene Wuethrich
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Benedikt W. Pelzer
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | - Yascha Khodamoradi
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt Am Main, Germany
| | - Maria J. G. T. Vehreschild
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt Am Main, Germany,CONTACT Maria J. G. T. Vehreschild Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt Am Main, Germany
| |
Collapse
|
62
|
Li X, Pan C, Sun P, Peng Z, Feng E, Wu J, Wang H, Zhu L. Orthogonal modular biosynthesis of nanoscale conjugate vaccines for vaccination against infection. NANO RESEARCH 2022; 15:1645-1653. [PMID: 34405037 PMCID: PMC8359766 DOI: 10.1007/s12274-021-3713-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 05/21/2023]
Abstract
UNLABELLED Conjugate vaccines represent one of the most effective means for controlling the occurrence of bacterial diseases. Although nanotechnology has been greatly applied in the field of vaccines, it is seldom used for conjugate vaccine research because it is very difficult to connect polysaccharides and nanocarriers. In this work, an orthogonal and modular biosynthesis method was used to produce nanoconjugate vaccines using the SpyTag/SpyCatcher system. When SpyTag/SpyCatcher system is combined with protein glycosylation technology, bacterial O-polysaccharide obtained from Shigela flexneri 2a can be conjugated onto the surfaces of different virus-like particles (VLPs) in a biocompatible and controlled manner. After confirming the excellent lymph node targeting and humoral immune activation abilities, these nanoconjugate vaccines further induced efficient prophylactic effects against infection in a mouse model. These results demonstrated that natural polysaccharide antigens can be easily connected to VLPs to prepare highly efficient nanoconjugate vaccines. To the best of the researchers' knowledge, this is the first time VLP-based nanoconjugate vaccines are produced efficiently, and this strategy could be applied to develop various pathogenic nanoconjugate vaccines. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (Figs. S1-S9) is available in the online version of this article at 10.1007/s12274-021-3713-4.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Zhehui Peng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Erling Feng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| |
Collapse
|
63
|
Tsakiri M, Naziris N, Demetzos C. Innovative vaccine platforms against infectious diseases: Under the scope of the COVID-19 pandemic. Int J Pharm 2021; 610:121212. [PMID: 34687816 PMCID: PMC8527590 DOI: 10.1016/j.ijpharm.2021.121212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 12/30/2022]
Abstract
While classic vaccines have proved greatly efficacious in eliminating serious infectious diseases, innovative vaccine platforms open a new pathway to overcome dangerous pandemics via the development of safe and effective formulations. Such platforms play a key role either as antigen delivery systems or as immune-stimulators that induce both innate and adaptive immune responses. Liposomes or lipid nanoparticles, virus-like particles, nanoemulsions, polymeric or inorganic nanoparticles, as well as viral vectors, all belong to the nanoscale and are the main categories of innovative vaccines that are currently on the market or in clinical and preclinical phases. In this paper, we review the above formulations used in vaccinology and we discuss their connection with the development of safe and effective prophylactic vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Maria Tsakiri
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Nikolaos Naziris
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece.
| |
Collapse
|
64
|
Heinimäki S, Lampinen V, Tamminen K, Hankaniemi MM, Malm M, Hytönen VP, Blazevic V. Antigenicity and immunogenicity of HA2 and M2e influenza virus antigens conjugated to norovirus-like, VP1 capsid-based particles by the SpyTag/SpyCatcher technology. Virology 2021; 566:89-97. [PMID: 34894525 DOI: 10.1016/j.virol.2021.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Virus-like particles (VLPs) modified through different molecular technologies are employed as delivery vehicles or platforms for heterologous antigen display. We have recently created a norovirus (NoV) VLP platform, where two influenza antigens, the extracellular domain of matrix protein M2 (M2e) or the stem domain of the major envelope glycoprotein hemagglutinin (HA2) are displayed on the surface of the NoV VLPs by SpyTag/SpyCatcher conjugation. To demonstrate the feasibility of the platform to deliver foreign antigens, this study examined potential interference of the conjugation with induction of antibodies against conjugated M2e peptide, HA2, and NoV VLP carrier. High antibody response was induced by HA2 but not M2e decorated VLPs. Furthermore, HA2-elicited antibodies did not neutralize the homologous influenza virus in vitro. Conjugated NoV VLPs retained intact receptor binding capacity and self-immunogenicity. The results demonstrate that NoV VLPs could be simultaneously used as a platform to deliver foreign antigens and a NoV vaccine.
Collapse
Affiliation(s)
- Suvi Heinimäki
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Vili Lampinen
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi Tamminen
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna M Hankaniemi
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maria Malm
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Vesa P Hytönen
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Tampere, Finland
| | - Vesna Blazevic
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
65
|
Mimicking Native Display of CD0873 on Liposomes Augments Its Potency as an Oral Vaccine against Clostridioides difficile. Vaccines (Basel) 2021; 9:vaccines9121453. [PMID: 34960199 PMCID: PMC8708880 DOI: 10.3390/vaccines9121453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/23/2022] Open
Abstract
Mucosal vaccination aims to prevent infection mainly by inducing secretory IgA (sIgA) antibody, which neutralises pathogens and enterotoxins by blocking their attachment to epithelial cells. We previously demonstrated that encapsulated protein antigen CD0873 given orally to hamsters induces neutralising antibodies locally as well as systemically, affording partial protection against Clostridioides difficile infection. The aim of this study was to determine whether displaying CD0873 on liposomes, mimicking native presentation, would drive a stronger antibody response. The recombinant form we previously tested resembles the naturally cleaved lipoprotein commencing with a cysteine but lacking lipid modification. A synthetic lipid (DHPPA-Mal) was designed for conjugation of this protein via its N-terminal cysteine to the maleimide headgroup. DHPPA-Mal was first formulated with liposomes to produce MalLipo; then, CD0873 was conjugated to headgroups protruding from the outer envelope to generate CD0873-MalLipo. The immunogenicity of CD0873-MalLipo was compared to CD0873 in hamsters. Intestinal sIgA and CD0873-specific serum IgG were induced in all vaccinated animals; however, neutralising activity was greatest for the CD0873-MalLipo group. Our data hold great promise for development of a novel oral vaccine platform driving intestinal and systemic immune responses.
Collapse
|
66
|
Krishnan R, Stuart PM. Developments in Vaccination for Herpes Simplex Virus. Front Microbiol 2021; 12:798927. [PMID: 34950127 PMCID: PMC8691362 DOI: 10.3389/fmicb.2021.798927] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
Herpes simplex virus (HSV) is an alpha herpes virus, with two subtypes: HSV-1 and HSV-2. HSV is one of the most prevalent sexually transmitted infections. It is the cause of severe neonatal infections and a leading cause of infectious blindness in the Western world. As of 2016, 13.2% of the global population ages 15-49 were existing with HSV-2 infection and 66.6% with HSV-1. This high prevalence of disease and the fact that resistance to current therapies is on the rise makes it imperative to develop and discover new methods of HSV prevention and management. Among the arsenal of therapies/treatments for this virus has been the development of a prophylactic or therapeutic vaccine to prevent the complications of HSV reactivation. Our current understanding of the immune responses involved in latency and reactivation provides a unique challenge to the development of vaccines. There are no approved vaccines currently available for either prophylaxis or therapy. However, there are various promising candidates in the pre-clinical and clinical phases of study. Vaccines are being developed with two broad focuses: preventative and therapeutic, some with a dual use as both immunotherapeutic and prophylactic. Within this article, we will review the current guidelines for the treatment of herpes simplex infections, our understanding of the immunological pathways involved, and novel vaccine candidates in development.
Collapse
Affiliation(s)
| | - Patrick M. Stuart
- Department of Ophthalmology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
67
|
Abdelzaher HM, Gabr AS, Saleh BM, Abdel Gawad RM, Nour AA, Abdelanser A. RNA Vaccines against Infectious Diseases: Vital Progress with Room for Improvement. Vaccines (Basel) 2021; 9:1211. [PMID: 34835142 PMCID: PMC8622374 DOI: 10.3390/vaccines9111211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 01/14/2023] Open
Abstract
mRNA vaccines have amassed a strong interest from scientists and nonscientists alike for their potential in treating cancer and curbing the spread of infectious diseases. Their success has been bolstered by the COVID-19 pandemic as mRNA vaccines for the SARS-CoV-2 virus showed unrivaled efficiency and success. The strategy relies on the delivery of an RNA transcript that carries the sequence of an antigenic molecule into the body's cells where the antigen is manufactured. The lack of use of infectious pathogens and the fact that they are made of nucleic acids render these vaccines a favorable alternative to other vaccination modalities. However, mRNA vaccination still suffers from a great deal of hurdles starting from their safety, cellular delivery, uptake and response to their manufacturing, logistics and storage. In this review, we examine the premise of RNA vaccination starting from their conceptualization to their clinical applications. We also thoroughly discuss the advances in the field of RNA vaccination for infectious diseases. Finally, we discuss the challenges impeding their progress and shed light on potential areas of research in the field.
Collapse
Affiliation(s)
| | | | | | | | | | - Anwar Abdelanser
- Institute of Global Public Health, School of Sciences and Engineering, The American University in Cairo, Cairo 11835, Egypt; (H.M.A.); (A.S.G.); (B.M.S.); (R.M.A.G.); (A.A.N.)
| |
Collapse
|
68
|
Rodrigues MQ, Alves PM, Roldão A. Functionalizing Ferritin Nanoparticles for Vaccine Development. Pharmaceutics 2021; 13:1621. [PMID: 34683914 PMCID: PMC8540537 DOI: 10.3390/pharmaceutics13101621] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022] Open
Abstract
In the last decade, the interest in ferritin-based vaccines has been increasing due to their safety and immunogenicity. Candidates against a wide range of pathogens are now on Phase I clinical trials namely for influenza, Epstein-Barr, and SARS-CoV-2 viruses. Manufacturing challenges related to particle heterogeneity, improper folding of fused antigens, and antigen interference with intersubunit interactions still need to be overcome. In addition, protocols need to be standardized so that the production bioprocess becomes reproducible, allowing ferritin-based therapeutics to become readily available. In this review, the building blocks that enable the formulation of ferritin-based vaccines at an experimental stage, including design, production, and purification are presented. Novel bioengineering strategies of functionalizing ferritin nanoparticles based on modular assembly, allowing the challenges associated with genetic fusion to be circumvented, are discussed. Distinct up/down-stream approaches to produce ferritin-based vaccines and their impact on production yield and vaccine efficacy are compared. Finally, ferritin nanoparticles currently used in vaccine development and clinical trials are summarized.
Collapse
Affiliation(s)
- Margarida Q. Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.Q.R.); (P.M.A.)
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.Q.R.); (P.M.A.)
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - António Roldão
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.Q.R.); (P.M.A.)
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
69
|
Mohammadzadeh R, Soleimanpour S, Pishdadian A, Farsiani H. Designing and development of epitope-based vaccines against Helicobacter pylori. Crit Rev Microbiol 2021; 48:489-512. [PMID: 34559599 DOI: 10.1080/1040841x.2021.1979934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori infection is the principal cause of serious diseases (e.g. gastric cancer and peptic ulcers). Antibiotic therapy is an inadequate strategy in H. pylori eradication because of which vaccination is an inevitable approach. Despite the presence of countless vaccine candidates, current vaccines in clinical trials have performed with poor efficacy which makes vaccination extremely challenging. Remarkable advancements in immunology and pathogenic biology have provided an appropriate opportunity to develop various epitope-based vaccines. The fusion of proper antigens involved in different aspects of H. pylori colonization and pathogenesis as well as peptide linkers and built-in adjuvants results in producing epitope-based vaccines with excellent therapeutic efficacy and negligible adverse effects. Difficulties of the in vitro culture of H. pylori, high genetic variation, and unfavourable immune responses against feeble epitopes in the complete antigen are major drawbacks of current vaccine strategies that epitope-based vaccines may overcome. Besides decreasing the biohazard risk, designing precise formulations, saving time and cost, and induction of maximum immunity with minimum adverse effects are the advantages of epitope-based vaccines. The present article is a comprehensive review of strategies for designing and developing epitope-based vaccines to provide insights into the innovative vaccination against H. pylori.
Collapse
Affiliation(s)
- Roghayeh Mohammadzadeh
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Reference Tuberculosis Laboratory, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Pishdadian
- Department of Immunology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
70
|
Xu X, Yu R, Xiao L, Wang J, Yu M, Xu J, Tan Y, Ma X, Wu X, Lian J, Huang K, Ouyang X, Bi S, Wu S, Wang X, Jin J, Yu L, Zhang H, Wei Q, Shi J, Chen W, Li L. Safety and Immunogenicity of a Recombinant Tetanus Vaccine in Healthy Adults in China: A Randomized, Double-Blind, Dose Escalation, Placebo- and Positive-Controlled, Phase 1/2 Trial. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002751. [PMID: 34081408 PMCID: PMC8336487 DOI: 10.1002/advs.202002751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 04/13/2021] [Indexed: 06/12/2023]
Abstract
Tetanus is a fatal but vaccine-preventable disease. The currently available tetanus vaccines are tetanus toxoid (TT)-based. Although these vaccines are generally effective, challenges in vaccine development and access remain. A randomized, double-blind, dose escalation, placebo- and positive-controlled, phase 1/2 trial (ChiCTR1800015865) is performed to evaluate the safety and immunogenicity of an alternative recombinant tetanus vaccine based on the Hc domain of tetanus neurotoxin (TeNT-Hc) in healthy adult volunteers. The primary outcome is the safety profile of the recombinant tetanus vaccine, and immunogenicity is the secondary outcome. 150 eligible participants were enrolled and randomly assigned to receive one of the three doses of recombinant tetanus vaccine (TeNT-Hc 10/20/30 µg), TT vaccine, or placebo. The recombinant tetanus vaccine shows a good safety profile. The frequency of any solicited and unsolicited adverse events after each vaccination does not differ across the vaccine and placebo recipients. No serious treatment-related adverse events occur. The recombinant tetanus vaccine shows strong immune responses (seroconversion rates, geometric mean titer, and antigen-specific CD4+/CD8+ T-cell responses), which are roughly comparable to those of the TT vaccine. In conclusion, the findings from this study support that recombinant tetanus vaccine is safe and immunogenic; thereby, it represents a novel vaccine candidate against tetanus.
Collapse
Affiliation(s)
- Xiaowei Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Rui Yu
- Beijing Institute of BiotechnologyBeijing100071China
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Meihong Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Junjie Xu
- Beijing Institute of BiotechnologyBeijing100071China
| | - Yajun Tan
- National Institutes for Food and Drug ControlBeijing102629China
| | - Xiao Ma
- National Institutes for Food and Drug ControlBeijing102629China
| | - Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Jiangshan Lian
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Kaizhou Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Xiaoxi Ouyang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Sheng Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Shipo Wu
- Beijing Institute of BiotechnologyBeijing100071China
| | - Xiaoyan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Jiandi Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Ling Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Huafen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| | - Qi Wei
- Sichuan Zihao Times Pharmaceutical Co., LtdMeishanSichuan Province610000China
| | - Jinfa Shi
- Sichuan Zihao Times Pharmaceutical Co., LtdMeishanSichuan Province610000China
| | - Wei Chen
- Beijing Institute of BiotechnologyBeijing100071China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Centre for Infectious DiseasesCollaborative Innovation Centre for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou310003China
| |
Collapse
|
71
|
Liu ZH, Xu HL, Han GW, Tao LN, Lu Y, Zheng SY, Fang WH, He F. A self-assembling nanoparticle: Implications for the development of thermostable vaccine candidates. Int J Biol Macromol 2021; 183:2162-2173. [PMID: 34102236 DOI: 10.1016/j.ijbiomac.2021.06.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022]
Abstract
Effective controls on viral infections rely on the continuous development in vaccine technology. Nanoparticle (NP) antigens are highly immunogenic based on their unique physicochemical properties, making them molecular scaffolds to present soluble vaccine antigens. Here, viral targets (113-354 aas) were genetically fused to N terminal of mi3, a protein that self-assembles into nanoparticles composed of 60 subunits. With transmission electron microscopy, it was confirmed that target-mi3 fusion proteins which have insertions of up to 354 aas in N terminal form intact NPs. Moreover, viral targets are surface-displayed on NPs as indicated in dynamic light scattering. NPs exhibit perfect stability after long-term storage at room temperature. Moreover, SP-E2-mi3 NPs enhance antigen uptake and maturation in dendritic cells (DCs) via up-regulating marker molecules and immunostimulatory cytokines. Importantly, in a mouse model, SP-E2-mi3 nanovaccines against Classical swine fever virus (CSFV) remarkably improved CSFV-specific neutralizing antibodies (NAbs) and cellular immunity related cytokines (IFN-γ and IL-4) as compared to monomeric E2. Specially, improved NAb response with more than tenfold increase in NAb titer against both CSFV Shimen and HZ-08 strains indicated better cross-protection against different genotypes. Collectively, this structure-based, self-assembling NP provides an attractive platform to improve the potency of subunit vaccine for emerging pathogens.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/pharmacology
- Cells, Cultured
- Classical Swine Fever/blood
- Classical Swine Fever/immunology
- Classical Swine Fever/prevention & control
- Classical Swine Fever/virology
- Classical Swine Fever Virus/immunology
- Cytokines/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Disease Models, Animal
- Drug Stability
- Female
- Immunogenicity, Vaccine
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nanoparticles
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacology
- Swine
- Temperature
- Vaccines, Subunit/immunology
- Vaccines, Subunit/pharmacology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/pharmacology
- Viral Vaccines/immunology
- Viral Vaccines/pharmacology
- Mice
Collapse
Affiliation(s)
- Ze-Hui Liu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Hui-Ling Xu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Guang-Wei Han
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Li-Na Tao
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Ying Lu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Su-Ya Zheng
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Wei-Huan Fang
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou 310058, China.
| | - Fang He
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou 310058, China.
| |
Collapse
|
72
|
Liu ZH, Xu HL, Han GW, Tao LN, Lu Y, Zheng SY, Fang WH, He F. Self-Assembling Nanovaccine Enhances Protective Efficacy Against CSFV in Pigs. Front Immunol 2021; 12:689187. [PMID: 34367147 PMCID: PMC8334734 DOI: 10.3389/fimmu.2021.689187] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/01/2021] [Indexed: 01/01/2023] Open
Abstract
Classical swine fever virus (CSFV) is a highly contagious pathogen, which pose continuous threat to the swine industry. Though most attenuated vaccines are effective, they fail to serologically distinguish between infected and vaccinated animals, hindering CSFV eradication. Beneficially, nanoparticles (NPs)-based vaccines resemble natural viruses in size and antigen structure, and offer an alternative tool to circumvent these limitations. Using self-assembling NPs as multimerization platforms provides a safe and immunogenic tool against infectious diseases. This study presented a novel strategy to display CSFV E2 glycoprotein on the surface of genetically engineered self-assembling NPs. Eukaryotic E2-fused protein (SP-E2-mi3) could self-assemble into uniform NPs as indicated in transmission electron microscope (TEM) and dynamic light scattering (DLS). SP-E2-mi3 NPs showed high stability at room temperature. This NP-based immunization resulted in enhanced antigen uptake and up-regulated production of immunostimulatory cytokines in antigen presenting cells (APCs). Moreover, the protective efficacy of SP-E2-mi3 NPs was evaluated in pigs. SP-E2-mi3 NPs significantly improved both humoral and cellular immunity, especially as indicated by the elevated CSFV-specific IFN-γ cellular immunity and >10-fold neutralizing antibodies as compared to monomeric E2. These observations were consistent to in vivo protection against CSFV lethal virus challenge in prime-boost immunization schedule. Further results revealed single dose of 10 μg of SP-E2-mi3 NPs provided considerable clinical protection against lethal virus challenge. In conclusion, these findings demonstrated that this NP-based technology has potential to enhance the potency of subunit vaccine, paving ways for nanovaccine development.
Collapse
Affiliation(s)
- Ze-Hui Liu
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Hui-Ling Xu
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Guang-Wei Han
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Li-Na Tao
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Ying Lu
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Su-Ya Zheng
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wei-Huan Fang
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China.,Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Fang He
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China.,Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
73
|
Cid R, Bolívar J. Platforms for Production of Protein-Based Vaccines: From Classical to Next-Generation Strategies. Biomolecules 2021; 11:1072. [PMID: 34439738 PMCID: PMC8394948 DOI: 10.3390/biom11081072] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
To date, vaccination has become one of the most effective strategies to control and reduce infectious diseases, preventing millions of deaths worldwide. The earliest vaccines were developed as live-attenuated or inactivated pathogens, and, although they still represent the most extended human vaccine types, they also face some issues, such as the potential to revert to a pathogenic form of live-attenuated formulations or the weaker immune response associated with inactivated vaccines. Advances in genetic engineering have enabled improvements in vaccine design and strategies, such as recombinant subunit vaccines, have emerged, expanding the number of diseases that can be prevented. Moreover, antigen display systems such as VLPs or those designed by nanotechnology have improved the efficacy of subunit vaccines. Platforms for the production of recombinant vaccines have also evolved from the first hosts, Escherichia coli and Saccharomyces cerevisiae, to insect or mammalian cells. Traditional bacterial and yeast systems have been improved by engineering and new systems based on plants or insect larvae have emerged as alternative, low-cost platforms. Vaccine development is still time-consuming and costly, and alternative systems that can offer cost-effective and faster processes are demanding to address infectious diseases that still do not have a treatment and to face possible future pandemics.
Collapse
Affiliation(s)
- Raquel Cid
- ADL Bionatur Solutions S.A., Av. del Desarrollo Tecnológico 11, 11591 Jerez de la Frontera, Spain
| | - Jorge Bolívar
- Department of Biomedicine, Biotechnology and Public Health-Biochemistry and Molecular Biology, Campus Universitario de Puerto Real, University of Cadiz, 11510 Puerto Real, Spain
| |
Collapse
|
74
|
Karch CP, Matyas GR. The current and future role of nanovaccines in HIV-1 vaccine development. Expert Rev Vaccines 2021; 20:935-944. [PMID: 34184607 DOI: 10.1080/14760584.2021.1945448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: An efficacious vaccine for HIV-1 has been sought for over 30 years to eliminate the virus from the human population. Many challenges have occurred in the attempt to produce a successful immunogen, mainly caused by the basic biology of the virus. Immunogens have been developed focusing on inducing one or more of the following types of immune responses; neutralizing antibodies, non-neutralizing antibodies, and T-cell mediated responses. One way to better present and develop an immunogen for HIV-1 is through the use of nanotechnology and nanoparticles.Areas covered: This article gives a basic overview of the HIV-1 vaccine field, as well as nanotechnology, specifically nanovaccines. It then covers the application of nanovaccines made from biological macromolecules to HIV-1 vaccine development for neutralizing antibodies, non-neutralizing antibodies, and T-cell-mediated responses.Expert opinion: Nanovaccines are an area that is ripe for further exploration in HIV-1 vaccine field. Not only are nanovaccines capable of carrying and presenting antigens in native-like conformations, but they have also repeatedly been shown to increase immunogenicity over recombinant antigens alone. Only through further research can the true role of nanovaccines in the development of an efficacious HIV-1 vaccine be established.
Collapse
Affiliation(s)
- Christopher P Karch
- Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Laboratory of Adjuvant and Antigen Research, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
75
|
Aebischer A, Wernike K, König P, Franzke K, Wichgers Schreur PJ, Kortekaas J, Vitikainen M, Wiebe M, Saloheimo M, Tchelet R, Audonnet JC, Beer M. Development of a Modular Vaccine Platform for Multimeric Antigen Display Using an Orthobunyavirus Model. Vaccines (Basel) 2021; 9:vaccines9060651. [PMID: 34203630 PMCID: PMC8232151 DOI: 10.3390/vaccines9060651] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging infectious diseases represent an increasing threat to human and animal health. Therefore, safe and effective vaccines that could be available within a short time frame after an outbreak are required for adequate prevention and control. Here, we developed a robust and versatile self-assembling multimeric protein scaffold particle (MPSP) vaccine platform using lumazine synthase (LS) from Aquifex aeolicus. This scaffold allowed the presentation of peptide epitopes by genetic fusion as well as the presentation of large antigens by bacterial superglue-based conjugation to the pre-assembled particle. Using the orthobunyavirus model Schmallenberg virus (SBV) we designed MPSPs presenting major immunogens of SBV and assessed their efficacy in a mouse model as well as in cattle, a target species of SBV. All prototype vaccines conferred protection from viral challenge infection and the multivalent presentation of the selected antigens on the MPSP markedly improved their immunogenicity compared to the monomeric subunits. Even a single shot vaccination protected about 80% of mice from an otherwise lethal dose of SBV. Most importantly, the MPSPs induced a virtually sterile immunity in cattle. Altogether, LS represents a promising platform for modular and rapid vaccine design.
Collapse
Affiliation(s)
- Andrea Aebischer
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (A.A.); (K.W.); (P.K.); (K.F.)
| | - Kerstin Wernike
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (A.A.); (K.W.); (P.K.); (K.F.)
| | - Patricia König
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (A.A.); (K.W.); (P.K.); (K.F.)
| | - Kati Franzke
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (A.A.); (K.W.); (P.K.); (K.F.)
| | - Paul J. Wichgers Schreur
- Laboratory of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (P.J.W.S.); (J.K.)
| | - Jeroen Kortekaas
- Laboratory of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (P.J.W.S.); (J.K.)
| | - Marika Vitikainen
- VTT Technical Research Centre of Finland Ltd., 02150 Espoo, Finland; (M.V.); (M.W.); (M.S.)
| | - Marilyn Wiebe
- VTT Technical Research Centre of Finland Ltd., 02150 Espoo, Finland; (M.V.); (M.W.); (M.S.)
| | - Markku Saloheimo
- VTT Technical Research Centre of Finland Ltd., 02150 Espoo, Finland; (M.V.); (M.W.); (M.S.)
| | - Ronen Tchelet
- Dyadic Netherland B.V., 6709 PA Wageningen, The Netherlands;
| | | | - Martin Beer
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (A.A.); (K.W.); (P.K.); (K.F.)
- Correspondence:
| |
Collapse
|
76
|
Rahman N, Miura S, Okawa M, Kibria MG, Islam MM, Kuroda Y. Solubility Controlling Peptide Tags of Opposite Charges Generate a Bivalent Immune Response Against Dengue ED3 Serotypes 3 and 4. Front Immunol 2021; 12:671590. [PMID: 34177912 PMCID: PMC8226127 DOI: 10.3389/fimmu.2021.671590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/04/2021] [Indexed: 11/24/2022] Open
Abstract
We previously demonstrated that a protein’s immunogenicity could be substantially increased by attaching a hydrophobic solubility controlling peptide tag (SCP-tag) producing small sub-visible aggregates. Here, we report the oligomerization of Dengue envelop protein domain 3 (ED3), and consequently, its immunogenicity increase by mixing ED3s attached with SCP-tags of opposite charges at equimolar concentration. We used ED3 of serotype 3 (D3ED3) and serotype 4 (D4ED3), which are, respectively, moderately and poorly immunogenic, and their SCP tagged variants constructed by attaching either a C-termini 5-Aspartic acid (C5D) or a 5-Lysine (C5K) tag. Light scattering indicated that the isolated tagged ED3s remained monomeric, but mixing the C5D and C5K tagged ED3s at equimolar concentration generated sub-visible aggregates or oligomers of ~500 nm through electrostatic interaction. In addition, the oligomerized ED3s remained in a native-like state, as assessed by fluorescence spectroscopy and circular dichroism. The in vivo immunogenicity of the D3ED3 and D4ED3 oligomers generated by the charged tags increased by 5 and 16 fold, respectively. Furthermore, injection of heterotypic ED3 oligomers (D3C5D+D4C5K) induced an immune response against both D3ED3 and D4ED3 in 3 of 4 responsive mice, and the IgG titer of the bivalent anti-D3C5D-D4C5K sera was over 100 times higher than that generated by co-injecting the untagged D3ED3 and D4ED3 (D3+D4). Altogether, these observations suggest that SCP-tags could be used as a platform for producing a long-sought tetravalent dengue vaccine.
Collapse
Affiliation(s)
- Nafsoon Rahman
- Department of Biotechnology and Life Sciences, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shiho Miura
- Department of Biotechnology and Life Sciences, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mami Okawa
- Department of Biotechnology and Life Sciences, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Md Golam Kibria
- Department of Biotechnology and Life Sciences, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mohammad Monirul Islam
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Yutaka Kuroda
- Department of Biotechnology and Life Sciences, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
77
|
Ruseska I, Fresacher K, Petschacher C, Zimmer A. Use of Protamine in Nanopharmaceuticals-A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1508. [PMID: 34200384 PMCID: PMC8230241 DOI: 10.3390/nano11061508] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/18/2022]
Abstract
Macromolecular biomolecules are currently dethroning classical small molecule therapeutics because of their improved targeting and delivery properties. Protamine-a small polycationic peptide-represents a promising candidate. In nature, it binds and protects DNA against degradation during spermatogenesis due to electrostatic interactions between the negatively charged DNA-phosphate backbone and the positively charged protamine. Researchers are mimicking this technique to develop innovative nanopharmaceutical drug delivery systems, incorporating protamine as a carrier for biologically active components such as DNA or RNA. The first part of this review highlights ongoing investigations in the field of protamine-associated nanotechnology, discussing the self-assembling manufacturing process and nanoparticle engineering. Immune-modulating properties of protamine are those that lead to the second key part, which is protamine in novel vaccine technologies. Protamine-based RNA delivery systems in vaccines (some belong to the new class of mRNA-vaccines) against infectious disease and their use in cancer treatment are reviewed, and we provide an update on the current state of latest developments with protamine as pharmaceutical excipient for vaccines.
Collapse
Affiliation(s)
| | | | | | - Andreas Zimmer
- Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, 8010 Graz, Austria; (I.R.); (K.F.); (C.P.)
| |
Collapse
|
78
|
Fourie KR, Choudhary P, Ng SH, Obradovic M, Brownlie R, Anand SK, Wilson HL. Evaluation of immunogenicity and protection mediated by Lawsonia intracellularis subunit vaccines. Vet Immunol Immunopathol 2021; 237:110256. [PMID: 33971523 DOI: 10.1016/j.vetimm.2021.110256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 02/06/2023]
Abstract
Lawsonia intracellularis is an economically important bacterium that causes ileitis in pigs. Current vaccines for L. intracellularis do not allow for differentiation between infected and vaccinated animals (DIVA), which is beneficial for disease tracking and surveillance. Previously, we identified five putative surface L. intracellularis proteins that were targeted by antibodies from pigs infected with L. intracellularis which could serve as antigens in a subunit vaccine. We conducted two trials to determine whether these antigens were immunogenic and provided protection against infectious challenge and whether truncated glycoprotein D could be used as a DIVA antigen. For Trial 1, 5 week-old piglets were administered intramuscular monovalent vaccines comprised of a recombinant (r) flagella subunit protein (rFliC,) and DIVA antigen (truncated glycoprotein D (TgD), a herpes virus antigen) both formulated with a combination adjuvant consisting of polyinosinic:polycytidylic acid(poly I:C), host defense peptide 1002 and polyphosphazene, referred to as Triple Adjuvant (TriAdj). Relative to control animals, animals vaccinated with rFliC and rTgD had significantly elevated antigen-specific humoral immunity in sera suggesting that rFliC and TgD are immunogenic. Control animals had negligible anti-TgD titres suggesting that TgD may be a suitable DIVA antigen for pigs. For Trial 2, piglets were immunized with a trivalent vaccine (FOG vaccine consisting of rFLiC, rOppA protein (a ABC Type dipeptide transport system) and rGroEL (a stress response protein)) and a divalent vaccine (CM vaccine consisting of rClpP (an ATP-dependent Clp protease proteolytic subunit) and rMetK (a S-adenosyl methionine synthase)) formulated with Emulsigen®. Relative to the control pigs, pigs immunized with the FOG vaccine produced robust and significantly higher serum IgG antibodies against rFliC and rGroEL, and significantly higher anti-FliC and anti-GroEL IgA antibodies in jejunal (GroEL only) and ileal intestinal mucosa. Pigs immunized with CM vaccine produced significantly higher serum antibodies against rClpP and rMetK and significantly higher anti-rClpP IgA antibodies in the ileum relative to the control pigs. Quantitative polymerase chain reaction (qPCR) analysis showed that 18 days after challenge with infectious L. intracellularis, challenged/control pigs and pigs that received the CM vaccine, but not the pigs vaccinated with the FOG vaccine, shed significantly more bacteria in feces than the unchallenged controls pigs. These data suggest that the FOG vaccinated pigs showed limited protection. While promising, more work is needed to enhance the efficiency of the intramuscular vaccine to show significant disease protection.
Collapse
Affiliation(s)
- Kezia R Fourie
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Pooja Choudhary
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Siew Hon Ng
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Milan Obradovic
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montréal, Montréal, Quebec, Canada
| | - Robert Brownlie
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | - Heather L Wilson
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
79
|
Blunck BN, Rezende W, Piedra PA. Profile of respiratory syncytial virus prefusogenic fusion protein nanoparticle vaccine. Expert Rev Vaccines 2021; 20:351-364. [PMID: 33733995 DOI: 10.1080/14760584.2021.1903877] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Respiratory Syncytial Virus (RSV) is a leading cause of acute lower respiratory infections worldwide. The RSV fusion (F) glycoprotein is a major focus of vaccine development. Despite over 60 years of research, there is no licensed vaccine for RSV. AREAS COVERED The primary focus of this review is a novel RSV-F recombinant nanoparticle vaccine from Novavax utilizing the F protein, a conserved and immunodominant surface glycoprotein. This RSV F recombinant nanoparticle vaccine adsorbed to 0.4 mg of aluminum phosphate was ultimately administered by a single intramuscular injection during the third trimester of pregnancy in an effort to induce passive immunity in newborns. Its mechanism, performance in clinical trials, and place in RSV vaccine history are discussed. EXPERT OPINION The vaccine was safe and well tolerated in pregnant women and the results suggest potential benefits with respect to other medically relevant end-point events involving RSV-associated respiratory and all-cause disease in infants. However, the RSV-F recombinant nanoparticle vaccine did not meet the pre-specified primary success criteria for efficacy against RSV-associated, medically significant lower respiratory tract infection in infants up to 90 days of life. The potential benefits to infants from maternal immunization and excellent safety profile warrant further confirmatory studies.
Collapse
Affiliation(s)
- Brittani N Blunck
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, USA
| | - Wanderson Rezende
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, USA.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, USA
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, United States
| |
Collapse
|
80
|
Nascimento Júnior JAC, Santos AM, Cavalcante RCM, Quintans-Júnior LJ, Walker CIB, Borges LP, Frank LA, Serafini MR. Mapping the technological landscape of SARS, MERS, and SARS-CoV-2 vaccines. Drug Dev Ind Pharm 2021; 47:673-684. [PMID: 33826439 PMCID: PMC8040490 DOI: 10.1080/03639045.2021.1908343] [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] [Indexed: 12/23/2022]
Abstract
PURPOSE The last two decades have seen the emergence of several viral outbreaks. Some of them are the severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and severe acute respiratory syndrome 2 (SARS-CoV2) - the cause of the coronavirus disease 2019 (COVID-19). Ever, vaccines for emergency use have been authorized for the control and prevention of COVID-19. Currently, there is an urgent need to develop a vaccine for prophylaxis of COVID-19 and for other future epidemics. METHODS This review describes patented vaccines for SARS and MERS-CoV and vaccines developed and approved for emergency use against the new coronavirus (COVID-19). The European Patent Office and the World Intellectual Property Organization were the patent databases used using specific terms. In addition, another search was carried out in the Clinical Trials in search of ongoing clinical studies focused on the COVID-19 vaccine. RESULTS The patent search showed that most vaccines are based on viral vector platforms, nucleic acids, or protein subunits. The review also includes an overview of completed and ongoing clinical trials for SARS-CoV-2 in several countries. CONCLUSION The information provided here lists vaccines for other types of coronavirus that have been used in the development of vaccines for COVID-19.
Collapse
Affiliation(s)
- José Adão Carvalho Nascimento Júnior
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.,Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | | | - Lucindo José Quintans-Júnior
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.,Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Cristiani Isabel Banderó Walker
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.,Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Lysandro Pinto Borges
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Luiza Abrahão Frank
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Escola de Saúde e Bem Estar UniRitter, Faculdade de Farmácia - Laureate International Universities, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mairim Russo Serafini
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.,Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| |
Collapse
|
81
|
Jiang HD, Li JX, Zhang P, Huo X, Zhu FC. The COVID-19 Vaccine in Clinical Trials: Where Are We Now? INFECTIOUS DISEASES & IMMUNITY 2021; 1:43-51. [PMID: 38630107 PMCID: PMC8057314 DOI: 10.1097/id9.0000000000000003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Indexed: 12/23/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to scale up around the world, costing severe health and economic losses. The development of an effective COVID-19 vaccine is of utmost importance. Most vaccine designs can be classified into three camps: protein based (inactivated vaccines, protein subunit, VLP and T-cell based vaccines), gene based (DNA or RNA vaccines, replicating or non-replicating viral/bacterial vectored vaccines), and a combination of both protein-based and gene-based (live-attenuated virus vaccines). Up to now, 237 candidate vaccines against SARS-CoV-2 are in development worldwide, of which 63 have been approved for clinical trials and 27 are evaluated in phase 3 clinical trials. Six candidate vaccines have been authorized for emergency use or conditional licensed, based on their efficacy data in phase 3 trials. This review summarizes the strengths and weaknesses of the candidate COVID-19 vaccines from various platforms, compares, and discusses their protective efficacy, safety, and immunogenicity according to the published clinical trials results.
Collapse
Affiliation(s)
- Hu-Dachuan Jiang
- School of Public Health, Southeast University, Nanjing 210009, China
| | - Jing-Xin Li
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Peng Zhang
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Xiang Huo
- Food Safety and Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Feng-Cai Zhu
- School of Public Health, Southeast University, Nanjing 210009, China
- NHC Key Laboratory of Enteric Pathogenic Microbiology Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| |
Collapse
|
82
|
Faber E, Tshilwane SI, Kleef MV, Pretorius A. Virulent African horse sickness virus serotype 4 interferes with the innate immune response in horse peripheral blood mononuclear cells in vitro. INFECTION GENETICS AND EVOLUTION 2021; 91:104836. [PMID: 33798756 DOI: 10.1016/j.meegid.2021.104836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022]
Abstract
African horse sickness (AHS) is caused by African horse sickness virus (AHSV), a double stranded RNA (dsRNA) virus of the genus Orbivirus, family Reoviridae. For the development of new generation AHS vaccines or antiviral treatments, it is crucial to understand the host immune response against the virus and the immune evasion strategies the virus employs. To achieve this, the current study used transcriptome analysis of RNA sequences to characterize and compare the innate immune responses activated during the attenuated AHSV serotype 4 (attAHSV4) (in vivo) and the virulent AHSV4 (virAHSV4) (in vitro) primary and secondary immune responses in horse peripheral blood mononuclear cells (PBMC) after 24 h. The pro-inflammatory cytokine and chemokine responses were negatively regulated by anti-inflammatory cytokines, whereas the parallel type I and type III IFN responses were maintained downstream of nucleic acid sensing pattern recognition receptor (PRR) signalling pathways during the attAHSV4 primary and secondary immune responses. It appeared that after translation, virAHSV4 proteins were able to interfere with the C-terminal IRF association domain (IAD)-type 1 (IAD1) containing IRFs, which inhibited the expression of type I and type III IFNs downstream of PRR signalling during the virAHSV4 primary and secondary immune responses. Viral interference resulted in an impaired innate immune response that was not able to eliminate virAHSV4-infected PBMC and gave rise to prolonged expression of pro-inflammatory cytokines and chemokines during the virAHSV4 induced primary immune response. Indicating that virAHSV4 interference with the innate immune response may give rise to an excessive inflammatory response that causes immunopathology, which could be a major contributing factor to the pathogenesis of AHS in a naïve horse. Viral interference was overcome by the fast kinetics and increased effector responses of innate immune cells due to trained innate immunity and memory T cells and B cells during the virAHSV4 secondary immune response.
Collapse
Affiliation(s)
- Erika Faber
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort 0110, South Africa; Department of Veterinary Tropical Disease, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa.
| | - Selaelo Ivy Tshilwane
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Mirinda Van Kleef
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort 0110, South Africa; Department of Veterinary Tropical Disease, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa
| | - Alri Pretorius
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort 0110, South Africa; Department of Veterinary Tropical Disease, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa
| |
Collapse
|
83
|
Gebre MS, Brito LA, Tostanoski LH, Edwards DK, Carfi A, Barouch DH. Novel approaches for vaccine development. Cell 2021; 184:1589-1603. [PMID: 33740454 PMCID: PMC8049514 DOI: 10.1016/j.cell.2021.02.030] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023]
Abstract
Vaccines are critical tools for maintaining global health. Traditional vaccine technologies have been used across a wide range of bacterial and viral pathogens, yet there are a number of examples where they have not been successful, such as for persistent infections, rapidly evolving pathogens with high sequence variability, complex viral antigens, and emerging pathogens. Novel technologies such as nucleic acid and viral vector vaccines offer the potential to revolutionize vaccine development as they are well-suited to address existing technology limitations. In this review, we discuss the current state of RNA vaccines, recombinant adenovirus vector-based vaccines, and advances from biomaterials and engineering that address these important public health challenges.
Collapse
Affiliation(s)
- Makda S. Gebre
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- co-first authors
| | | | - Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- co-first authors
| | | | - Andrea Carfi
- Moderna, Inc., Cambridge, MA, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, USA
| |
Collapse
|
84
|
Karch CP, Burkhard P, Matyas GR, Beck Z. The diversity of HIV-1 fights against vaccine efficacy: how self-assembling protein nanoparticle technology may fight back. Nanomedicine (Lond) 2021; 16:673-680. [PMID: 33715403 DOI: 10.2217/nnm-2020-0450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An efficacious HIV-1 vaccine has remained an elusive target for almost 40 years. The sheer diversity of the virus is one of the major roadblocks for vaccine development. HIV-1 frequently mutates and various strains predominate in different geographic regions, making the development of a globally applicable vaccine extremely difficult. Multiple approaches have been taken to overcome the issue of viral diversity, including sequence optimization, development of consensus and mosaic sequences and the use of different prime-boost approaches. To develop an efficacious vaccine, these approaches may need to be combined. One way to potentially synergize these approaches is to use a rationally designed protein nanoparticle that allows for the native-like presentation of antigens, such as the self-assembling protein nanoparticle.
Collapse
Affiliation(s)
- Christopher P Karch
- US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910, USA.,Henry M Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Peter Burkhard
- Alpha-O Peptides, Lörracherstrasse 50, 4125 Riehen, Switzerland
| | - Gary R Matyas
- US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910, USA
| | - Zoltan Beck
- US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910, USA.,Henry M Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA.,Current address: VRD, Pfizer, 401 N Middletown Rd, Pearl River, NY 10965, USA
| |
Collapse
|
85
|
Kodama Y, Tokunaga A, Hashizume J, Nakagawa H, Harasawa H, Kurosaki T, Nakamura T, Nishida K, Nakashima M, Hashida M, Kawakami S, Sasaki H. Evaluation of transgene expression characteristics and DNA vaccination against melanoma metastasis of an intravenously injected ternary complex with biodegradable dendrigraft poly-L-lysine in mice. Drug Deliv 2021; 28:542-549. [PMID: 33685317 PMCID: PMC7946064 DOI: 10.1080/10717544.2021.1895904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
We developed a biocompatible splenic vector for a DNA vaccine against melanoma. The splenic vector is a ternary complex composed of plasmid DNA (pDNA), biodegradable dendrigraft poly-L-lysine (DGL), and γ-polyglutamic acid (γ-PGA), the selective uptake of which by the spleen has already been demonstrated. The ternary complex containing pDNA encoding luciferase (pCMV-Luc) exhibited stronger luciferase activity for RAW264.7 mouse macrophage-like cells than naked pCMV-Luc. Although the ternary complex exhibited strong luciferase activity in the spleen after its tail vein injection, luciferase activity in the liver and spleen was significantly decreased by a pretreatment with clodronate liposomes, which depleted macrophages in the liver and spleen. These results indicate that the ternary complex is mainly transfected in macrophages and is a suitable formulation for DNA vaccination. We applied the ternary complex to a pUb-M melanoma DNA vaccine. The ternary complex containing pUb-M suppressed the growth of melanoma and lung metastasis by B16-F10 mouse melanoma cells. We also examined the acute and liver toxicities of the pUb-M ternary complex at an excess pDNA dose in mice. All mice survived the injection of the excess amount of the ternary complex. Liver toxicity was negligible in mice injected with the excess amount of the ternary complex. In conclusion, we herein confirmed that the ternary complex was mainly transfected into macrophages in the spleen after its tail vein injection. We also showed the prevention of melanoma metastasis by the DNA vaccine and the safety of the ternary complex.
Collapse
Affiliation(s)
- Yukinobu Kodama
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Ayako Tokunaga
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Junya Hashizume
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroo Nakagawa
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Hitomi Harasawa
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Tomoaki Kurosaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Tadahiro Nakamura
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan.,Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Mikiro Nakashima
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Mitsuru Hashida
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Shigeru Kawakami
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hitoshi Sasaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| |
Collapse
|
86
|
Galloway JM, Bray HEV, Shoemark DK, Hodgson LR, Coombs J, Mantell JM, Rose RS, Ross JF, Morris C, Harniman RL, Wood CW, Arthur C, Verkade P, Woolfson DN. De Novo Designed Peptide and Protein Hairpins Self-Assemble into Sheets and Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100472. [PMID: 33590708 PMCID: PMC11475375 DOI: 10.1002/smll.202100472] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 06/12/2023]
Abstract
The design and assembly of peptide-based materials has advanced considerably, leading to a variety of fibrous, sheet, and nanoparticle structures. A remaining challenge is to account for and control different possible supramolecular outcomes accessible to the same or similar peptide building blocks. Here a de novo peptide system is presented that forms nanoparticles or sheets depending on the strategic placement of a "disulfide pin" between two elements of secondary structure that drive self-assembly. Specifically, homodimerizing and homotrimerizing de novo coiled-coil α-helices are joined with a flexible linker to generate a series of linear peptides. The helices are pinned back-to-back, constraining them as hairpins by a disulfide bond placed either proximal or distal to the linker. Computational modeling indicates, and advanced microscopy shows, that the proximally pinned hairpins self-assemble into nanoparticles, whereas the distally pinned constructs form sheets. These peptides can be made synthetically or recombinantly to allow both chemical modifications and the introduction of whole protein cargoes as required.
Collapse
Affiliation(s)
- Johanna M. Galloway
- School of ChemistryUniversity of BristolCantock's CloseBristolBS8 1TSUK
- School of ChemistryUniversity of LeedsLeedsLS2 9JTUK
| | | | - Deborah K. Shoemark
- School of BiochemistryUniversity of BristolMedical Sciences BuildingUniversity WalkBristolBS8 1TDUK
| | - Lorna R. Hodgson
- School of BiochemistryUniversity of BristolMedical Sciences BuildingUniversity WalkBristolBS8 1TDUK
- BrisSynBio/Bristol Biodesign InstituteUniversity of BristolLife Sciences Building, Tyndall AvenueBristolBS8 1TQUK
| | - Jennifer Coombs
- School of BiochemistryUniversity of BristolMedical Sciences BuildingUniversity WalkBristolBS8 1TDUK
- Bristol Centre for Functional NanomaterialsSchool of PhysicsUniversity of BristolHH Wills Physics LaboratoryTyndall AvenueBristolBS8 1TLUK
| | - Judith M. Mantell
- School of BiochemistryUniversity of BristolMedical Sciences BuildingUniversity WalkBristolBS8 1TDUK
| | - Ruth S. Rose
- School of Biological and Chemical SciencesFogg BuildingQueen Mary University of LondonMile End RoadLondonE1 4QDUK
| | - James F. Ross
- School of ChemistryUniversity of BristolCantock's CloseBristolBS8 1TSUK
- School of ChemistryUniversity of LeedsLeedsLS2 9JTUK
| | - Caroline Morris
- School of ChemistryUniversity of BristolCantock's CloseBristolBS8 1TSUK
- BrisSynBio/Bristol Biodesign InstituteUniversity of BristolLife Sciences Building, Tyndall AvenueBristolBS8 1TQUK
- School of ChemistryUniversity of Glasgow0/1 125 Novar DriveGlasgowG12 9TAUK
| | | | - Christopher W. Wood
- School of ChemistryUniversity of BristolCantock's CloseBristolBS8 1TSUK
- BrisSynBio/Bristol Biodesign InstituteUniversity of BristolLife Sciences Building, Tyndall AvenueBristolBS8 1TQUK
- School of Biological SciencesRoger Land Building, King's BuildingsEdinburghEH9 3JQUK
| | | | - Paul Verkade
- School of BiochemistryUniversity of BristolMedical Sciences BuildingUniversity WalkBristolBS8 1TDUK
- BrisSynBio/Bristol Biodesign InstituteUniversity of BristolLife Sciences Building, Tyndall AvenueBristolBS8 1TQUK
| | - Derek N. Woolfson
- School of ChemistryUniversity of BristolCantock's CloseBristolBS8 1TSUK
- School of BiochemistryUniversity of BristolMedical Sciences BuildingUniversity WalkBristolBS8 1TDUK
- BrisSynBio/Bristol Biodesign InstituteUniversity of BristolLife Sciences Building, Tyndall AvenueBristolBS8 1TQUK
| |
Collapse
|
87
|
Utterström J, Naeimipour S, Selegård R, Aili D. Coiled coil-based therapeutics and drug delivery systems. Adv Drug Deliv Rev 2021; 170:26-43. [PMID: 33378707 DOI: 10.1016/j.addr.2020.12.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/20/2022]
Abstract
Coiled coils are characterized by an arrangement of two or more α-helices into a superhelix and one of few protein motifs where the sequence-to-structure relationship to a large extent have been decoded and understood. The abundance of both natural and de novo designed coil coils provides a rich molecular toolbox for self-assembly of elaborate bespoke molecular architectures, nanostructures, and materials. Leveraging on the numerous possibilities to tune both affinities and preferences for polypeptide oligomerization, coiled coils offer unique possibilities to design modular and dynamic assemblies that can respond in a predictable manner to biomolecular interactions and subtle physicochemical cues. In this review, strategies to use coiled coils in design of novel therapeutics and advanced drug delivery systems are discussed. The applications of coiled coils for generating drug carriers and vaccines, and various aspects of using coiled coils for controlling and triggering drug release, and for improving drug targeting and drug uptake are described. The plethora of innovative coiled coil-based molecular systems provide new knowledge and techniques for improving efficacy of existing drugs and can facilitate development of novel therapeutic strategies.
Collapse
|
88
|
Lutz H, Popowski KD, Dinh PUC, Cheng K. Advanced Nanobiomedical Approaches to Combat Coronavirus Disease of 2019. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000063. [PMID: 33681865 PMCID: PMC7917381 DOI: 10.1002/anbr.202000063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
New infectious diseases are making themselves known as the human population grows, expands into new regions, and becomes more dense, increasing contact with each other and animal populations. Ease of travel has also increased infectious disease transmission and has now culminated into a global pandemic. The emergence of the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019 has already infected over 83.7 million people and caused over 1.8 million deaths. While there have been vaccine candidates produced and supportive care implemented, the world is impatiently waiting for a commercially approved vaccine and treatment for the coronavirus disease of 2019 (COVID-19). The different vaccine types investigated for the prevention of COVID-19 all have great promise but face safety obstacles that must be first addressed. Some vaccine candidates of key interest are whole inactivated viruses, adeno-associated viruses, virus-like particles, and lipid nanoparticles. This review examines nanobiomedical techniques for combatting COVID-19 in terms of vaccines and therapeutics.
Collapse
Affiliation(s)
- Halle Lutz
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
| | - Kristen D. Popowski
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
| | - Phuong-Uyen C. Dinh
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
| | - Ke Cheng
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill/North Carolina State UniversityRaleigh/Chapel HillNC27607/27599USA
- Division of Pharmacoengineering and Molecular PharmaceuticsUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| |
Collapse
|
89
|
Moran MC, Bence AR, Vallecillo MFS, Lützelschwab CM, Rodriguez MG, Pardo R, Goldbaum FA, Zylberman V, Palma SD, Maletto BA, Estein SM. Polymeric antigen BLSOmp31 formulated with class B CpG-ODN in a nanostructure (BLSOmp31/CpG-ODN/Coa-ASC16) administered by parenteral or mucosal routes confers protection against Brucella ovis in Balb/c mice. Res Vet Sci 2021; 135:217-227. [PMID: 33631456 DOI: 10.1016/j.rvsc.2021.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 01/22/2021] [Accepted: 02/14/2021] [Indexed: 01/18/2023]
Abstract
Previously, we demonstrated that the chimera BLSOmp31 formulated in chitosan microspheres or Poloxamer407-Chitosan administered via the nasal and the ocular mucosa conferred partial protection in sheep against B. ovis. In this work, we tested a new delivery system for mucosal immunization with BLSOmp31 in the murine model to improve the efficacy of previously used formulations. First, we evaluated the protective efficacy against B. ovis induced by BLSOmp31 administered by the subcutaneous route using either BLSOmp31 alone, co-administered with immunostimulatory synthetic oligodeoxynucleotides containing unmethylated cytosine-guanine motifs (CpG-ODN) or with CpG-ODN in a nanostructure called Coa-ASC16 compared with BLSOmp31 emulsified in Incomplete Freund Adjuvant. Then, we evaluated the protection conferred by the best performing formulation (BLSOmp31/CpG-ODN/Coa-ASC16) administered by both subcutaneous and ocular routes. BLSOmp31/CpG-ODN/Coa-ASC16 injected subcutaneously did not induce higher IgG antibody levels compared to BLSOmp31 alone or BLSOmp31/CpG-ODN but it did stimulate a mixed immune Th1-Th2 response with the highest levels of IFN-ɣ and conferred significant protection against the B. ovis challenge. Although ocular instillation of BLSOmp31/CpG-ODN/Coa-ASC16 showed a similar degree of protection compared to the parenteral route (3.66 and 3.60 logs of protection, respectively), it induced lower levels in serum of specific IgG (with mixed IgG1/IgG2a) and IgA antibodies and, less IFN-ɣ and IL-4 than the subcutaneous route. No antibodies were detected in vaginal lavages or saliva. Fecal antigen-specific IgA was slightly higher in mice immunized with BLSOmp31/CpG-ODN/Coa-ASC16 subcutaneously compared with the ocular route. These results indicate that BLSOmp31/CpG-ODN/Coa-ASC16 was a safe and effective vaccine against B. ovis in mice.
Collapse
Affiliation(s)
- María Celeste Moran
- Laboratorio de Inmunología, Departamento de Sanidad Animal y Medicina Preventiva (SAMP), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET-CICPBA), Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Buenos Aires, Argentina; Laboratorio de Microbiología Clínica y Experimental, Departamento SAMP, CIVETAN-CONICET-CICPBA., F.C.V, U.N.C.P.B.A., Tandil, Buenos Aires, Argentina
| | - Angel Ricardo Bence
- Laboratorio de Inmunología, Departamento de Sanidad Animal y Medicina Preventiva (SAMP), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET-CICPBA), Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Buenos Aires, Argentina; Departamento de Fisiopatología, F.C.V, U.N.C.P.B.A., Tandil, Buenos Aires., Argentina; Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CICPBA), Argentina
| | - María Fernanda Sánchez Vallecillo
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, CIBICI (CONICET), Córdoba, Argentina
| | - Claudia María Lützelschwab
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, SLU, Box 7028, SE-750-07, Uppsala, Sweden
| | | | | | | | | | - Santiago Daniel Palma
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, UNITEFA (CONICET), Córdoba, Argentina
| | - Belkys Angélica Maletto
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, CIBICI (CONICET), Córdoba, Argentina
| | - Silvia Marcela Estein
- Laboratorio de Inmunología, Departamento de Sanidad Animal y Medicina Preventiva (SAMP), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET-CICPBA), Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Buenos Aires, Argentina.
| |
Collapse
|
90
|
Mehmood I, Ijaz M, Ahmad S, Ahmed T, Bari A, Abro A, Allemailem KS, Almatroudi A, Tahir ul Qamar M. SARS-CoV-2: An Update on Genomics, Risk Assessment, Potential Therapeutics and Vaccine Development. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:1626. [PMID: 33567746 PMCID: PMC7915969 DOI: 10.3390/ijerph18041626] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a great threat to public health, being a causative pathogen of a deadly coronavirus disease (COVID-19). It has spread to more than 200 countries and infected millions of individuals globally. Although SARS-CoV-2 has structural/genomic similarities with the previously reported SARS-CoV and MERS-CoV, the specific mutations in its genome make it a novel virus. Available therapeutic strategies failed to control this virus. Despite strict standard operating procedures (SOPs), SARS-CoV-2 has spread globally and it is mutating gradually as well. Diligent efforts, special care, and awareness are needed to reduce transmission among susceptible masses particularly elder people, children, and health care workers. In this review, we highlighted the basic genome organization and structure of SARS-CoV-2. Its transmission dynamics, symptoms, and associated risk factors are discussed. This review also presents the latest mutations identified in its genome, the potential therapeutic options being used, and a brief explanation of vaccine development efforts against COVID-19. The effort will not only help readers to understand the deadly SARS-CoV-2 virus but also provide updated information to researchers for their research work.
Collapse
Affiliation(s)
- Iqra Mehmood
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (I.M.); (M.I.)
| | - Munazza Ijaz
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (I.M.); (M.I.)
| | - Sajjad Ahmad
- Department of Microbiology and Pharmacy, Abasyn University, Peshawar 25000, Pakistan;
| | - Temoor Ahmed
- Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China;
| | - Amna Bari
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China;
| | - Asma Abro
- Department of Biotechnology, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology Engineering and Management Sciences, Quetta 87100, Pakistan;
| | - Khaled S. Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | | |
Collapse
|
91
|
Kumar P, Sunita, Dubey KK, Shukla P. Whole-Cell Vaccine Preparation: Options and Perspectives. Methods Mol Biol 2021; 2183:249-266. [PMID: 32959248 DOI: 10.1007/978-1-0716-0795-4_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Vaccines are biological preparations to elicit a specific immune response in individuals against the targetted microorganisms. The use of vaccines has caused the near eradication of many critical diseases and has had an everlasting impact on public health at a relatively low cost. Most of the vaccines developed today are based on techniques which were developed a long time ago. In the beginning, vaccines were prepared from tissue fluids obtained from infected animals or people, but at present, the scenario has changed with the development of vaccines from live or killed whole microorganisms and toxins or using genetic engineering approaches. Considerable efforts have been made in vaccine development, but there are still many diseases that need attention, and new technologies are being developed in vaccinology to combat them. In this chapter, we discuss different approaches for vaccine development, including the properties and preparation of whole-cell vaccines.
Collapse
Affiliation(s)
- Punit Kumar
- Department of Biotechnology, University Institute of Engineering and Technology, Maharshi Dayanand University Rohtak, Rohtak, Haryana, India.,Department of Clinical Immunology, Allergology and Microbiology, Karaganda Medical University, 40 Gogol Street, Karaganda, Kazakhstan
| | - Sunita
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University Rohtak, Rohtak, Haryana, India
| | - Kashyap Kumar Dubey
- Department of Biotechnology, Central University of Haryana, Mahendergarh, Haryana, India.
| | - Pratyoosh Shukla
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University Rohtak, Rohtak, Haryana, India.
| |
Collapse
|
92
|
Kangabam R, Sahoo S, Ghosh A, Roy R, Silla Y, Misra N, Suar M. Next-generation computational tools and resources for coronavirus research: From detection to vaccine discovery. Comput Biol Med 2021; 128:104158. [PMID: 33301953 PMCID: PMC7705366 DOI: 10.1016/j.compbiomed.2020.104158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
The COVID-19 pandemic has affected 215 countries and territories around the world with 60,187,347 coronavirus cases and 17,125,719 currently infected patients confirmed as of the November 25, 2020. Currently, many countries are working on developing new vaccines and therapeutic drugs for this novel virus strain, and a few of them are in different phases of clinical trials. The advancement in high-throughput sequence technologies, along with the application of bioinformatics, offers invaluable knowledge on genomic characterization and molecular pathogenesis of coronaviruses. Recent multi-disciplinary studies using bioinformatics methods like sequence-similarity, phylogenomic, and computational structural biology have provided an in-depth understanding of the molecular and biochemical basis of infection, atomic-level recognition of the viral-host receptor interaction, functional annotation of important viral proteins, and evolutionary divergence across different strains. Additionally, various modern immunoinformatic approaches are also being used to target the most promiscuous antigenic epitopes from the SARS-CoV-2 proteome for accelerating the vaccine development process. In this review, we summarize various important computational tools and databases available for systematic sequence-structural study on coronaviruses. The features of these public resources have been comprehensively discussed, which may help experimental biologists with predictive insights useful for ongoing research efforts to find therapeutics against the infectious COVID-19 disease.
Collapse
Affiliation(s)
- Rajiv Kangabam
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India
| | - Susrita Sahoo
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India
| | - Arpan Ghosh
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India; School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India
| | - Riya Roy
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India
| | - Yumnam Silla
- Advanced Computation and Data Sciences Division, CSIR-North East Institute of Science and Technology (CSIR-NEIST), Jorhat, 785006, India
| | - Namrata Misra
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India; School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India
| | - Mrutyunjay Suar
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India; School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, 751024, India.
| |
Collapse
|
93
|
Garcia-Del Rio L, Diaz-Rodriguez P, Landin M. Design of novel orotransmucosal vaccine-delivery platforms using artificial intelligence. Eur J Pharm Biopharm 2020; 159:36-43. [PMID: 33383169 DOI: 10.1016/j.ejpb.2020.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022]
Abstract
The linings of the oral cavity are excellent needle-free vaccination sites, able to induce immune responses at distal sites and confer systemic protection. However, owing to the mucosal tissues' intrinsic characteristics, the design of effective antigen-delivery systems is not an easy task. In the present work, we propose to develop and characterize thermosensitive and mucoadhesive hydrogels for orotransmucosal vaccination taking advantage of artificial intelligence tools (AIT). Hydrogels of variable composition were obtained combining Pluronic® F127 (PF127), Hybrane® S1200 (HS1200) and Gantrez® AN119 (AN119) or S97 (S97). Systems were characterized in terms of physicochemical properties, adhesion capacity to mucosal tissues and antigen-like microspheres release. Additionally, polymers biocompatibility and their immune-stimulation capacity was assessed in human macrophages. Interestingly, cells treated with HS1200 exhibited a significant proliferation enhancement compared to control. The use of AIT allowed to determine the effect of each polymer on formulations properties. The proportions of PF127 and Gantrez® are mainly the factors controlling gelation temperature, mucoadhesion, adhesion work and gel strength. Meanwhile, cohesion and short-term microsphere release are dependent on the PF127 concentration. However, long-term microsphere release varies depending on the Gantrez® variety and the PF127 concentration used. Hydrogels prepared with S97 showed slower microsphere release. The use of AIT allowed to establish the conditions able to produce ternary hydrogels with immune-stimulatory properties together with adequate mucoadhesion capacity and antigen-like microspheres release.
Collapse
Affiliation(s)
- Lorena Garcia-Del Rio
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), AeMat, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain
| | - Patricia Diaz-Rodriguez
- Departamento de Ingeniería Química y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de La Laguna, Campus Anchieta, La Laguna 38200, Spain; Institute of Biomedical Technologies (ITB), Center for Biomedical Research of the Canary Islands (CIBICAN), Universidad de La Laguna, 38200 La Laguna, Spain.
| | - Mariana Landin
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), AeMat, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain
| |
Collapse
|
94
|
Lee YJ, Han Y, Joo HG. Bordetella bronchiseptica is a potent and safe adjuvant that enhances the antigen-presenting capability of dendritic cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:47-52. [PMID: 31908574 PMCID: PMC6940501 DOI: 10.4196/kjpp.2020.24.1.47] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 01/01/2023]
Abstract
We previously demonstrated that Bordetella bronchiseptica (B. bronchiseptica) antigen (Ag) enhances the Mycoplasma hyopneumoniae Ag-specific immune response. The focus of this study was whether acellular bacterin of B. bronchiseptica could be used as an adjuvant to increase antigen-presenting capability of dendritic cells (DCs) by increasing the level of activation. The metabolic activity of DCs was increased by B. bronchiseptica, similar to lipopolysaccharide (LPS). Flow cytometry analysis revealed that B. bronchiseptica increases the expression of major histocompatibility complex class-2, cluster of differentiation (CD)40, CD54, and CD86 which are closely related to DC-mediated immune responses. B. bronchiseptica enhanced the production of cytokines related to adaptive immune responses. Furthermore, the survival rate of B. bronchiseptica-injected groups was 100% at 15 and 20 mg/kg doses, whereas that of LPS-injected groups was only 20%, 0% at 15 and 20 mg/kg doses respectively, and so B. bronchiseptica is likely to be safer than LPS. Taken together, these results indicate that B. bronchiseptica can be used as an adjuvant to enhance the antigen-presenting capability of DCs. B. bronchiseptica is a candidate for producing vaccines, especially in case of DC-mediating efficacy and safety demands. This study provides researchers and clinicians with valuable information regarding the usage of B. bronchiseptica as a safe bacteria-derived immunostimulating agent for developing efficient vaccines.
Collapse
Affiliation(s)
- You-Jeong Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Yong Han
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Hong-Gu Joo
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea.,Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
95
|
Cossette B, Kelly SH, Collier JH. Intranasal Subunit Vaccination Strategies Employing Nanomaterials and Biomaterials. ACS Biomater Sci Eng 2020; 7:1765-1779. [DOI: 10.1021/acsbiomaterials.0c01291] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Benjamin Cossette
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| | - Sean H. Kelly
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| | - Joel H. Collier
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| |
Collapse
|
96
|
Han HJ, Nwagwu C, Anyim O, Ekweremadu C, Kim S. COVID-19 and cancer: From basic mechanisms to vaccine development using nanotechnology. Int Immunopharmacol 2020; 90:107247. [PMID: 33307513 PMCID: PMC7709613 DOI: 10.1016/j.intimp.2020.107247] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022]
Abstract
Shows updated understanding of SARS-CoV-2, including the interaction between ACE 2 and the viral spike protein. More effective vaccines are required for immunocompromised cancer patients. Cancer alters the immune system through different levels of D-Dimer, albumin, prothrombin, and neutrophils. Nanomaterials assist vaccine delivery, including viral vector and mRNA vaccines with lipid nanoparticles.
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global pandemic which has induced unprecedented ramifications, severely affecting our society due to the long incubation time, unpredictably high prevalence and lack of effective vaccines. One of the interesting notions is that there is an association between COVID-19 and cancer. Cancer patients seem to exhibit exacerbated conditions and a higher mortality rate when exposed to the virus. Therefore, vaccines are the promising solution to minimise the problem amongst cancer patients threatened by the new viral strains. However, there are still limitations to be considered, including the efficacy of COVID vaccines for immunocompromised individuals, possible interactions between the vaccine and cancer, and personalised medicine. Not only to eradicate the pandemic, but also to make it more effective for immunocompromised patients who are suffering from cancer, a successful vaccine platform is required through the implementation of nanotechnology which can also enable scalable manufacturing and worldwide distribution along with its faster and precise delivery. In this review, we summarise the current understanding of COVID-19 with clinical perspectives, highlighting the association between COVID-19 and cancer, followed by a vaccine development for this association using nanotechnology. We suggest different administration methods for the COVID-19 vaccine formulation options. This study will contribute to paving the way towards the prevention and treatment of COVID-19, especially for the immunocompromised individuals.
Collapse
Affiliation(s)
- Hyun Jee Han
- University College London, Department of Neonatology, United Kingdom.
| | - Chinekwu Nwagwu
- Department of Pharmaceutics, University of Nigeria Nsukka, Nigeria.
| | - Obumneme Anyim
- Department of Internal Medicine, University of Nigeria Teaching Hospital Ituku-Ozalla, Enugu, Nigeria
| | - Chinedu Ekweremadu
- Department of Pharmaceutics and Pharmaceutical Technology Enugu State University of Science and Technology, Nigeria.
| | - San Kim
- Basildon and Thurrock University Hospital, United Kingdom.
| |
Collapse
|
97
|
Russell RL, Pelka P, Mark BL. Frontrunners in the race to develop a SARS-CoV-2 vaccine. Can J Microbiol 2020; 67:189-212. [PMID: 33264067 DOI: 10.1139/cjm-2020-0465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Numerous studies continue to be published on the COVID-19 pandemic that is being caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Given the rapidly evolving global response to SARS-CoV-2, here we primarily review the leading COVID-19 vaccine strategies that are currently in Phase III clinical trials. Nonreplicating viral vector strategies, inactivated virus, recombinant protein subunit vaccines, and nucleic acid vaccine platforms are all being pursued in an effort to combat the infection. Preclinical and clinal trial results of these efforts are examined as well as the characteristics of each vaccine strategy from the humoral and cellular immune responses they stimulate, effects of any adjuvants used, and the potential risks associated with immunization such as antibody-dependent enhancement. A number of promising advancements have been made toward the development of multiple vaccine candidates. Preliminary data now emerging from phase III clinical trials show encouraging results for the protective efficacy and safety of at least 3 frontrunning candidates. There is hope that one or more will emerge as potent weapons to protect against SARS-CoV-2.
Collapse
Affiliation(s)
- Raquel L Russell
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Peter Pelka
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Brian L Mark
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
98
|
A novel lamprey antibody sequence to multimerize and increase the immunogenicity of recombinant viral and bacterial vaccine antigens. Vaccine 2020; 38:7905-7915. [PMID: 33153770 DOI: 10.1016/j.vaccine.2020.10.073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022]
Abstract
Hemagglutinin, the major surface protein of influenza viruses, was recombinantly expressed in eukaryotic cells as a monomer instead of its native trimer, and was only immunogenic when administered with an adjuvant [Pion et al. 2014]. In order to multimerize this antigen to increase its immunogenicity, a cysteine-rich peptide sequence found at the extreme C-terminus of lamprey variable lymphocyte receptor (VLR)-B antibodies was fused to various recombinant hemagglutinin (rHA) proteins from A and B influenza virus strains. The rHA-Lamp fusion (rHA fused to the lamprey sequence) protein was expressed in Leishmania tarentolae and Chinese hamster ovary (CHO) cells and shown to produce several multimeric forms. The multimers produced were very stable and more immunogenic in mice than monomeric rHA. The lamprey VLR-B sequence was also used to multimerize the neuraminidase (NA) of influenza viruses expressed in CHO cells. For some viral strains, the NA was expressed as a tetramer like the native viral NA form. In addition, the lamprey VLR-B sequence was fused with two surface antigens of Shigella flexneri 2a, the invasion plasmid antigen D and a double mutated soluble form of the membrane expression of the invasion plasmid antigen H namely MxiH. The fusion proteins were expressed in Escherichia coli to produce the respective multimer protein forms. The resulting proteins had similar multimeric forms as rHA-Lamp protein and were more immunogenic in mice than the monomer forms. In conclusion, the VLR-B sequence can be used to increase the immunogenicity of recombinant viral and bacterial antigens, thus negating the need for adjuvants.
Collapse
|
99
|
Silvane L, Celias DP, Romagnoli PA, Maletto BA, Sanchez Vallecillo MF, Chiapello LS, Palma SD, Allemandi DA, Sanabria REF, Pruzzo CI, Motrán CC, Cervi L. A Vaccine Based on Kunitz-Type Molecule Confers Protection Against Fasciola hepatica Challenge by Inducing IFN-γ and Antibody Immune Responses Through IL-17A Production. Front Immunol 2020; 11:2087. [PMID: 33193292 PMCID: PMC7641617 DOI: 10.3389/fimmu.2020.02087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/31/2020] [Indexed: 01/03/2023] Open
Abstract
Fasciola hepatica is helminth parasite found around the world that causes fasciolosis, a chronic disease affecting mainly cattle, sheep, and occasionally humans. Triclabendazole is the drug of choice to treat this parasite. However, the continuous use of this drug has led to the development of parasite resistance and, consequently, the limitation of its effectiveness. Hence, vaccination appears as an attractive option to develop. In this work, we evaluated the potential of F. hepatica Kunitz-type molecule (FhKTM) as an antigen formulated with a liquid crystal nanostructure formed by self-assembly of 6-O-ascorbyl palmitate ester (Coa-ASC16) and the synthetic oligodeoxynucleotide containing unmethylated cytosine-guanine motifs (CpG-ODN) during an experimental model of fasciolosis in mice, and we further dissected the immune response associated with host protection. Our results showed that immunization of mice with FhKTM/CpG-ODN/Coa-ASC16 induces protection against F. hepatica challenge by preventing liver damage and improving survival after F. hepatica infection. FhKTM/CpG-ODN/Coa-ASC16-immunized mice elicited potent IFN-γ and IL-17A with high levels of antigen-specific IgG1, IgG2a, and IgA serum antibodies. Strikingly, IL-17A blockade during infection decreased IgG2a and IgA antibody levels as well as IFN-γ production, leading to an increase in mortality of vaccinated mice. The present study highlights the potential of a new vaccine formulation to improve control and help the eradication of F. hepatica infection, with potential applications for natural hosts such as cattle and sheep.
Collapse
Affiliation(s)
- Leonardo Silvane
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Daiana Pamela Celias
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Pablo Alberto Romagnoli
- Centro de Investigación en Medicina Traslacional Severo Amuchastegui (CIMETSA), Córdoba, Argentina.,Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Belkys Angélica Maletto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - María Fernanda Sanchez Vallecillo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Silvina Chiapello
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Santiago Daniel Palma
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Unidad de Investigación y desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Daniel Alberto Allemandi
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Unidad de Investigación y desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Rodrigo Eduardo Fabrizio Sanabria
- Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, La Plata, Argentina.,Instituto Tecnológico Chascomús (INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín (CONICET/UNSAM), Chascomús, Argentina
| | - César Iván Pruzzo
- Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, La Plata, Argentina
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| |
Collapse
|
100
|
Chen L, Liu B, Sun P, Wang W, Luo S, Zhang W, Yang Y, Wang Z, Lin J, Chen PR. Severe Acute Respiratory Syndrome Coronavirus-2 Spike Protein Nanogel as a Pro-Antigen Strategy with Enhanced Protective Immune Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004237. [PMID: 33107685 PMCID: PMC7645880 DOI: 10.1002/smll.202004237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/10/2020] [Indexed: 06/11/2023]
Abstract
Prevention and intervention methods are urgently needed to curb the global pandemic of coronavirus disease-19 caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Herein, a general pro-antigen strategy for subunit vaccine development based on the reversibly formulated receptor binding domain of SARS-CoV-2 spike protein (S-RBD) is reported. Since the poor lymph node targeting and uptake of S-RBD by antigen-presenting cells prevent effective immune responses, S-RBD protein is formulated into a reversible nanogel (S-RBD-NG), which serves as a pro-antigen with enhanced lymph node targeting and dendritic cell and macrophage accumulation. Synchronized release of S-RBD monomers from the internalized S-RBD-NG pro-antigen triggers more potent immune responses in vivo. In addition, by optimizing the adjuvant used, the potency of S-RBD-NG is further improved, which may provide a generally applicable, safer, and more effective strategy for subunit vaccine development against SARS-CoV-2 as well as other viruses.
Collapse
Affiliation(s)
- Long Chen
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
| | - Bo Liu
- Department of Microorganism EngineeringBeijing Institute of BiotechnologyBeijing100071China
| | - Peng Sun
- Department of Microorganism EngineeringBeijing Institute of BiotechnologyBeijing100071China
| | - Wenjun Wang
- Key Laboratory of Infection and ImmunityInstitute of BiophysicsChinese Academy of SciencesCollege of Life SciencesUniversity of the Chinese Academy of SciencesBeijing100101China
| | - Shiqiang Luo
- Department of Microorganism EngineeringBeijing Institute of BiotechnologyBeijing100071China
- Institute of Physical Science and InformationAnhui UniversityHefei230601China
| | - Wenyuan Zhang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
| | - Yuanfan Yang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
| | - Zihao Wang
- Beijing Institute of Pharmacology and ToxicologyBeijing100850China
| | - Jian Lin
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
| | - Peng R. Chen
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| |
Collapse
|