51
|
Dong C, Wang BZ. Engineered Nanoparticulate Vaccines to Combat Recurring and Pandemic Influenza Threats. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100122. [PMID: 35754779 PMCID: PMC9231845 DOI: 10.1002/anbr.202100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Reoccurring seasonal flu epidemics and occasional pandemics are among the most severe threats to public health. Current seasonal influenza vaccines provide limited protection against drifted circulating strains and no protection against influenza pandemics. Next-generation influenza vaccines, designated as universal influenza vaccines, should be safe, affordable, and elicit long-lasting cross-protective influenza immunity. Nanotechnology plays a critical role in the development of such novel vaccines. Engineered nanoparticles can incorporate multiple advantageous properties into the same nanoparticulate platforms to improve vaccine potency and breadth. These immunological properties include virus-like biomimicry, high antigen-load, controlled antigen release, targeted delivery, and induction of innate signaling pathways. Many nanoparticle influenza vaccines have shown promising results in generating potent and broadly protective immune responses. This review will summarize the necessity and characteristics of next-generation influenza vaccines and the immunological correlates of broad influenza immunity and focus on how cutting-edge nanoparticle technology contributes to such vaccine development. The review will give new insights into the rational design of nanoparticle universal vaccines to combat influenza epidemics and pandemics.
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| |
Collapse
|
52
|
Spatial, Temporal, and Demographic Patterns in the Prevalence of Hemorrhagic Septicemia in 41 Countries in 2005–2019: A Systematic Analysis with Special Focus on the Potential Development of a New-Generation Vaccine. Vaccines (Basel) 2022; 10:vaccines10020315. [PMID: 35214771 PMCID: PMC8880277 DOI: 10.3390/vaccines10020315] [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: 12/27/2021] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 01/10/2023] Open
Abstract
Hemorrhagic septicemia (HS) caused by Pasteurella multocida B:2 and E:2 is among the fatal bacterial diseases in cattle and buffaloes that are economically valuable in Asian and African countries. The current work aims to study the prevalence of HS among buffaloes, cattle, sheep, and goats in 41 countries in 2005–2019. The data analysis revealed that 74.4% of the total infection rate in the world was distributed among cattle, followed by buffaloes (13.1%). The mortality of HS among cattle and buffaloes increased in 2017–2019 compared to the period between 2014 and 2016. The best measure to control the disease is through vaccination programs. Current commercial vaccines, including live-attenuated vaccines and inactivated vaccines, have some shortcomings and undesirable effects. Virus-like particles (VLPs) have more potential as a vaccine platform due to their unique properties to enhance immune response and the ability to use them as a platform for foreign antigens against infectious diseases. VLPs-based vaccines are among the new-generation subunit vaccine approaches that have been licensed for the human and veterinary fields. However, most studies are still in the late stages of vaccine evaluation.
Collapse
|
53
|
Bhat T, Cao A, Yin J. Virus-like Particles: Measures and Biological Functions. Viruses 2022; 14:383. [PMID: 35215979 PMCID: PMC8877645 DOI: 10.3390/v14020383] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/10/2022] [Accepted: 02/10/2022] [Indexed: 12/25/2022] Open
Abstract
Virus-like particles resemble infectious virus particles in size, shape, and molecular composition; however, they fail to productively infect host cells. Historically, the presence of virus-like particles has been inferred from total particle counts by microscopy, and infectious particle counts or plaque-forming-units (PFUs) by plaque assay; the resulting ratio of particles-to-PFUs is often greater than one, easily 10 or 100, indicating that most particles are non-infectious. Despite their inability to hijack cells for their reproduction, virus-like particles and the defective genomes they carry can exhibit a broad range of behaviors: interference with normal virus growth during co-infections, cell killing, and activation or inhibition of innate immune signaling. In addition, some virus-like particles become productive as their multiplicities of infection increase, a sign of cooperation between particles. Here, we review established and emerging methods to count virus-like particles and characterize their biological functions. We take a critical look at evidence for defective interfering virus genomes in natural and clinical isolates, and we review their potential as antiviral therapeutics. In short, we highlight an urgent need to better understand how virus-like genomes and particles interact with intact functional viruses during co-infection of their hosts, and their impacts on the transmission, severity, and persistence of virus-associated diseases.
Collapse
Affiliation(s)
| | | | - John Yin
- Department of Chemical and Biological Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, 330 N. Orchard Street, Madison, WI 53715, USA; (T.B.); (A.C.)
| |
Collapse
|
54
|
Dong C, Wang Y, Zhu W, Ma Y, Kim J, Wei L, Gonzalez GX, Wang BZ. Polycationic HA/CpG Nanoparticles Induce Cross-Protective Influenza Immunity in Mice. ACS APPLIED MATERIALS & INTERFACES 2022; 14:6331-6342. [PMID: 35084819 PMCID: PMC8832387 DOI: 10.1021/acsami.1c19192] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 05/28/2023]
Abstract
The intranasal (i.n.) route is an ideal vaccination approach for infectious respiratory diseases like influenza. Polycationic polyethylenimine (PEI) could form nanoscale complexes with negatively charged viral glycoproteins. Here we fabricated PEI-hemagglutinin (HA) and PEI-HA/CpG nanoparticles and investigated their immune responses and protective efficacies with an i.n. vaccination regimen in mice. Our results revealed that the nanoparticles significantly enhanced HA immunogenicity, providing heterologous cross-protection. The conserved HA stalk region induced substantial antibodies in the nanoparticle immunization groups. In contrast to the Th2-biased, IgG1-dominant antibody response generated by PEI-HA nanoparticles, PEI-HA/CpG nanoparticles generated more robust and balanced IgG1/IgG2a antibody responses with augmented neutralization activity and Fc-mediated antibody-dependent cellular cytotoxicity (ADCC). PEI-HA/CpG nanoparticles also induced enhanced local and systemic cellular immune responses. These immune responses did not decay over six months of observation postimmunization. PEI and CpG synergized these comprehensive immune responses. Thus, the PEI-HA/CpG nanoparticle is a potential cross-protective influenza vaccine candidate. Polycationic PEI nanoplatforms merit future development into mucosal vaccine systems.
Collapse
|
55
|
Utomo DIS, Pambudi S, Park EY. Humoral immune response induced with dengue virus-like particles serotypes 1 and 4 produced in silkworm. AMB Express 2022; 12:8. [PMID: 35102445 PMCID: PMC8802989 DOI: 10.1186/s13568-022-01353-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/22/2022] [Indexed: 11/12/2022] Open
Abstract
Dengue is an arboviral disease, which threatens almost half the global population, and has emerged as the most significant of current global public health challenges. In this study, we prepared dengue virus-like particles (DENV-LPs) consisting of Capsid-premembrane-envelope (CprME) and premembrane-envelope (prME) polypeptides from serotype 1 and 4, which were expressed in the silkworms using Bombyx mori nucleopolyhedrovirus (BmNPV) bacmid. 1CprME, 1prME, 4CprME, and 4prME expressed proteins in hemolymph, and the molecular weight of the purified proteins was 55 kDa, respectively. The purified polypeptides formed spherical Dengue virus-like particles (DENV-LPs) with ~ 30–55 nm in diameter. The immunoelectron microscopy (IEM) images revealed antigens to the surface of a lipid bilayer of DENV-LPs. The heparin-binding assay shows a positive relationship between absorbance and E protein domain III (EDIII) quantity, which is supported by the isothermal titration calorimetry assay. This indicates a moderate binding affinity between heparin and DENV-LP. The high correlation between patient sera and DENV-LP reactivities revealed that these DENV-LPs shared similar epitopes with the natural dengue virus. IgG elicitation studies in mice have demonstrated that DENV-LPs/CPrMEs elicit a stronger immune response than DENV-LP/prMEs, which lends credence to this claim. Dengue virus-like particles for serotype 1 and serotype 4 (DENV-LPs/1 and DENV-LPs/4) were produced in silkworm. Heparin-binding assay by ELISA and ITC showed that DENV-LPs/1 and DENV-LPs/4 contain Envelope Domain III. DENV-LPs/1 and DENV-LPs/4 showed affinity to sera from human dengue patients and immunized mice.
Collapse
|
56
|
McFall-Boegeman H, Huang X. Mechanisms of cellular and humoral immunity through the lens of VLP-based vaccines. Expert Rev Vaccines 2022; 21:453-469. [PMID: 35023430 PMCID: PMC8960355 DOI: 10.1080/14760584.2022.2029415] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Vaccination can be effective defense against many infectious agents and the corresponding diseases. Discoveries elucidating the mechanisms of the immune system have given hopes to developing vaccines against diseases recalcitrant to current treatment/prevention strategies. One such finding is the ability of immunogenic biological nanoparticles to powerfully boost the immunogenicity of poorer antigens conjugated to them with virus-like particle (VLP)-based vaccines as a key example. VLPs take advantage of the well-defined molecular structures associated with sub-unit vaccines and the immunostimulatory nature of conjugate vaccines. AREAS COVERED In this review, we will discuss how advances in understanding the immune system can inform VLP-based vaccine design and how VLP-based vaccines have uncovered underlying mechanisms in the immune system. EXPERT OPINION As our understanding of mechanisms underlying the immune system increases, that knowledge should inform our vaccine design. Testing of proof-of-concept vaccines in the lab should seek to elucidate the underlying mechanisms of immune responses. The integration of these approaches will allow for VLP-based vaccines to live up to their promise as a powerful plug-and-play platform for next generation vaccine development.
Collapse
Affiliation(s)
- Hunter McFall-Boegeman
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA.,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
57
|
Taha EA, Lee J, Hotta A. Delivery of CRISPR-Cas tools for in vivo genome editing therapy: Trends and challenges. J Control Release 2022; 342:345-361. [PMID: 35026352 DOI: 10.1016/j.jconrel.2022.01.013] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
The discovery of clustered regularly interspaced short palindromic repeats (CRISPR) genome editing technology opened the door to provide a versatile approach for treating multiple diseases. Promising results have been shown in numerous pre-clinical studies and clinical trials. However, a safe and effective method to deliver genome-editing components is still a key challenge for in vivo genome editing therapy. Adeno-associated virus (AAV) is one of the most commonly used vector systems to date, but immunogenicity against capsid, liver toxicity at high dose, and potential genotoxicity caused by off-target mutagenesis and genomic integration remain unsolved. Recently developed transient delivery systems, such as virus-like particle (VLP) and lipid nanoparticle (LNP), may solve some of the issues. This review summarizes existing in vivo delivery systems and possible solutions to overcome their limitations. Also, we highlight the ongoing clinical trials for in vivo genome editing therapy and recently developed genome editing tools for their potential applications.
Collapse
Affiliation(s)
- Eman A Taha
- Center for iPS cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Biochemistry, Ain Shams University Faculty of Science, Cairo 11566, Egypt
| | - Joseph Lee
- Center for iPS cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Takeda-CiRA Joint Program (T-CiRA), Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
58
|
Mohsen MO, Balke I, Zinkhan S, Zeltina V, Liu X, Chang X, Krenger PS, Plattner K, Gharailoo Z, Vogt AS, Augusto G, Zwicker M, Roongta S, Rothen DA, Josi R, da Costa JJ, Sobczak JM, Nonic A, Brand L, Nuss K, Martina B, Speiser DE, Kündig T, Jennings GT, Walton SM, Vogel M, Zeltins A, Bachmann MF. A scalable and highly immunogenic virus-like particle-based vaccine against SARS-CoV-2. Allergy 2022; 77:243-257. [PMID: 34496033 PMCID: PMC8653185 DOI: 10.1111/all.15080] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND SARS-CoV-2 caused one of the most devastating pandemics in the recent history of mankind. Due to various countermeasures, including lock-downs, wearing masks, and increased hygiene, the virus has been controlled in some parts of the world. More recently, the availability of vaccines, based on RNA or adenoviruses, has greatly added to our ability to keep the virus at bay; again, however, in some parts of the world only. While available vaccines are effective, it would be desirable to also have more classical vaccines at hand for the future. Key feature of vaccines for long-term control of SARS-CoV-2 would be inexpensive production at large scale, ability to make multiple booster injections, and long-term stability at 4℃. METHODS Here, we describe such a vaccine candidate, consisting of the SARS-CoV-2 receptor-binding motif (RBM) grafted genetically onto the surface of the immunologically optimized cucumber mosaic virus, called CuMVTT -RBM. RESULTS Using bacterial fermentation and continuous flow centrifugation for purification, the yield of the production process is estimated to be >2.5 million doses per 1000-litre fermenter run. We demonstrate that the candidate vaccine is highly immunogenic in mice and rabbits and induces more high avidity antibodies compared to convalescent human sera. The induced antibodies are more cross-reactive to mutant RBDs of variants of concern (VoC). Furthermore, antibody responses are neutralizing and long-lived. In addition, the vaccine candidate was stable for at least 14 months at 4℃. CONCLUSION Thus, the here presented VLP-based vaccine may be a good candidate for use as conventional vaccine in the long term.
Collapse
|
59
|
Naz SS, Munir I. An Outline of Contributing Vaccine Technologies for SARS CoV2 Advancing in Clinical and Preclinical Phase-Trials. Recent Pat Biotechnol 2022; 16:122-143. [PMID: 35040422 DOI: 10.2174/1872208316666220118094344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/11/2021] [Accepted: 11/27/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Severe Acute Respiratory Syndrome Coronavirus 2 (SARS CoV2) is an RNA virus involving 4 structural and 16 non-structural proteins, and exhibiting high transmission potential and fatality. The emergence of this newly encountered beta coronavirus-SARS CoV2 has brought over 2 million people to death, and more than 10 billion people got infected across the globe as yet. Consequently, the global scientific community has contributed to the synthesis and design of effective immunization technologies to combat this virus. OBJECTIVES This literature review was intended to gather an update on published reports of the vaccines advancing in the clinical trial phases or preclinical trials, to summarize the foundations and implications of contributing vaccine candidates inferring their impact in the pandemic repression. In addition, this literature review distinctly facilitates an outline of the overall vaccine effectiveness at current doses. METHODS The reported data in this review was extracted from research articles, review articles and patents published from January 2020 to July 2021, available on Google Scholar, Pubmed, Pubmed Central, Research Gate, Science direct, and Free Patent Online Database by using combination of keywords. Moreover, some information is retrieved from native web pages of vaccine manufacturing companies' due to progressing research and unavailability of published research papers. CONCLUSION Contributing vaccine technologies include: RNA (Ribonucleic acid) vaccines, DNA (Deoxyribonucleic acid) vaccines, viral vector vaccines, protein-based vaccines, inactivated vaccines, viruses-like particles, protein superglue, and live-attenuated vaccines. Some vaccines are prepared by establishing bacterial and yeast cell lines and as self-assembling adenovirus- derived multimeric protein-based self-assembling nanoparticle (ADDOmer). On May 19, WHO has issued an emergency use sanction of Moderna, Pfizer, Sinopharm, AstraZeneca, and Covishield vaccine candidates on account of clinical credibility from experimental data.
Collapse
Affiliation(s)
- Sheikh Saba Naz
- Department of Microbiology, Jinnah University for Women, Pakistan
| | - Iqra Munir
- Department of Microbiology, Jinnah University for Women, Pakistan
- National Nanotechnology Research Center-UNAM, Bilkent University, Turkey
| |
Collapse
|
60
|
Chen Z, Gao X, Yu D. Longevity of vaccine protection: Immunological mechanism, assessment methods, and improving strategy. VIEW 2022. [DOI: 10.1002/viw.20200103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Zhian Chen
- The University of Queensland Diamantina Institute, Faculty of Medicine The University of Queensland Brisbane Queensland Australia
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| | - Xin Gao
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine The University of Queensland Brisbane Queensland Australia
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| |
Collapse
|
61
|
Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine Delivery Approaches and Advanced Delivery Systems for the Prevention of Viral Infections: From Development to Clinical Application. Pharmaceutics 2021; 13:2091. [PMID: 34959372 PMCID: PMC8707864 DOI: 10.3390/pharmaceutics13122091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections causing pandemics and chronic diseases are the main culprits implicated in devastating global clinical and socioeconomic impacts, as clearly manifested during the current COVID-19 pandemic. Immunoprophylaxis via mass immunisation with vaccines has been shown to be an efficient strategy to control such viral infections, with the successful and recently accelerated development of different types of vaccines, thanks to the advanced biotechnological techniques involved in the upstream and downstream processing of these products. However, there is still much work to be done for the improvement of efficacy and safety when it comes to the choice of delivery systems, formulations, dosage form and route of administration, which are not only crucial for immunisation effectiveness, but also for vaccine stability, dose frequency, patient convenience and logistics for mass immunisation. In this review, we discuss the main vaccine delivery systems and associated challenges, as well as the recent success in developing nanomaterials-based and advanced delivery systems to tackle these challenges. Manufacturing and regulatory requirements for the development of these systems for successful clinical and marketing authorisation were also considered. Here, we comprehensively review nanovaccines from development to clinical application, which will be relevant to vaccine developers, regulators, and clinicians.
Collapse
Affiliation(s)
- Ana Sara Cordeiro
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Yogita Patil-Sen
- Wrightington, Wigan and Leigh Teaching Hospitals NHS Foundation Trust, National Health Service, Wigan WN6 0SZ, UK;
| | - Maitreyi Shivkumar
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Ronak Patel
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Abdulwahhab Khedr
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A. Elsawy
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| |
Collapse
|
62
|
Marfe G, Perna S, Shukla AK. Effectiveness of COVID-19 vaccines and their challenges (Review). Exp Ther Med 2021; 22:1407. [PMID: 34676000 PMCID: PMC8524740 DOI: 10.3892/etm.2021.10843] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
At the end of 2019, a new disease recognized such as severe acute respiratory syndrome (SARS), was reported in Wuhan, China. This disease was caused by an unknown SARS coronavirus 2 (SARS-CoV-2); a virus is characterized by high infectivity among humans. In some cases, this disease can be asymptomatic, while in other cases can induce flu-like symptoms or acute respiratory distress syndrome, pneumonia and death. For this reason, the World Health Organization and Public Health Emergency of International Concern declared a pandemic status in January 2020. Currently, numerous countries have been involved in the development of effective vaccines to protect humans against SARS-CoV-2 infection. The present review will discuss the four vaccines, AZD1222 (AstraZeneca or Vaxzevria), Janssen (Ad26.COV2.S), Moderna/mRNA-1273 and BioNTech/Fosun/Pfizer BNT162b1, that are currently in use worldwide to understand their efficacy, but also evaluate the difficulties and challenges of vaccine development. Although several questions should be addressed regarding these vaccines, the current review will examine the viral elements used in the coronavirus-19 vaccine that can play a crucial role in inducing a strong immune response, as well as the different adverse effects that they can cause to individuals.
Collapse
Affiliation(s)
- Gabriella Marfe
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Stefania Perna
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
- Inventra Medclin Biomedical Healthcare and Research Center, Katemanivli, Kalyan, Thane, Maharashtra 421306, India
| |
Collapse
|
63
|
González-Domínguez I, Lorenzo E, Bernier A, Cervera L, Gòdia F, Kamen A. A Four-Step Purification Process for Gag VLPs: From Culture Supernatant to High-Purity Lyophilized Particles. Vaccines (Basel) 2021; 9:vaccines9101154. [PMID: 34696262 PMCID: PMC8539588 DOI: 10.3390/vaccines9101154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 01/15/2023] Open
Abstract
Gag-based virus-like particles (VLPs) have high potential as scaffolds for the development of chimeric vaccines and delivery strategies. The production of purified preparations that can be preserved independently from cold chains is highly desirable to facilitate distribution and access worldwide. In this work, a nimble purification has been developed, facilitating the production of Gag VLPs. Suspension-adapted HEK 293 cells cultured in chemically defined cell culture media were used to produce the VLPs. A four-step downstream process (DSP) consisting of membrane filtration, ion-exchange chromatography, polishing, and lyophilization was developed. The purification of VLPs from other contaminants such as host cell proteins (HCP), double-stranded DNA, or extracellular vesicles (EVs) was confirmed after their DSP. A concentration of 2.2 ± 0.8 × 109 VLPs/mL in the lyophilized samples was obtained after its storage at room temperature for two months. Morphology and structural integrity of purified VLPs was assessed by cryo-TEM and NTA. Likewise, the purification methodologies proposed here could be easily scaled up and applied to purify similar enveloped viruses and vesicles.
Collapse
Affiliation(s)
- Irene González-Domínguez
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
- Correspondence:
| | - Elianet Lorenzo
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
| | - Alice Bernier
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; (A.B.); (A.K.)
| | - Laura Cervera
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
| | - Francesc Gòdia
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; (A.B.); (A.K.)
| |
Collapse
|
64
|
Abstract
INTRODUCTION Vaccination is so far the most effective way of eradicating infections. Rapidly emerging drug resistance against infectious diseases and chemotherapy-related toxicities in cancer warrant immediate vaccine development to save mankind. Subunit vaccines alone, however, fail to elicit sufficiently strong and long-lasting protective immunity against deadly pathogens. Nanoparticle (NP)-based delivery vehicles like microemulsions, liposomes, virosomes, nanogels, micelles and dendrimers offer promising strategies to overcome limitations of traditional vaccine adjuvants. Nanovaccines can improve targeted delivery, antigen presentation, stimulation of body's innate immunity, strong T cell response combined with safety to combat infectious diseases and cancers. Further, nanovaccines can be highly beneficial to generate effective immutherapeutic formulations against cancer. AREAS COVERED This review summarizes the emerging nanoparticle strategies highlighting their success and challenges in preclinical and clinical trials in infectious diseases and cancer. It provides a concise overview of current nanoparticle-based vaccines, their adjuvant potential and their cellular delivery mechanisms. EXPERT OPINION The nanovaccines (50-250 nm in size) are most efficient in terms of tissue targeting, prolonged circulation and preferential uptake by the professional APCs chiefly due to their small size. More rational designing, improved antigen loading, extensive functionalization and targeted delivery are some of the future goals of ideal nanovaccines.
Collapse
Affiliation(s)
- Amrita Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
65
|
Jiménez-Chávez ÁDJ, Nava-García BK, Bustos-Jaimes I, Moreno-Fierros L. B19-VLPs as an effective delivery system for tumour antigens to induce humoral and cellular immune responses against triple negative breast cancer. Immunol Lett 2021; 239:77-87. [PMID: 34508790 DOI: 10.1016/j.imlet.2021.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/25/2021] [Accepted: 09/03/2021] [Indexed: 01/16/2023]
Abstract
Cancer immunotherapy is emerging as a viable treatment option for several types of cancer. Active immunotherapy aims for the induction of specific antitumor immune responses; this goal requires strategies capable of increasing the immunogenicity of tumour antigens. Parvovirus B19 virus-like particles (B19-VLPs) formed of VP2 protein had been shown to be an effective multi-neoepitope delivery system capable of inducing specific cellular responses towards coupled antigens and reducing tumour growth and lung metastases in triple negative breast cancer mouse model. These findings encouraged us to further characterise these VP2 B19-VLPs by testing their capacity to simultaneously induce cellular and humoral responses towards other tumour-associated antigens, as this had not yet been evaluated. Here, we designed and evaluated in the 4T1 breast cancer model the prophylactic and therapeutic effect of VP2 B19-VLPs decorated with cellular (P53) and humoral (MUC1) epitopes. Balb/c mice were immunised with chimaeric VLPs, vehicle, or VLPs plus adjuvant. Tumour establishment and growth, lung metastasis, and cellular and humoral immune responses were evaluated. The prophylactic administration of chimaeric VLPs without adjuvant prevented the establishment of the tumour, while by therapeutic administration, chimaeric VLPs induced smaller tumour growth and decreased the number of metastases in the lung compared to wild-type VLPs. chimaeric VLPs induced high antibody titres towards the MUC1 epitope, as well as specific cellular responses towards P53 epitopes in lymph nodes local to the tumour. Our results reinforce and extend the utility of VP2 B19-VLPs as an encouraging tumour antigen delivery system in cancer immunotherapy able to improve tumour immunity in TNBC by inducing cellular and humoral immune responses.
Collapse
Affiliation(s)
- Ángel de Jesús Jiménez-Chávez
- Biomedicine Research Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico. Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
| | - Brenda Katherine Nava-García
- Biomedicine Research Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico. Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
| | - Ismael Bustos-Jaimes
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Leticia Moreno-Fierros
- Biomedicine Research Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico. Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México.
| |
Collapse
|
66
|
Calvo Fernández E, Zhu LY. Racing to immunity: Journey to a COVID-19 vaccine and lessons for the future. Br J Clin Pharmacol 2021; 87:3408-3424. [PMID: 33289156 PMCID: PMC7753785 DOI: 10.1111/bcp.14686] [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: 10/19/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 is the novel coronavirus behind the COVID-19 pandemic. Since its emergence, the global scientific community has mobilized to study this virus, and an overwhelming effort to identify COVID-19 treatments is currently ongoing for a variety of therapeutics and prophylactics. To better understand these efforts, we compiled a list of all COVID-19 vaccines undergoing preclinical and clinical testing using the WHO and ClinicalTrials.gov database, with details surrounding trial design and location. The most advanced vaccines are discussed in more detail, with a focus on their technology, advantages and disadvantages, as well as any available recent clinical findings. We also cover some of the primary challenges, safety concerns and public responses to COVID-19 vaccine trials, and consider what this can mean for the future. By compiling this information, we aim to facilitate a more thorough understanding of the extensive COVID-19 clinical testing vaccine landscape as it unfolds, and better highlight some of the complexities and challenges being faced by the joint effort of the scientific community in finding a prophylactic against COVID-19.
Collapse
Affiliation(s)
- Ester Calvo Fernández
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNYUSA
| | - Lucie Y. Zhu
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNYUSA
| |
Collapse
|
67
|
Zilberzwige-Tal S, Alon DM, Gazit D, Zachariah S, Hollander A, Gazit E, Elbaz J. Genetically Encoding Ultrastable Virus-like Particles Encapsulating Functional DNA Nanostructures in Living Bacteria. ACS Synth Biol 2021; 10:1798-1807. [PMID: 34077194 DOI: 10.1021/acssynbio.0c00586] [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: 11/29/2022]
Abstract
DNA nanotechnology is leading the field of in vitro molecular-scale device engineering, accumulating to a dazzling array of applications. However, while DNA nanostructures' function is robust under in vitro settings, their implementation in real-world conditions requires overcoming their rapid degradation and subsequent loss of function. Viruses are sophisticated supramolecular assemblies, able to protect their nucleic acid content in inhospitable biological environments. Inspired by this natural ability, we engineered in vitro and in vivo technologies, enabling the encapsulation and protection of functional DNA nanostructures inside MS2 bacteriophage virus-like particles (VLPs). We demonstrate the ssDNA-VLPs nanocomposites' (NCs) abilities to encapsulate single-stranded-DNA (ssDNA) in a variety of sizes (200-1500 nucleotides (nt)), sequences, and structures while retaining their functionality. Moreover, by exposing these NCs to hostile biological conditions, such as human blood serum, we exhibit that the VLPs serve as an excellent protective shell. These engineered NCs pose critical properties that are yet unattainable by current fabrication methods.
Collapse
Affiliation(s)
- Shai Zilberzwige-Tal
- The Shmunis School of Molecular Cell Biology & Biotechnology, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dan Mark Alon
- The Shmunis School of Molecular Cell Biology & Biotechnology, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Danielle Gazit
- The Shmunis School of Molecular Cell Biology & Biotechnology, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shahar Zachariah
- The Shmunis School of Molecular Cell Biology & Biotechnology, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Amit Hollander
- The Shmunis School of Molecular Cell Biology & Biotechnology, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ehud Gazit
- The Shmunis School of Molecular Cell Biology & Biotechnology, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel
- BLAVATNIK Center for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Materials Science and Engineering Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Johann Elbaz
- The Shmunis School of Molecular Cell Biology & Biotechnology, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
68
|
Song SJ, Shin GI, Noh J, Lee J, Kim DH, Ryu G, Ahn G, Jeon H, Diao HP, Park Y, Kim MG, Kim WY, Kim YJ, Sohn EJ, Song CS, Hwang I. Plant-based, adjuvant-free, potent multivalent vaccines for avian influenza virus via Lactococcus surface display. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2021; 63:1505-1520. [PMID: 34051041 DOI: 10.1111/jipb.13141] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/26/2021] [Indexed: 05/28/2023]
Abstract
Influenza epidemics frequently and unpredictably break out all over the world, and seriously affect the breeding industry and human activity. Inactivated and live attenuated viruses have been used as protective vaccines but exhibit high risks for biosafety. Subunit vaccines enjoy high biosafety and specificity but have a few weak points compared to inactivated virus or live attenuated virus vaccines, especially in low immunogenicity. In this study, we developed a new subunit vaccine platform for a potent, adjuvant-free, and multivalent vaccination. The ectodomains of hemagglutinins (HAs) of influenza viruses were expressed in plants as trimers (tHAs) to mimic their native forms. tHAs in plant extracts were directly used without purification for binding to inactivated Lactococcus (iLact) to produce iLact-tHAs, an antigen-carrying bacteria-like particle (BLP). tHAs BLP showed strong immune responses in mice and chickens without adjuvants. Moreover, simultaneous injection of two different antigens by two different formulas, tHAH5N6 + H9N2 BLP or a combination of tHAH5N6 BLP and tHAH9N2 BLP, led to strong immune responses to both antigens. Based on these results, we propose combinations of plant-based antigen production and BLP-based delivery as a highly potent and cost-effective platform for multivalent vaccination for subunit vaccines.
Collapse
Affiliation(s)
- Shi-Jian Song
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Gyeong-Im Shin
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, 660-701, Korea
| | | | - Jiho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Korea
| | - Deok-Hwan Kim
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Korea
| | - Gyeongryul Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, 660-701, Korea
| | - Gyeongik Ahn
- Division of Applied Life Science (BK21 PLUS), Institute of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Korea
| | - Hyungmin Jeon
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Hai-Ping Diao
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Youngmin Park
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
- Bioapp, Inc., Pohang Technopark Complex, Pohang, 37668, Korea
| | - Min Gab Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, 660-701, Korea
| | - Woe-Yeon Kim
- Division of Applied Life Science (BK21 PLUS), Institute of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Korea
| | - Young-Jin Kim
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Eun-Ju Sohn
- Bioapp, Inc., Pohang Technopark Complex, Pohang, 37668, Korea
| | - Chang Seon Song
- KCAV Inc., Gwangjin-gu, 05029, Korea
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Korea
| | - Inhwan Hwang
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| |
Collapse
|
69
|
Warner NL, Frietze KM. Development of Bacteriophage Virus-Like Particle Vaccines Displaying Conserved Epitopes of Dengue Virus Non-Structural Protein 1. Vaccines (Basel) 2021; 9:726. [PMID: 34358143 PMCID: PMC8310087 DOI: 10.3390/vaccines9070726] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
Dengue virus (DENV) is a major global health problem, with over half of the world's population at risk of infection. Despite over 60 years of efforts, no licensed vaccine suitable for population-based immunization against DENV is available. Here, we describe efforts to engineer epitope-based vaccines against DENV non-structural protein 1 (NS1). NS1 is present in DENV-infected cells as well as secreted into the blood of infected individuals. NS1 causes disruption of endothelial cell barriers, resulting in plasma leakage and hemorrhage. Immunizing against NS1 could elicit antibodies that block NS1 function and also target NS1-infected cells for antibody-dependent cell cytotoxicity. We identified highly conserved regions of NS1 from all four DENV serotypes. We generated synthetic peptides to these regions and chemically conjugated them to bacteriophage Qβ virus-like particles (VLPs). Mice were immunized two times with the candidate vaccines and sera were tested for the presence of antibodies that bound to the cognate peptide, recombinant NS1 from all four DENV serotypes, and DENV-2-infected cells. We found that two of the candidate vaccines elicited antibodies that bound to recombinant NS1, and one candidate vaccine elicited antibodies that bound to DENV-infected cells. These results show that an epitope-specific vaccine against conserved regions of NS1 could be a promising approach for DENV vaccines or therapeutics to bind circulating NS1 protein.
Collapse
Affiliation(s)
- Nikole L. Warner
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences, Albuquerque, NM 87131, USA;
| | - Kathryn M. Frietze
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences, Albuquerque, NM 87131, USA;
- Clinical and Translational Science Center, University of New Mexico Health Sciences, Albuquerque, NM 87131, USA
| |
Collapse
|
70
|
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
|
71
|
Patel SP, Patel GS, Suthar JV. Inside the story about the research and development of COVID-19 vaccines. Clin Exp Vaccine Res 2021; 10:154-170. [PMID: 34222129 PMCID: PMC8217575 DOI: 10.7774/cevr.2021.10.2.154] [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: 03/01/2021] [Accepted: 05/03/2021] [Indexed: 12/29/2022] Open
Abstract
The ongoing coronavirus threat from China has spread rapidly to other nations and has been declared a global health emergency by the World Health Organization (WHO). The pandemic has resulted in over half of the world's population living under conditions of lockdown. Several academic institutions and pharmaceutical companies that are in different stages of development have plunged into the vaccine development race against coronavirus disease 2019 (COVID-19). The demand for immediate therapy and potential prevention of COVID-19 is growing with the increase in the number of individuals affected due to the seriousness of the disease, global dissemination, lack of prophylactics, and therapeutics. The challenging part is a need for vigorous testing for immunogenicity, safety, efficacy, and level of protection conferred in the hosts for the vaccines. As the world responds to the COVID-19 pandemic, we face the challenge of an overabundance of information related to the virus. Inaccurate information and myths spread widely and at speed, making it more difficult for the public to identify verified facts and advice from trusted sources, such as their local health authority or WHO. This review focuses on types of vaccine candidates against COVID-19 in clinical as well as in the preclinical development platform.
Collapse
Affiliation(s)
- Shrina P Patel
- Ramanbhai Patel College of Pharmacy, Charusat University, Anand, India
| | - Gayatri S Patel
- Ramanbhai Patel College of Pharmacy, Charusat University, Anand, India
| | - Jalpa V Suthar
- Ramanbhai Patel College of Pharmacy, Charusat University, Anand, India
| |
Collapse
|
72
|
Cimica V, Saleem S, Matuczinski E, Adams-Fish D, McMahon C, Rashid S, Stedman TT. A Virus-Like Particle-Based Vaccine Candidate against the Tick-Borne Powassan Virus Induces Neutralizing Antibodies in a Mouse Model. Pathogens 2021; 10:pathogens10060680. [PMID: 34072726 PMCID: PMC8229747 DOI: 10.3390/pathogens10060680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Powassan virus (POWV) is a tick-borne flavivirus circulating in North America and the Russian Far East that can cause severe neuroinvasive diseases, including encephalitis, meningitis, and meningoencephalitis. The reported neuroinvasive case fatality is about 10%, and approximately 50% of the survivors from the neuroinfection exhibit long-lasting or permanent neurological sequelae. Currently, treatment of POWV infection is supportive, and no FDA-approved vaccines or specific therapeutics are available. A novel Powassan vaccine candidate was created using virus-like particle technology (POW-VLP) and assembled with the viral structural proteins pre-Membrane (prM) and Envelope (E). Western blot immunoassay demonstrated high antigenicity of POW-VLP structural proteins. Transmission electron microscopy indicated that the POW-VLP exhibited icosahedral morphology typical of flaviviruses. A dose-escalation study in a murine model was performed to test immunogenicity and safety. Serum antibody was tested by ELISA, demonstrating that POW-VLP afforded 100% seroconversion to the E protein. Reporter viral-particle neutralization assay demonstrated high levels of neutralizing antibodies in the serum of immunized mice. Hybridomas expressing monoclonal antibodies were produced following POW-VLP immunization. The POW-VLP vaccine candidate created in this study provides a strategy for inducing protective antibodies against Powassan neuroinvasive infection.
Collapse
|
73
|
Maria Vasconcelos Queiroz A, Aleksandrovna Yanshina Y, Thays da Silva Rodrigues E, Luciano Neves Santos F, Alejandra Fiorani Celedon P, Maheshwari S, Beatriz Gabelli S, Stephanie Peucelle Rubio C, Durana A, Guérin DMA, Sousa Silva M. Antibodies response induced by recombinant virus-like particles from Triatoma virus and chimeric antigens from Trypanosoma cruzi. Vaccine 2021; 39:4723-4732. [PMID: 34053789 DOI: 10.1016/j.vaccine.2021.05.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/04/2021] [Accepted: 05/14/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND The infection caused by the protozoan Trypanosoma cruzi affects humans and is called as Chagas disease. Currently, the main measures available to reduce the incidence of this disease are drug treatment and vector control. Traditionally, the development of vaccines occurs mainly through the use of antigenic candidates of the etiologic agent in the form of a vaccine preparation. Virus-like particles (VLPs) are structures analogous to viral capsids composed essentially of structural proteins and are widely used in vaccination protocols because of their immunostimulatory properties. In this context, the objective of this study was to use strategies in a murine immunization model to characterize the immunostimulatory capacity of VLPs from Triatoma virus (TrV-VLPs), analysed in the presence or absence of the aluminium vaccine adjuvant. In parallel, to characterize the immunogenic behaviour of four T. cruzi chimeric recombinant proteins (mix-IBMP) associated with TrV-VLPs or aluminium vaccine adjuvant. METHOD We immunized BALB/c mice once or twice, depending on the strategy, and collected serum samples at 15, 30 and 45 days after the immunization. Subsequently, serum samples from animals immunized with TrV-VLPs were used to determine total IgG, IgG1, IgG2a, IgG2b and IgG3 anti-TrV-VLPs by enzyme-linked immunosorbent assay (ELISA). RESULTS Data obtained demonstrate the ability of TrV-VLPs to preferably induce IgG2b and IgG3 type antibodies in the absence of aluminium adjuvant. In fact, the use of aluminium did not interfere with the total IgG profile of anti-TrV-VLPs. Interestingly, mix-IBMP had a better profile of total IgG, IgG1 and IgG3 subclasses when mixed with TrV-VLPs. CONCLUSION In conclusion, these results suggest the potential of TrV-VLPs as a vaccine adjuvant and the use of T. cruzi chimeric antigens as a rational strategy for the development of vaccines against the experimental model of Chagas disease.
Collapse
Affiliation(s)
- Aline Maria Vasconcelos Queiroz
- Postgraduate Programme in Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Rua Gen, Gustavo Cordeiro de Farias, 384, 59012-570 Natal, Brazil; Immunoparasitology Laboratory, Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Rua Gen, Gustavo Cordeiro de Farias, 384, 59012-570 Natal, Brazil
| | - Yulia Aleksandrovna Yanshina
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, Universidade Nova de Lisboa, Rua da Juqueira, 100, 1800-166 Lisbon, Portugal
| | - Emily Thays da Silva Rodrigues
- Immunoparasitology Laboratory, Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Rua Gen, Gustavo Cordeiro de Farias, 384, 59012-570 Natal, Brazil
| | - Fred Luciano Neves Santos
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Fiocruz, Rua Waldemar Falcão, 121, 40296-710 Salvador, Brazil.
| | | | - Sweta Maheshwari
- School of Medicine, Johns Hopkins University, 725 N Wolfe St, Baltimore, MD 21205, USA
| | | | - Carla Stephanie Peucelle Rubio
- Departamento de Bioquímica y Biología Molecular, Instituto Biofisika, Universidad del País Vasco (UBF, CSIC, UPV-EHU), B° Sarriena S/N, 48940 Leioa, Bizkaia, Spain; Ikosaedrika Biologicals S.L. ZITEK Edificio Rectorado UPV/EHU, B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain
| | - Aritz Durana
- Instituto Biofisika (CSIC, UPV/EHU), Fundación Biofísica Bizkaia, B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain
| | - Diego M A Guérin
- Departamento de Bioquímica y Biología Molecular, Instituto Biofisika, Universidad del País Vasco (UBF, CSIC, UPV-EHU), B° Sarriena S/N, 48940 Leioa, Bizkaia, Spain; Ikosaedrika Biologicals S.L. ZITEK Edificio Rectorado UPV/EHU, B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain.
| | - Marcelo Sousa Silva
- Postgraduate Programme in Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Rua Gen, Gustavo Cordeiro de Farias, 384, 59012-570 Natal, Brazil; Immunoparasitology Laboratory, Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Rua Gen, Gustavo Cordeiro de Farias, 384, 59012-570 Natal, Brazil; Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, Universidade Nova de Lisboa, Rua da Juqueira, 100, 1800-166 Lisbon, Portugal.
| |
Collapse
|
74
|
Bayat M, Asemani Y, Najafi S. Essential considerations during vaccine design against COVID-19 and review of pioneering vaccine candidate platforms. Int Immunopharmacol 2021; 97:107679. [PMID: 33930707 PMCID: PMC8049400 DOI: 10.1016/j.intimp.2021.107679] [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: 02/27/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023]
Abstract
The calamity of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), COVID-19, is still a global human tragedy. To date, no specific antiviral drug or therapy has been able to break the widespread of SARS-CoV2. It has been generally believed that stimulating protective immunity via universal vaccination is the individual strategy to manage this pandemic. Achieving an effective COVID-19 vaccine requires attention to the immunological and non-immunological standpoints mentioned in this article. Here, we try to introduce the considerable immunological aspects, potential antigen targets, appropriate adjuvants as well as key points in the various stages of COVID-19 vaccine development. Also, the principal features of the preclinical and clinical studies of pioneering COVID-19 vaccine candidates were pointed out by reviewing the available information. Finally, we discuss the key challenges in the successful design of the COVID-19 vaccine and address the most fundamental strengths and weaknesses of common vaccine platforms.
Collapse
Affiliation(s)
- Maryam Bayat
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yahya Asemani
- Department of Immunology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sajad Najafi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
75
|
Schijns V, Majhen D, van der Ley P, Thakur A, Summerfield A, Berisio R, Nativi C, Fernández-Tejada A, Alvarez-Dominguez C, Gizurarson S, Zamyatina A, Molinaro A, Rosano C, Jakopin Ž, Gursel I, McClean S. Rational Vaccine Design in Times of Emerging Diseases: The Critical Choices of Immunological Correlates of Protection, Vaccine Antigen and Immunomodulation. Pharmaceutics 2021; 13:501. [PMID: 33917629 PMCID: PMC8067490 DOI: 10.3390/pharmaceutics13040501] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 01/21/2023] Open
Abstract
Vaccines are the most effective medical intervention due to their continual success in preventing infections and improving mortality worldwide. Early vaccines were developed empirically however, rational design of vaccines can allow us to optimise their efficacy, by tailoring the immune response. Establishing the immune correlates of protection greatly informs the rational design of vaccines. This facilitates the selection of the best vaccine antigens and the most appropriate vaccine adjuvant to generate optimal memory immune T cell and B cell responses. This review outlines the range of vaccine types that are currently authorised and those under development. We outline the optimal immunological correlates of protection that can be targeted. Finally we review approaches to rational antigen selection and rational vaccine adjuvant design. Harnessing current knowledge on protective immune responses in combination with critical vaccine components is imperative to the prevention of future life-threatening diseases.
Collapse
Affiliation(s)
- Virgil Schijns
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
- Epitopoietic Research Corporation (ERC), 5374 RE Schaijk, The Netherlands
| | - Dragomira Majhen
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Instiute, HR-10000 Zagreb, Croatia;
| | - Peter van der Ley
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
| | - Aneesh Thakur
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Artur Summerfield
- Institute of Virology and Immunology, 3147 Mittelhausern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, National Research Council, I-80134 Naples, Italy;
| | - Cristina Nativi
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy;
| | - Alberto Fernández-Tejada
- Chemical Immunology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Biscay Science and Technology Park, 48160 Derio-Bilbao, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Carmen Alvarez-Dominguez
- Facultativo en plantilla (Research Faculty), Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Sveinbjörn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, 107 Reykjavik, Iceland;
- Department of Pharmacy, College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria;
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Santangelo, I-80126 Napoli, Italy;
- Department of Chemistry, School of Science, Osaka University, 1-1 Osaka University Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Camillo Rosano
- Proteomics and Mass Spectrometry Unit, IRCCS Policlinico San Martino, 16132 Genova-1, Italy;
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubiljana, Slovenia;
| | - Ihsan Gursel
- Molecular Biology and Genetics Department, Science Faculty, Bilkent University, Bilkent, 06800 Ankara, Turkey;
| | - Siobhán McClean
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
76
|
Darwish RM. COVID-19 immunity and vaccines: what a pharmacist needs to know. ASIAN BIOMED 2021; 15:51-67. [PMID: 37551403 PMCID: PMC10388771 DOI: 10.2478/abm-2021-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
COVID-19 vaccines are being produced using different platforms by different companies, some of which are entering Phase 3 and 4 trials. Due to the pandemic, this production has been accelerated, which leaves a window for speculation on the method of production and safety. Pharmacists are familiar with vaccination; however, COVID-19 vaccines are still new and further work is needed to clarify many aspects, including side effects, methods of storage, and number of doses. Prioritization of vaccination has been implemented to a certain extent, but no clear strategy is available. A comprehensive overview on immunity and immunological principles for the design of COVID-19 vaccine strategies is provided in this narrative review and the current COVID-19 vaccine landscape is discussed, in addition to exploring the principles for prioritization of vaccination using data from articles available in PubMed and from health organizations. Pharmacists should have a better understanding of COVID-19 vaccines and their manufacture. This would also allow better counseling of the public on COVID 19, immunization, and explaining prioritization basis and vaccination programs.
Collapse
Affiliation(s)
- Rula M. Darwish
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, The University of Jordan, Aljubeiha, Amman00962, Jordan
| |
Collapse
|
77
|
A smart viral vector for targeted delivery of hydrophobic drugs. Sci Rep 2021; 11:7030. [PMID: 33782428 PMCID: PMC8007742 DOI: 10.1038/s41598-021-86198-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Targeted delivery of hydrophobic chemotherapeutic drugs to tumor cells remains a fundamental problem in cancer therapy. Effective encapsulation of hydrophobic drugs in nano-vehicles can improve their pharmacokinetics, bioavailability and prevent off-target localization. We have devised a method for easy chemical conjugation and multivalent display of a tumor-homing peptide to virus-like particles of a non-mammalian virus, Flock House Virus (FHV), to engineer it into a smart vehicle for targeted delivery of hydrophobic drugs. This conjugation method provides dual functionalization to the VLPs, first, a 2 kDa PEG spacer arm shields VLPs from immune reactivity, and second, attachment of the tumor homing peptide tLyP-1 chauffeurs the encapsulated hydrophobic drugs to target cells. The fortuitous affinity of the FHV capsid towards hydrophobic molecules, and dependence on Ca2+ for maintaining a stable capsid shell, were utilized for incorporation of hydrophobic drugs—doxorubicin and ellipticine—in tLyP-1 conjugated VLPs. The drug release profile from the VLP was observed to be gradual, and strictly endosomal pH dependent. We propose that this accessible platform empowers surface functionalization of VLP with numerous ligands containing terminal cysteines, for generating competent delivery vehicles, antigenic display and other biomedical applications.
Collapse
|
78
|
An Overview of Nanocarrier-Based Adjuvants for Vaccine Delivery. Pharmaceutics 2021; 13:pharmaceutics13040455. [PMID: 33801614 PMCID: PMC8066039 DOI: 10.3390/pharmaceutics13040455] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
The development of vaccines is one of the most significant medical accomplishments which has helped to eradicate a large number of diseases. It has undergone an evolutionary process from live attenuated pathogen vaccine to killed whole organisms or inactivated toxins (toxoids), each of them having its own advantages and disadvantages. The crucial parameters in vaccination are the generation of memory response and protection against infection, while an important aspect is the effective delivery of antigen in an intelligent manner to evoke a robust immune response. In this regard, nanotechnology is greatly contributing to developing efficient vaccine adjuvants and delivery systems. These can protect the encapsulated antigen from the host’s in-vivo environment and releasing it in a sustained manner to induce a long-lasting immunostimulatory effect. In view of this, the present review article summarizes nanoscale-based adjuvants and delivery vehicles such as viral vectors, virus-like particles and virosomes; non-viral vectors namely nanoemulsions, lipid nanocarriers, biodegradable and non-degradable nanoparticles, calcium phosphate nanoparticles, colloidally stable nanoparticles, proteosomes; and pattern recognition receptors covering c-type lectin receptors and toll-like receptors.
Collapse
|
79
|
Chimeric VLPs Based on HIV-1 Gag and a Fusion Rabies Glycoprotein Induce Specific Antibodies against Rabies and Foot-and-Mouth Disease Virus. Vaccines (Basel) 2021; 9:vaccines9030251. [PMID: 33809060 PMCID: PMC7999769 DOI: 10.3390/vaccines9030251] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/25/2022] Open
Abstract
Foot and mouth disease is a livestock acute disease, causing economic losses in affected areas. Currently, control of this disease is performed by mandatory vaccination campaigns using inactivated viral vaccines. In this work, we describe the development of a chimeric VLP-based vaccine candidate for foot-and-mouth disease virus (FMDV), based on the co-expression of the HIV-1 Gag protein and a novel fusion rabies glycoprotein (RVG), which carries in its N-term the FMDV main antigen: the G-H loop. It is demonstrated by confocal microscopy that both Gag-GFP polyprotein and the G-H loop colocalize at the cell membrane and, that the Gag polyprotein of the HIV virus acts as a scaffold for enveloped VLPs that during the budding process acquires the proteins that are being expressed in the cell membrane. The obtained VLPs were spherical particles of 130 ± 40 nm in diameter (analyzed by TEM, Cryo-TEM and NTA) carrying an envelope membrane that efficiently display the GH-RVG on its surface (analyzed by gold immunolabeling). Immunostainings with a FMDV hyperimmune serum showed that the heterologous antigenic site, genetically fused to RVG, is recognized by specific G-H loop antibodies. Additionally, the cVLPs produced expose the G-H loop to the liquid surrounding (analyzed by specific ELISA). Finally, we confirmed that these FMD cVLPs are able to induce a specific humoral immune response, based on antibodies directed to the G-H loop in experimental animals.
Collapse
|
80
|
Cherepushkin SA, Tsibezov VV, Yuzhakov AG, Latyshev OE, Alekseev KP, Altayeva EG, Khametova KM, Vorkunova GK, Yuzhakova KA, Grebennikova TV. [Synthesis and characterization of human rotavirus A ( Reoviridae: Sedoreovirinae: Rotavirus: Rotavirus A) virus-like particles]. Vopr Virusol 2021; 66:55-64. [PMID: 33683066 DOI: 10.36233/0507-4088-27] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/07/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Rotavirus infection is the leading cause of acute gastroenteritis among infants. The development of new vaccines against rotavirus A is urgent because the virus has many genotypes, some of which have regional prevalence. Virus-like particles (VLP) is a promising way to create effective and safe vaccine preparations.The purpose of the study is to develop the technology for the production of VLP, containing VP2, VP4, VP6 and VP7 of viral genotypes prevalent on the territory of the Russian Federation, and to give its molecular genetic and virological characteristics. MATERIAL AND METHODS The virulent strain Wa G1P[8] of human RV A adapted to MARC-145 cell culture has been used. It was cultured and purified according to the method described by the authors earlier. Standard molecular genetic and cytological methods were used: gene synthesis; cloning into transfer plasmids; recombinant baculoviruses production in Bac-to-Bac expression system; VLP production in the insect cells; centrifugation in sucrose solution; enzyme-linked immunosorbent assay (ELISA); electron microscopy (EM); polyacrylamide gel electrophoresis (SDS-PAGE) and western blot analysis. RESULTS VP4 and VP7 of the six most represented in Russia genotypes: G1, G2, G4, G9, P4, P8, as well as VP2 and VP6 were selected for VLP production. Recombinant baculoviruses were obtained with codon frequencies optimized for insect cells. Cabbage loopper (Trichoplusia ni) cell culture was coinfected with different combinations of baculoviruses, and VLP consisting of 2-4 proteins were produced. VLP were purified by centrifugation. The size and morphology of the particles matched the rotavirus A virion (by EM). The presence of rotavirus A proteins in VLP was confirmed by the ELISA, SDS-PAGE and western blot analysis. CONCLUSION The technology for the synthesis of three-layer VLP consisting of VP2, VP4, VP6 and VP7 has been developed and optimized. The resulting VLP composition represents 6 serotypes of VP4 and VP7, which are most represented on the territory of Russia, and can be used for vaccine development.
Collapse
Affiliation(s)
- S A Cherepushkin
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | - V V Tsibezov
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | - A G Yuzhakov
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | - O E Latyshev
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | - K P Alekseev
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | | | - K M Khametova
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | - G K Vorkunova
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | - K A Yuzhakova
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| | - T V Grebennikova
- FSBI National Research Center for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya
| |
Collapse
|
81
|
Dry Formulation of Virus-Like Particles in Electrospun Nanofibers. Vaccines (Basel) 2021; 9:vaccines9030213. [PMID: 33802376 PMCID: PMC8000389 DOI: 10.3390/vaccines9030213] [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: 01/22/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 11/16/2022] Open
Abstract
Biologics can be combined with liquid polymer materials and electrospun to produce a dry nanofibrous scaffold. Unlike spray-drying and freeze-drying, electrospinning minimizes the physiological stress on sensitive materials, and nanofiber mat properties such as hydrophobicity, solubility, and melting temperature can be tuned based on the polymer composition. In this study, we explored the dry formulation of a virus-like particle (VLP) vaccine by electrospinning VLP derived from rabbit hemorrhagic disease virus modified to carry the MHC-I gp100 tumor-associated antigen epitope. VLP were added to a polyvinylpyrrolidone (PVP) solution (15% w/v) followed by electrospinning at 24 kV. Formation of a nanofibrous mat was confirmed by scanning electron microscopy, and the presence of VLP was confirmed by transmission electron microscopy and Western blot. VLP from the nanofibers induced T-cell activation and interferon- (IFN-) γ production in vitro. To confirm in vivo cytotoxicity, Pmel mice treated by injection with gp100 VLP from nanofibers induced a gp100 specific immune response, lysing approximately 65% of gp100-pulsed target cells, comparable to mice vaccinated with gp100 VLP in PBS. VLP from nanofibers also induced an antibody response. This work shows that electrospinning can be used to dry-formulate VLP, preserving both humoral and cell-mediated immunity.
Collapse
|
82
|
Kim YI, Kim D, Yu KM, Seo HD, Lee SA, Casel MAB, Jang SG, Kim S, Jung W, Lai CJ, Choi YK, Jung JU. Development of Spike Receptor-Binding Domain Nanoparticles as a Vaccine Candidate against SARS-CoV-2 Infection in Ferrets. mBio 2021; 12:e00230-21. [PMID: 33653891 PMCID: PMC8092224 DOI: 10.1128/mbio.00230-21] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/31/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a causative agent of the CoV disease 2019 (COVID-19) pandemic, enters host cells via the interaction of its receptor-binding domain (RBD) of the spike protein with host angiotensin-converting enzyme 2 (ACE2). Therefore, the RBD is a promising vaccine target to induce protective immunity against SARS-CoV-2 infection. In this study, we report the development of an RBD protein-based vaccine candidate against SARS-CoV-2 using self-assembling Helicobacter pylori-bullfrog ferritin nanoparticles as an antigen delivery system. RBD-ferritin protein purified from mammalian cells efficiently assembled into 24-mer nanoparticles. Sixteen- to 20-month-old ferrets were vaccinated with RBD-ferritin nanoparticles (RBD nanoparticles) by intramuscular or intranasal inoculation. All vaccinated ferrets with RBD nanoparticles produced potent neutralizing antibodies against SARS-CoV-2. Strikingly, vaccinated ferrets demonstrated efficient protection from SARS-CoV-2 challenge, showing no fever, body weight loss, or clinical symptoms. Furthermore, vaccinated ferrets showed rapid clearance of infectious virus in nasal washes and lungs as well as of viral RNA in respiratory organs. This study demonstrates that spike RBD-nanoparticles are an effective protein vaccine candidate against SARS-CoV-2.
Collapse
Affiliation(s)
- Young-Il Kim
- College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Dokyun Kim
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kwang-Min Yu
- College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Hogyu David Seo
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Shin-Ae Lee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mark Anthony B Casel
- College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Seung-Gyu Jang
- College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Stephanie Kim
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - WooRam Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Chih-Jen Lai
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Young Ki Choi
- College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Jae U Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
83
|
Chung JY, Thone MN, Kwon YJ. COVID-19 vaccines: The status and perspectives in delivery points of view. Adv Drug Deliv Rev 2021; 170:1-25. [PMID: 33359141 PMCID: PMC7759095 DOI: 10.1016/j.addr.2020.12.011] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/29/2022]
Abstract
Due to the high prevalence and long incubation periods often without symptoms, the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has infected millions of individuals globally, causing the coronavirus disease 2019 (COVID-19) pandemic. Even with the recent approval of the anti-viral drug, remdesivir, and Emergency Use Authorization of monoclonal antibodies against S protein, bamlanivimab and casirimab/imdevimab, efficient and safe COVID-19 vaccines are still desperately demanded not only to prevent its spread but also to restore social and economic activities via generating mass immunization. Recent Emergency Use Authorization of Pfizer and BioNTech's mRNA vaccine may provide a pathway forward, but monitoring of long-term immunity is still required, and diverse candidates are still under development. As the knowledge of SARS-CoV-2 pathogenesis and interactions with the immune system continues to evolve, a variety of drug candidates are under investigation and in clinical trials. Potential vaccines and therapeutics against COVID-19 include repurposed drugs, monoclonal antibodies, antiviral and antigenic proteins, peptides, and genetically engineered viruses. This paper reviews the virology and immunology of SARS-CoV-2, alternative therapies for COVID-19 to vaccination, principles and design considerations in COVID-19 vaccine development, and the promises and roles of vaccine carriers in addressing the unique immunopathological challenges presented by the disease.
Collapse
Affiliation(s)
- Jee Young Chung
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States of America
| | - Melissa N Thone
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States of America
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States of America; Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, United States of America; Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States of America; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States of America.
| |
Collapse
|
84
|
Nooraei S, Bahrulolum H, Hoseini ZS, Katalani C, Hajizade A, Easton AJ, Ahmadian G. Virus-like particles: preparation, immunogenicity and their roles as nanovaccines and drug nanocarriers. J Nanobiotechnology 2021; 19:59. [PMID: 33632278 PMCID: PMC7905985 DOI: 10.1186/s12951-021-00806-7] [Citation(s) in RCA: 416] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Virus-like particles (VLPs) are virus-derived structures made up of one or more different molecules with the ability to self-assemble, mimicking the form and size of a virus particle but lacking the genetic material so they are not capable of infecting the host cell. Expression and self-assembly of the viral structural proteins can take place in various living or cell-free expression systems after which the viral structures can be assembled and reconstructed. VLPs are gaining in popularity in the field of preventive medicine and to date, a wide range of VLP-based candidate vaccines have been developed for immunization against various infectious agents, the latest of which is the vaccine against SARS-CoV-2, the efficacy of which is being evaluated. VLPs are highly immunogenic and are able to elicit both the antibody- and cell-mediated immune responses by pathways different from those elicited by conventional inactivated viral vaccines. However, there are still many challenges to this surface display system that need to be addressed in the future. VLPs that are classified as subunit vaccines are subdivided into enveloped and non- enveloped subtypes both of which are discussed in this review article. VLPs have also recently received attention for their successful applications in targeted drug delivery and for use in gene therapy. The development of more effective and targeted forms of VLP by modification of the surface of the particles in such a way that they can be introduced into specific cells or tissues or increase their half-life in the host is likely to expand their use in the future. Recent advances in the production and fabrication of VLPs including the exploration of different types of expression systems for their development, as well as their applications as vaccines in the prevention of infectious diseases and cancers resulting from their interaction with, and mechanism of activation of, the humoral and cellular immune systems are discussed in this review.
Collapse
Affiliation(s)
- Saghi Nooraei
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), P. O. BOX: 14155-6343, Tehran, 1497716316, Iran
| | - Howra Bahrulolum
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), P. O. BOX: 14155-6343, Tehran, 1497716316, Iran
| | - Zakieh Sadat Hoseini
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), P. O. BOX: 14155-6343, Tehran, 1497716316, Iran
| | - Camellia Katalani
- Sari Agriculture Science and Natural Resource University (SANRU), Genetics and Agricultural Biotechnology Institute of Tabarestan (GABIT), Sari, Iran
| | - Abbas Hajizade
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Andrew J Easton
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, UK.
| | - Gholamreza Ahmadian
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), P. O. BOX: 14155-6343, Tehran, 1497716316, Iran.
| |
Collapse
|
85
|
Gerstweiler L, Bi J, Middelberg APJ. Virus-like particle preparation is improved by control over capsomere-DNA interactions during chromatographic purification. Biotechnol Bioeng 2021; 118:1707-1720. [PMID: 33484156 DOI: 10.1002/bit.27687] [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: 10/13/2020] [Revised: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 11/09/2022]
Abstract
Expression of viral capsomeres in bacterial systems and subsequent in vitro assembly into virus-like particles is a possible pathway for affordable future vaccines. However, purification is challenging as viral capsomeres show poor binding to chromatography media. In this study, the behavior of capsomeres in unfractionated bacterial lysate was compared with that for purified capsomeres, with or without added microbial DNA, to better understand reasons for poor bioprocess behavior. We show that aggregates or complexes form through the interaction between viral capsomeres and DNA, especially in bacterial lysates rich in contaminating DNA. The formation of these complexes prevents the target protein capsomeres from accessing the pores of chromatography media. We find that protein-DNA interactions can be modulated by controlling the ionic strength of the buffer and that at elevated ionic strengths the protein-DNA complexes dissociate. Capsomeres thus released show enhanced bind-elute behavior on salt-tolerant chromatography media. DNA could therefore be efficiently removed. We believe this is the first report of the use of an optimized salt concentration that dissociates capsomere-DNA complexes yet enables binding to salt-tolerant media. Post purification, assembly experiments indicate that DNA-protein interactions can play a negative role during in vitro assembly, as DNA-protein complexes could not be assembled into virus-like particles, but formed worm-like structures. This study reveals that the control over DNA-protein interaction is a critical consideration during downstream process development for viral vaccines.
Collapse
Affiliation(s)
- Lukas Gerstweiler
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jingxiu Bi
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia, Australia
| | | |
Collapse
|
86
|
Kim YI, Kim D, Yu KM, Seo HD, Lee SA, Casel MAB, Jang SG, Kim S, Jung W, Lai CJ, Choi YK, Jung JU. Development of spike receptor-binding domain nanoparticle as a vaccine candidate against SARS-CoV-2 infection in ferrets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.28.428743. [PMID: 33532767 PMCID: PMC7852231 DOI: 10.1101/2021.01.28.428743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a causative agent of COVID-19 pandemic, enters host cells via the interaction of its Receptor-Binding Domain (RBD) of Spike protein with host Angiotensin-Converting Enzyme 2 (ACE2). Therefore, RBD is a promising vaccine target to induce protective immunity against SARS-CoV-2 infection. In this study, we report the development of RBD protein-based vaccine candidate against SARS-CoV-2 using self-assembling H. pylori -bullfrog ferritin nanoparticles as an antigen delivery. RBD-ferritin protein purified from mammalian cells efficiently assembled into 24-mer nanoparticles. 16-20 months-old ferrets were vaccinated with RBD-ferritin nanoparticles (RBD-nanoparticles) by intramuscular or intranasal inoculation. All vaccinated ferrets with RBD-nanoparticles produced potent neutralizing antibodies against SARS-CoV-2. Strikingly, vaccinated ferrets demonstrated efficient protection from SARS-CoV-2 challenge, showing no fever, body weight loss and clinical symptoms. Furthermore, vaccinated ferrets showed rapid clearance of infectious viruses in nasal washes and lungs as well as viral RNA in respiratory organs. This study demonstrates the Spike RBD-nanoparticle as an effective protein vaccine candidate against SARS-CoV-2.
Collapse
Affiliation(s)
- Young-Il Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Dokyun Kim
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kwang-Min Yu
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Hogyu David Seo
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shin-Ae Lee
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mark Anthony B. Casel
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Seung-Gyu Jang
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Stephanie Kim
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - WooRam Jung
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Chih-Jen Lai
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Young Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
- Zoonotic Infectious Disease Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Jae U. Jung
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
87
|
Gerstweiler L, Billakanti J, Bi J, Middelberg A. Comparative evaluation of integrated purification pathways for bacterial modular polyomavirus major capsid protein VP1 to produce virus-like particles using high throughput process technologies. J Chromatogr A 2021; 1639:461924. [PMID: 33545579 PMCID: PMC7825977 DOI: 10.1016/j.chroma.2021.461924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/11/2021] [Accepted: 01/16/2021] [Indexed: 12/21/2022]
Abstract
Modular virus-like particles and capsomeres are potential vaccine candidates that can induce strong immune responses. There are many described protocols for the purification of microbially-produced viral protein in the literature, however, they suffer from inherent limitations in efficiency, scalability and overall process costs. In this study, we investigated alternative purification pathways to identify and optimise a suitable purification pathway to overcome some of the current challenges. Among the methods, the optimised purification strategy consists of an anion exchange step in flow through mode followed by a multi modal cation exchange step in bind and elute mode. This approach allows an integrated process without any buffer adjustment between the purification steps. The major contaminants like host cell proteins, DNA and aggregates can be efficiently removed by the optimised strategy, without the need for a size exclusion polishing chromatography step, which otherwise could complicate the process scalability and increase overall cost. High throughput process technology studies were conducted to optimise binding and elution conditions for multi modal cation exchanger, Capto™ MMC and strong anion exchanger Capto™ Q. A dynamic binding capacity of 14 mg ml−1 was achieved for Capto™ MMC resin. Samples derived from each purification process were thoroughly characterized by RP-HPLC, SEC-HPLC, SDS-PAGE and LC-ESI-MS/MS Mass Spectrometry analytical methods. Modular polyomavirus major capsid protein could be purified within hours using the optimised process achieving purities above 87% and above 96% with inclusion of an initial precipitation step. Purified capsid protein could be easily assembled in-vitro into well-defined virus-like particles by lowering pH with addition of calcium chloride to the eluate. High throughout studies allowed the screening of a vast design space within weeks, rather than months, and unveiled complicated binding behaviour for CaptoTM MMC.
Collapse
Affiliation(s)
- Lukas Gerstweiler
- The University of Adelaide, School of Chemical Engineering and Advanced Materials, Adelaide, SA 5005, Australia
| | - Jagan Billakanti
- Cytiva, Product and Application Specialist Downstream Design-In ANZ, Suite 547, Level 5, 7 Eden Park Drive, Macquarie Park, NSW 2113, Australia
| | - Jingxiu Bi
- The University of Adelaide, School of Chemical Engineering and Advanced Materials, Adelaide, SA 5005, Australia
| | - Anton Middelberg
- The University of Adelaide, Division of Research and Innovation, Adelaide, SA 5005, Australia.
| |
Collapse
|
88
|
Perera PY, Perera LP. Development of leading first-generation vaccines against SARS-CoV-2. Microbes Infect 2021; 23:104841. [PMID: 34022375 PMCID: PMC8133830 DOI: 10.1016/j.micinf.2021.104841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 01/29/2023]
Abstract
SARS-CoV-2 has infected more than 167 million individuals globally. Highly effective and safe vaccines are required to accelerate the development of herd immunity to end the pandemic. This review focuses on vaccines that are being developed at unprecedented speed globally and are completing late phase clinical trials to meet this urgent need.
Collapse
Affiliation(s)
- Pin-Yu Perera
- Pathology and Laboratory Medicine, Veterans Affairs Medical Center, Washington, DC, USA.
| | - Liyanage P Perera
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
89
|
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
|
90
|
Marchel M, Niewisiewicz J, Coroadinha AS, Marrucho IM. Purification of virus-like particles using aqueous biphasic systems composed of natural deep eutectic solvents. Sep Purif Technol 2020. [DOI: 10.1016/j.seppur.2020.117480] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
91
|
Promotion of Cellular and Humoral Immunity against Foot-and-Mouth Disease Virus by Immunization with Virus-Like Particles Encapsulated in Monophosphoryl Lipid A and Liposomes. Vaccines (Basel) 2020; 8:vaccines8040633. [PMID: 33142799 PMCID: PMC7712044 DOI: 10.3390/vaccines8040633] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 01/08/2023] Open
Abstract
Virus-like particles (VLPs) have emerged as promising vaccine candidates against foot-and-mouth disease (FMD). However, such vaccines provide a relatively low level of protection against FMD virus (FMDV) because of their poor immunogenicity. Therefore, it is necessary to design effective vaccine strategies that induce more potent immunogenicity. In order to investigate the means to improve FMD VLP vaccine (VLPFMDV) immunogenicity, we encapsulated VLPs (MPL/DDA-VLPFMDV) with cationic liposomes based on dimethyldioctadecylammonium bromide (DDA) and/or monophosphoryl lipid A (MPL, TLR4 agonist) as adjuvants. Unlike inactivated whole-cell vaccines, VLPFMDV were successfully encapsulated in this MPL/DDA system. We found that MPL/DDA-VLPFMDV could induce strong cell-mediated immune responses by inducing not only VLP-specific IFN-γ+CD4+ (Th1), IL-17A+CD4+ (Th17), and IFN-γ+CD8+ (activated CD8 response) T cells, but also the development of VLP-specific multifunctional CD4+ and CD8+ memory T cells co-expressing IFN-γ, TNF-α, and IL-2. In addition, the MPL/DDA-VLPFMDV vaccine markedly induced VLP-specific antibody titers; in particular, the vaccine induced greater Th1-predominant IgG responses than VLPFMDV only and DDA-VLPFMDV. These results are expected to provide important clues for the development of an effective VLPFMDV that can induce cellular and humoral immune responses, and address the limitations seen in current VLP vaccines for various diseases.
Collapse
|
92
|
Jeyanathan M, Afkhami S, Smaill F, Miller MS, Lichty BD, Xing Z. Immunological considerations for COVID-19 vaccine strategies. Nat Rev Immunol 2020; 20:615-632. [PMID: 32887954 PMCID: PMC7472682 DOI: 10.1038/s41577-020-00434-6] [Citation(s) in RCA: 673] [Impact Index Per Article: 134.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the most formidable challenge to humanity in a century. It is widely believed that prepandemic normalcy will never return until a safe and effective vaccine strategy becomes available and a global vaccination programme is implemented successfully. Here, we discuss the immunological principles that need to be taken into consideration in the development of COVID-19 vaccine strategies. On the basis of these principles, we examine the current COVID-19 vaccine candidates, their strengths and potential shortfalls, and make inferences about their chances of success. Finally, we discuss the scientific and practical challenges that will be faced in the process of developing a successful vaccine and the ways in which COVID-19 vaccine strategies may evolve over the next few years.
Collapse
MESH Headings
- Antibodies, Viral/biosynthesis
- Betacoronavirus/drug effects
- Betacoronavirus/immunology
- Betacoronavirus/pathogenicity
- COVID-19
- COVID-19 Vaccines
- Clinical Trials as Topic
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/virology
- Genetic Vectors/chemistry
- Genetic Vectors/immunology
- Humans
- Immunity, Herd/drug effects
- Immunity, Innate/drug effects
- Immunization Schedule
- Immunogenicity, Vaccine
- Pandemics/prevention & control
- Patient Safety
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- SARS-CoV-2
- Severe Acute Respiratory Syndrome/epidemiology
- Severe Acute Respiratory Syndrome/immunology
- Severe Acute Respiratory Syndrome/prevention & control
- Severe Acute Respiratory Syndrome/virology
- Vaccines, Attenuated
- Vaccines, DNA
- Vaccines, Subunit
- Vaccines, Virus-Like Particle
- Viral Vaccines/administration & dosage
- Viral Vaccines/biosynthesis
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Fiona Smaill
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Brian D Lichty
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| | - Zhou Xing
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
93
|
Tao Y, Yang R, Shu J, Zheng W, Chen J, Wu Y, He Y. Immune responses induced by a combined vaccination with a recombinant chimera of Mycoplasma hyopneumoniae antigens and capsid virus-like particles of porcine circovirus type 2. BMC Vet Res 2020; 16:342. [PMID: 32938456 PMCID: PMC7493066 DOI: 10.1186/s12917-020-02560-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 09/08/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Mycoplasma hyopneumoniae (Mhp) and porcine circovirus type 2 (PCV2) are two important pathogens causing Mycoplasma pneumonia of swine (MPS) and porcine circovirus diseases and porcine circovirus-associated diseases (PCVDs/PCVADs), respectively, and resulted in considerable economic loss to the swine industry worldwide. Currently, vaccination is one of the main measures to control these two diseases; however, there are few combination vaccines that can prevent these two diseases. To determine the effect of combination immunization, we developed capsid-derived (Cap) virus-like particles (VLPs) of PCV2 and a new recombinant chimera composed of the P97R1, P46, and P42 antigens of Mhp. Then we investigated the immune responses induced by the immunization with this combination vaccine in mice and piglets. RESULTS The high level antibodies against three protein antigens (P97R1, P46, and P42 of Mhp) were produced after immunization, up to or higher than 1:400,000; the antibody levels in Pro group continuously increased throughout the 42 days for all the antigens tested. The lymphocyte proliferative response in PCV2 group was stronger than that in PBS, VP, Mhp CV in mice. The antibody levels for Cap remained stable and reached the peak at 35 DAI. The IFN-γ and IL-4 in sera were significantly enhanced in the Pro group than that in the negative control-VP group on Day 14 and 28 post-the first immunization in piglets. CONCLUSIONS Above all, the combination immunization could induce humoral and cellular immune responses against all four antigens in mice and piglets. Therefore, our approach is a simple and effective vaccination strategy to protect pigs against MPS and PCVD/PCVAD.
Collapse
Affiliation(s)
- Yu Tao
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China
| | - Rui Yang
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China
| | - Jianhong Shu
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China
| | - Wenqian Zheng
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China
| | - Jian Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China
| | - Yuehong Wu
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China
| | - Yulong He
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China.
| |
Collapse
|
94
|
Basak S, Chu KB, Kang HJ, Kim MJ, Lee SH, Yoon KW, Jin H, Suh JW, Moon EK, Quan FS. Orally administered recombinant baculovirus vaccine elicits partial protection against avian influenza virus infection in mice. Microb Pathog 2020; 149:104495. [PMID: 32910984 DOI: 10.1016/j.micpath.2020.104495] [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: 05/28/2020] [Revised: 08/20/2020] [Accepted: 08/31/2020] [Indexed: 12/23/2022]
Abstract
Avian influenza outbreaks have placed a tremendous economic burden on the poultry industry, necessitating the need for an effective vaccine. Although multiple vaccine candidates are available, its development is hindered by several drawbacks associated with the vaccine platforms and as such, more improvements to the vaccines are needed. Therefore, in this study, the vaccine efficacy in the murine models was assessed prior to evaluation in chickens. An oral recombinant baculovirus (rBV) vaccine expressing influenza hemagglutinin (HA) (A/H5N1) was generated and its efficacy was investigated against homologous avian influenza infection in mice. Our results confirmed that oral administration of rBVs enhanced the level of virus-specific antibodies in the sera following boost immunization. Upon challenge infection with a lethal dose of highly pathogenic avian influenza virus (HPAI, H5N1) virus, a marked increase in mucosal IgG and IgA were observed. Drastically increased antibody secretory cell responses from the bone marrow cells and splenocytes of vaccinated mice were observed, in addition to the strongly elicited germinal center responses in the lungs and the spleens. Vaccinated mice showed significantly reduced lung pro-inflammatory cytokine responses, lung viral loads, body weight loss, and mortality. Though mice were only partially protected upon challenge infection, these results highlight the potential of orally administered rBVs expressing the HA as a vaccine candidate for controlling avian influenza outbreaks.
Collapse
Affiliation(s)
- Swarnendu Basak
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Su-Hwa Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hui Jin
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Gyeonggi-do, Republic of Korea
| | - Joo Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Gyeonggi-do, Republic of Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Republic of Korea; Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
95
|
Irvine DJ, Aung A, Silva M. Controlling timing and location in vaccines. Adv Drug Deliv Rev 2020; 158:91-115. [PMID: 32598970 PMCID: PMC7318960 DOI: 10.1016/j.addr.2020.06.019] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Vaccines are one of the most powerful technologies supporting public health. The adaptive immune response induced by immunization arises following appropriate activation and differentiation of T and B cells in lymph nodes. Among many parameters impacting the resulting immune response, the presence of antigen and inflammatory cues for an appropriate temporal duration within the lymph nodes, and further within appropriate subcompartments of the lymph nodes- the right timing and location- play a critical role in shaping cellular and humoral immunity. Here we review recent advances in our understanding of how vaccine kinetics and biodistribution impact adaptive immunity, and the underlying immunological mechanisms that govern these responses. We discuss emerging approaches to engineer these properties for future vaccines, with a focus on subunit vaccines.
Collapse
Affiliation(s)
- Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Murillo Silva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
96
|
Puente-Massaguer E, Saccardo P, Ferrer-Miralles N, Lecina M, Gòdia F. Coupling Microscopy and Flow Cytometry for a Comprehensive Characterization of Nanoparticle Production in Insect Cells. Cytometry A 2020; 97:921-932. [PMID: 32515126 DOI: 10.1002/cyto.a.24033] [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: 06/09/2019] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 01/01/2023]
Abstract
Advancements in the field of characterization techniques have broadened the opportunities to deepen into nanoparticle production bioprocesses. Gag-based virus-like particles (VLPs) have shown their potential as candidates for recombinant vaccine development. However, comprehensive characterization of the production process is still a requirement to meet the desired critical quality attributes. In this work, the production process of Gag VLPs by baculovirus (BV) infection in the reference High Five and Sf9 insect cell lines is characterized in detail. To this end, the Gag polyprotein was fused in frame to the enhanced green fluorescent protein (eGFP) to favor process evaluation with multiple analytical tools. Tracking of the infection process using confocal microscopy and flow cytometry revealed a pronounced increase in the complexity of High Five over Sf9 cells. Cryogenic transmission electron microscopy (cryo-TEM) characterization determined that changes in cell complexity could be attributed to the presence of occlusion-derived BV in High Five cells, whereas Sf9 cells evidenced a larger proportion of the budded virus phenotype (23-fold). Initial evaluation of the VLP production process using spectrofluorometry showed that higher levels of the Gag-eGFP polyprotein were obtained in High Five cells (3.6-fold). However, comparative analysis based on nanoparticle quantification by flow virometry and nanoparticle tracking analysis (NTA) proved that Sf9 cells were 1.7- and 1.5-fold more productive in terms of assembled VLPs, respectively. Finally, analytical ultracentrifugation coupled to flow virometry evidenced a larger sedimentation coefficient of High Five-derived VLPs, indicating a possible interaction with other cellular compounds. Taken together, these results highlight the combined use of microscopy and flow cytometry techniques to improve vaccine development processes using the insect cell/BV expression vector system. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Departament d'Enginyeria Química, Biològica i Ambiental, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Paolo Saccardo
- Plataforma de Producción de Proteínas, CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Neus Ferrer-Miralles
- Plataforma de Producción de Proteínas, CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Martí Lecina
- IQS School of Engineering, Universitat Ramón Llull, Barcelona, Spain
| | - Francesc Gòdia
- Departament d'Enginyeria Química, Biològica i Ambiental, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
97
|
Cheng K, Kang Q, Zhao X. Biogenic nanoparticles as immunomodulator for tumor treatment. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1646. [DOI: 10.1002/wnan.1646] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST) Beijing China
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province College of Materials, Xiamen University Xiamen Fujian China
| | - Qinglin Kang
- School of Chemistry and Molecular Biosciences The University of Queensland Brisbane Queensland Australia
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST) Beijing China
| |
Collapse
|
98
|
Kelly HG, Tan HX, Juno JA, Esterbauer R, Ju Y, Jiang W, Wimmer VC, Duckworth BC, Groom JR, Caruso F, Kanekiyo M, Kent SJ, Wheatley AK. Self-assembling influenza nanoparticle vaccines drive extended germinal center activity and memory B cell maturation. JCI Insight 2020; 5:136653. [PMID: 32434990 DOI: 10.1172/jci.insight.136653] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/15/2020] [Indexed: 01/10/2023] Open
Abstract
Protein-based, self-assembling nanoparticles elicit superior immunity compared with soluble protein vaccines, but the immune mechanisms underpinning this effect remain poorly defined. Here, we investigated the immunogenicity of a prototypic ferritin-based nanoparticle displaying influenza hemagglutinin (HA) in mice and macaques. Vaccination of mice with HA-ferritin nanoparticles elicited higher serum antibody titers and greater protection against experimental influenza challenge compared with soluble HA protein. Germinal centers in the draining lymph nodes were expanded and persistent following HA-ferritin vaccination, with greater deposition of antigen that colocalized with follicular dendritic cells. Our findings suggest that a highly ordered and repetitive antigen array may directly drive germinal centers through a B cell-intrinsic mechanism that does not rely on ferritin-specific T follicular helper cells. In contrast to mice, enhanced immunogenicity of HA-ferritin was not observed in pigtail macaques, where antibody titers and lymph node immunity were comparable to soluble vaccination. An improved understanding of factors that drive nanoparticle vaccine immunogenicity in small and large animal models will facilitate the clinical development of nanoparticle vaccines for broad and durable protection against diverse pathogens.
Collapse
Affiliation(s)
- Hannah G Kelly
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and
| | - Yi Ju
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and.,Department of Chemical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Wenbo Jiang
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | | | - Brigette C Duckworth
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Joanna R Groom
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and.,Department of Chemical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and.,Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and
| |
Collapse
|
99
|
Identification of Novel Adjuvants for Ebola Virus-Like Particle Vaccine. Vaccines (Basel) 2020; 8:vaccines8020215. [PMID: 32397625 PMCID: PMC7349346 DOI: 10.3390/vaccines8020215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 11/16/2022] Open
Abstract
Ebola virus disease is a severe disease, often fatal, with a mortality rate of up to 90%. Presently, effective treatment and safe prevention options for Ebola virus disease are not available. Therefore, there is an urgent need to develop control measures to prevent or limit future Ebola virus outbreaks. Ebola virus protein-based virus-like particle (VLP) and inactivated whole virion vaccines have demonstrated efficacy in animal models, and the addition of appropriate adjuvants may provide additional benefits to these vaccines, including enhanced immune responses. In this study, we screened 24 compounds from injectable excipients approved for human use in Japan and identified six compounds that significantly enhanced the humoral response to Ebola VLP vaccine in a murine model. Our novel adjuvant candidates for Ebola VLP vaccine have already been demonstrated to be safe when administered intramuscularly or subcutaneously, and therefore, they are closer to clinical trials than adjuvants whose safety profiles are unknown.
Collapse
|
100
|
Jonsdottir S, Fettelschoss V, Olomski F, Talker SC, Mirkovitch J, Rhiner T, Birkmann K, Thoms F, Wagner B, Bachmann MF, Kündig TM, Marti E, Fettelschoss-Gabriel A. Safety Profile of a Virus-Like Particle-Based Vaccine Targeting Self-Protein Interleukin-5 in Horses. Vaccines (Basel) 2020; 8:vaccines8020213. [PMID: 32397549 PMCID: PMC7349629 DOI: 10.3390/vaccines8020213] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Insect bite hypersensitivity (IBH) is an eosinophilic allergic dermatitis of horses caused by type I/IVb reactions against mainly Culicoides bites. The vaccination of IBH-affected horses with equine IL-5 coupled to the Cucumber mosaic virus-like particle (eIL-5-CuMVTT) induces IL-5-specific auto-antibodies, resulting in a significant reduction in eosinophil levels in blood and clinical signs. Objective: the preclinical and clinical safety of the eIL-5-CuMVTT vaccine. Methods: The B cell responses were assessed by longitudinal measurement of IL-5- and CuMVTT-specific IgG in the serum and plasma of vaccinated and unvaccinated horses. Further, peripheral blood mononuclear cells (PBMCs) from the same horses were re-stimulated in vitro for the proliferation and IFN-γ production of specific T cells. In addition, we evaluated longitudinal kidney and liver parameters and the general blood status. An endogenous protein challenge was performed in murine IL-5-vaccinated mice. Results: The vaccine was well tolerated as assessed by serum and cellular biomarkers and also induced reversible and neutralizing antibody titers in horses and mice. Endogenous IL-5 stimulation was unable to re-induce anti-IL-5 production. The CD4+ T cells of vaccinated horses produced significantly more IFN-γ and showed a stronger proliferation following stimulation with CuMVTT as compared to the unvaccinated controls. Re-stimulation using E. coli-derived proteins induced low levels of IFNγ+CD4+ cells in vaccinated horses; however, no IFN-γ and proliferation were induced following the HEK-eIL-5 re-stimulation. Conclusions: Vaccination using eIL-5-CuMVTT induces a strong B-cell as well as CuMVTT-specific T cell response without the induction of IL-5-specific T cell responses. Hence, B-cell unresponsiveness against self-IL-5 can be bypassed by inducing CuMVTT carrier-specific T cells, making the vaccine a safe therapeutic option for IBH-affected horses.
Collapse
Affiliation(s)
- Sigridur Jonsdottir
- Clinical Immunology Group, Department for Clinical Research VPH, Vetsuisse Faculty of the University of Bern, Länggassstrasse 124, 3012 Bern, Switzerland; (S.J.); (J.M.); (E.M.)
- Department of Dermatology, University Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (V.F.); (F.O.); (T.R.); (F.T.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Victoria Fettelschoss
- Department of Dermatology, University Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (V.F.); (F.O.); (T.R.); (F.T.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
- Evax AG, Hörnlistrass 3, 9542 Münchwilen, Switzerland;
| | - Florian Olomski
- Department of Dermatology, University Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (V.F.); (F.O.); (T.R.); (F.T.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Stephanie C. Talker
- Institute of Virology and Immunology, Länggassstrasse 122, 3012 Bern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Jelena Mirkovitch
- Clinical Immunology Group, Department for Clinical Research VPH, Vetsuisse Faculty of the University of Bern, Länggassstrasse 124, 3012 Bern, Switzerland; (S.J.); (J.M.); (E.M.)
| | - Tanya Rhiner
- Department of Dermatology, University Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (V.F.); (F.O.); (T.R.); (F.T.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | | | - Franziska Thoms
- Department of Dermatology, University Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (V.F.); (F.O.); (T.R.); (F.T.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Bettina Wagner
- Departments of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853-0001, USA;
| | - Martin F. Bachmann
- RIA Immunology, Inselspital, University of Bern, 3012 Bern, Switzerland;
- Jenner Institute, Nuffield Department of Medicine, Henry Welcome Building for Molecular Physiology, University of Oxford, OX1 2JD Oxford, UK
| | - Thomas M. Kündig
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland;
| | - Eliane Marti
- Clinical Immunology Group, Department for Clinical Research VPH, Vetsuisse Faculty of the University of Bern, Länggassstrasse 124, 3012 Bern, Switzerland; (S.J.); (J.M.); (E.M.)
| | - Antonia Fettelschoss-Gabriel
- Department of Dermatology, University Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (V.F.); (F.O.); (T.R.); (F.T.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
- Evax AG, Hörnlistrass 3, 9542 Münchwilen, Switzerland;
- Correspondence:
| |
Collapse
|