1
|
Casmil IC, Bathula NV, Huang C, Wayne CJ, Cairns ES, Friesen JJ, Soriano SK, Liao S, Ho CH, Kong KYS, Blakney AK. Alphaviral backbone of self-amplifying RNA enhances protein expression and immunogenicity against SARS-CoV-2 antigen. Mol Ther 2025; 33:514-528. [PMID: 39741413 PMCID: PMC11852984 DOI: 10.1016/j.ymthe.2024.12.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Self-amplifying RNA (saRNA) vectors are a next-generation RNA technology that extends the expression of heterologous genes. Clinical trials have shown the dose-sparing capacity of saRNA vectors in a vaccine context compared with conventional messenger RNA. However, saRNA vectors have historically been based on a limited number of alphaviruses, and only the Venezuelan equine encephalitis virus-based saRNA vaccines have been used clinically. Here, we designed genotypically distinct alphaviral saRNA vectors and characterized their performance in mammalian cell lines, human skin explants and mice. Five of the 12 vectors had substantial luciferase expression in mice with variable pharmacokinetics, enabling modulation of both the magnitude and duration of protein expression. Additionally, we demonstrated that the alphaviral genotype of the saRNA significantly impacts the immunogenicity of saRNA vaccines, including the humoral and cellular responses in mice. Given the differences in RNA reactogenicity and expression between mice and humans, we assessed the saRNA vectors in human skin explants obtained from patients and observed high transgene expression. saRNA bioluminescence and immunogenicity in different mice strains were highly correlative, while minimal correlation was observed when compared with human explants and mammalian cell lines. This work demonstrates that efficacious saRNA vaccines and therapies can be produced by adapting genetically diverse alphaviruses into vectors.
Collapse
Affiliation(s)
- Irafasha C Casmil
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Nuthan V Bathula
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Cynthia Huang
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Christopher J Wayne
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Evan S Cairns
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Josh J Friesen
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Shekinah K Soriano
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Suiyang Liao
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada; Life Science Institute, University of British Columbia, Vancouver V6T1Z3, BC, Canada
| | - Chia H Ho
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Kristen Y S Kong
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada
| | - Anna K Blakney
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver V6T1Z4, BC, Canada.
| |
Collapse
|
2
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
3
|
Vasquez R, Song JH, Mendoza RM, Hwang I, Bagon BB, Engstrand L, Valeriano VD, Kang D. Oral Immunisation With Non-GMO Surface Displayed SARS-CoV-2 Spike Epitopes on Bacteria-Like Particles Provokes Robust Humoral and Cellular Immune Responses, and Modulated the Gut Microbiome in Mice. Microb Biotechnol 2025; 18:e70073. [PMID: 39797809 PMCID: PMC11724470 DOI: 10.1111/1751-7915.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a fatal disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). To date, several vaccines have been developed to combat the spread of this virus. Mucosal vaccines using food-grade bacteria, such as Lactobacillus spp., are promising strategies for developing safe and effective vaccines against SARS-CoV-2. In this study, we designed a non-GMO surface-displayed SARS-CoV-2 spike S1 epitope on Limosilactobacillus fermentum-derived bacteria-like particles (BLPs). After that, we evaluated its efficacy to induce immune responses in immunocompetent mice. Moreover, we examined the influence of oral immunisation on the gut microbiome and microbiota metabolites. Twenty-eight 6-week-old male C57BL/6 mice were orally immunised with the following: PBS (control), Lm. fermentum-derived BLPs only, BLPs displaying SARS-CoV-2 spike S1-2, or BLPs displaying SARS-CoV-2 spike S1-3 epitopes. Our results showed that mucosal immunisation of mice with surface-displayed SARS-CoV-2 spike epitopes provoked high-level secretory IgA and systemic IgG production. Moreover, the immunisation exhibited a Th1-like immune response, characterised by an elevated IgG2a-to-IgG1 ratio and high antiviral IFN-γ production. In addition, we observed gut microbiome modulation and increased butyrate production in immunised mice. Overall, the use of Lm. fermentum-derived BLPs and the anchor CshA to display SARS-CoV-2 spike S1epitopes is a promising novel strategy in developing a cost-effective, non-GMO mucosal vaccine alternative against SARS-CoV-2.
Collapse
Affiliation(s)
- Robie Vasquez
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
| | - Ji Hoon Song
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
| | | | - In‐Chan Hwang
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR)Karolinska InstitutetStockholmSweden
| | | | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR)Karolinska InstitutetStockholmSweden
| | - Valerie Diane Valeriano
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR)Karolinska InstitutetStockholmSweden
| | - Dae‐Kyung Kang
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
| |
Collapse
|
4
|
Silva-Pilipich N, Beloki U, Salaberry L, Smerdou C. Self-Amplifying RNA: A Second Revolution of mRNA Vaccines against COVID-19. Vaccines (Basel) 2024; 12:318. [PMID: 38543952 PMCID: PMC10974399 DOI: 10.3390/vaccines12030318] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 11/12/2024] Open
Abstract
SARS-CoV-2 virus, the causative agent of COVID-19, has produced the largest pandemic in the 21st century, becoming a very serious health problem worldwide. To prevent COVID-19 disease and infection, a large number of vaccines have been developed and approved in record time, including new vaccines based on mRNA encapsulated in lipid nanoparticles. While mRNA-based vaccines have proven to be safe and effective, they are more expensive to produce compared to conventional vaccines. A special type of mRNA vaccine is based on self-amplifying RNA (saRNA) derived from the genome of RNA viruses, mainly alphaviruses. These saRNAs encode a viral replicase in addition to the antigen, usually the SARS-CoV-2 spike protein. The replicase can amplify the saRNA in transfected cells, potentially reducing the amount of RNA needed for vaccination and promoting interferon I responses that can enhance adaptive immunity. Preclinical studies with saRNA-based COVID-19 vaccines in diverse animal models have demonstrated the induction of robust protective immune responses, similar to conventional mRNA but at lower doses. Initial clinical trials have confirmed the safety and immunogenicity of saRNA-based vaccines in individuals that had previously received authorized COVID-19 vaccines. These findings have led to the recent approval of two of these vaccines by the national drug agencies of India and Japan, underscoring the promising potential of this technology.
Collapse
Affiliation(s)
- Noelia Silva-Pilipich
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Uxue Beloki
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Laura Salaberry
- Facultad de Ingeniería, Universidad ORT Uruguay, Montevideo 11100, Uruguay;
- Nanogrow Biotech, Montevideo 11500, Uruguay
| | - Cristian Smerdou
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| |
Collapse
|
5
|
Najary S, Vatankhah M, Khadivi G, Salehi SN, Tabari MAK, Samieefar N, Behnaz M. A comprehensive review of oral microenvironment changes and orofacial adverse reactions after COVID-19 vaccination: The good, the bad, and the ugly. Health Sci Rep 2024; 7:e1967. [PMID: 38482134 PMCID: PMC10935892 DOI: 10.1002/hsr2.1967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 02/04/2024] [Accepted: 02/26/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND AND AIMS Anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines have the potential to alter several biological systems concurrently with remolding the immune system, most of which are related to immunization, while some others are known as adverse effects. This review aims to explore the potential effects of vaccination on the oral microenvironment and classifies them as good, bad, or ugly, with a brief review of facial diseases following coronavirus disease 2019 (COVID-19) vaccination. METHODS This study was a comprehensive review conducted through searching related articles in Medline, Scopus, and Google Scholar databases. RESULTS On one side, the "Good" impacts of vaccination on the oro-nasal mucosa are explained as if the mucosal immune responses followed by SARS-CoV-2 vaccines are enough to provide immunity. On the other side, the possible "Bad" and "Ugly" effects of the vaccine, which manifest as orofacial adverse events and autoimmune reactivations, respectively, should be noted. Exacerbation of pre-existing autoimmune conditions such as lichen planus, pemphigus vulgaris, bullous pemphigoid, and Stevens-Johnson syndrome have been reported. CONCLUSION COVID-19 vaccines could affect different biological systems alongside stimulating the immune system, and some of these effects are referred to as adverse effects. Nonetheless, these adverse effects are treatable, and healthcare professionals should not prevent patients from taking the first available vaccination.
Collapse
Affiliation(s)
- Shaghayegh Najary
- School of DentistryShahid Beheshti University of Medical SciencesTehranIran
- USERN OfficeShahid Beheshti University of Medical SciencesTehranIran
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Mohammadreza Vatankhah
- Center for Craniofacial Molecular Biology, Herman Ostrow School of DentistryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Gita Khadivi
- School of DentistryShahid Beheshti University of Medical SciencesTehranIran
- USERN OfficeShahid Beheshti University of Medical SciencesTehranIran
| | - Seyyede N. Salehi
- USERN OfficeShahid Beheshti University of Medical SciencesTehranIran
- Dentistry Student, Executive Secretary of Research Committee, Board Director of Scientific Society, Dental FacultyIslamic Azad UniversityTehranIran
| | - Mohammad A. K. Tabari
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
- Student Research CommitteeMazandaran University of Medical SciencesSariIran
- USERN OfficeMazandaran University of Medical SciencesSariIran
| | - Noosha Samieefar
- USERN OfficeShahid Beheshti University of Medical SciencesTehranIran
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Mohammad Behnaz
- USERN OfficeShahid Beheshti University of Medical SciencesTehranIran
- Dental Research Center, Research Institute of Dental Sciences, School of DentistryShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
6
|
Moukafih B, Belaroussi L, Achour S, Kartouti AE. Side Effects Reported by Moroccan Medical Students Who Received COVID-19 Vaccines. Curr Drug Saf 2024; 19:268-276. [PMID: 37138485 DOI: 10.2174/1574886318666230503113713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND Low confidence in the safety of COVID-19 vaccines was found to be a key promoter of vaccine reluctance especially among youth. Furthermore, young adults are an important demographic for building herd immunity through vaccination. As a result, their reactions to getting COVID-19 vaccines are crucial in our fight against SARS-CoV-2. OBJECTIVE The overall goal of this study was to look into the shortterm side effects experienced by Moroccan medical and pharmacy students after receiving COVID-19 vaccines. METHODS A cross-sectional survey-based study to assess the COVID-19 vaccines' short-term AEFIs among Moroccan medical and pharmacy students. The validated questionnaire was delivered in a digital form to explore the side effects (SE) they encountered after the first or the second dose of one of three vaccines namely: AstraZeneca Vaxzevria, PfizerBioNTeck, and SinoPharm vaccines. RESULTS There were 510 students in total who took part. After the first and second doses, approximately 72 percent and 78 percent of subjects, respectively, reported no SE. The remainder had localized injection site side effects (26%). Fatigue (21%), fever (19%), headache (17%), and myalgia (16%) were the most common systemic adverse effects after the first dose. There were no serious SEs reported. CONCLUSION The majority of the reported AEFIs in our data were mild to moderate in intensity and lasted only one or two days. COVID-19 vaccinations are highly likely safe for young adults, according to the findings of this study.
Collapse
Affiliation(s)
- Badreddine Moukafih
- Central Pharmacy Department, CHU Hassan II, Medical Center for Biomedical and Translational Research, Faculty of Medicine, Pharmacy and dentistry of Fez, Sidi Mohammed Ben Abdellah University, Fez, Morocco
| | - Leila Belaroussi
- Occupational Health Department, CHU Hassan II-Fez, Epidemiology and Health Sciences Research Laboratory, Faculty of Medicine and Pharmacy of Fez, Sidi Mohammed Ben Abdellah University, Fez, Morocco
| | - Sanae Achour
- Toxicology Department, CHU Hassan II, Fez, Medical Center for Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dentistry of Fez, Sidi Mohammed Ben Abdellah University, Morocco Hospital, Fez, Morocco
| | - Abdeslam El Kartouti
- Pharmacy Service, Moulay Ismaïl Military Hospital, Meknès, Medical Center for Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dentistry of Fez, Sidi Mohammed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
7
|
Bellocchio L, Dipalma G, Inchingolo AM, Inchingolo AD, Ferrante L, Del Vecchio G, Malcangi G, Palermo A, Qendro A, Inchingolo F. COVID-19 on Oral Health: A New Bilateral Connection for the Pandemic. Biomedicines 2023; 12:60. [PMID: 38255167 PMCID: PMC10813615 DOI: 10.3390/biomedicines12010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/14/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission are generally known to be produced by respiratory droplets and aerosols from the oral cavity (O.C.) of infected subjects, as stated by the World Health Organization. Saliva also retains the viral particles and aids in the spread of COVID-19. Angiotensin-converting enzyme Type 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) are two of the numerous factors that promote SARS-CoV-2 infection, expressed by O.C. structures, various mucosa types, and the epithelia of salivary glands. A systemic SARS-CoV-2 infection might result from viral replication in O.C. cells. On the other hand, cellular damage of different subtypes in the O.C. might be associated with various clinical signs and symptoms. Factors interfering with SARS-CoV-2 infection potential might represent fertile ground for possible local pharmacotherapeutic interventions, which may confine SARS-CoV-2 virus entry and transmission in the O.C., finally representing a way to reduce COVID-19 incidence and severity.
Collapse
Affiliation(s)
- Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, University of Bordeaux, 33063 Bordeaux, France;
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Laura Ferrante
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Gaetano Del Vecchio
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Andis Qendro
- Faculty of Dental Medicine, University of Medicine, 1005 Tirana, Albania;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| |
Collapse
|
8
|
Yu X, Kong Q. Potential value of neuroimmunotherapy for COVID-19: efficacies and mechanisms of vagus nerve stimulation, electroacupuncture, and cholinergic drugs. Front Immunol 2023; 14:1197467. [PMID: 37475861 PMCID: PMC10355152 DOI: 10.3389/fimmu.2023.1197467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/23/2023] [Indexed: 07/22/2023] Open
Abstract
COVID-19 is an inflammatory disease with multiple organs involved, mainly respiratory symptoms. Although the majority of patients with COVID-19 present with a mild to moderate self-limited course of illness, about 5-10% of patients with inflammatory disorders in severe COVID-19 have life-threatening progression. With the exception of a few drugs that have shown outstanding anti-COVID-19 effects, the efficacy of most drugs remains controversial. An increasing number of animal and clinical studies have shown that neuromodulation has a significant effect on reducing inflammatory markers of COVID-19, thus exerting an effective neuroimmunotherapeutic value. Currently, the main neuroimmunomodulatory measures effective against COVID-19 include vagus nerve stimulation, electroacupuncture, and cholinergic drugs. In this review, we will summarize the research progress of potential value of this neuroimmunotherapy measures for COVID-19 and elaborate its efficacies and mechanisms, in order to provide reliable evidence for clinical intervention.
Collapse
Affiliation(s)
- Xianqiang Yu
- Women and Children's Hospital Affiliated to Qingdao University, Heart center, Qingdao, China
- University of California, Los Angeles, Department of Cardiology, Los Angeles, CA, United States
| | - Qingming Kong
- School of Laboratory Medicine and Bioengineering, Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang province, Key Laboratory of Bio-tech Vaccine of Zhejiang Province, Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
9
|
Letafati A, Eyvazzadeh N, Gharehkhani A, Khorshidian A, Chalabiani S, Soufiani EK, Khakpoor N, Shamsodini B, Beheshti T, Bavili Olyaei RT, Soleimani A, Melyani F, Hossein GM. Comparison of AstraZeneca and sinopharm vaccines as boosters in protection against COVID-19 infection. Biologicals 2023; 82:101668. [PMID: 37004277 PMCID: PMC10008804 DOI: 10.1016/j.biologicals.2023.101668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/19/2022] [Accepted: 03/06/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND As the global number of confirmed cases rises past 640 million, vaccination remains the most effective measure in controlling COVID-19. Studies have shown that two doses of vaccination can significantly reduce hospitalization and mortality rates among patients, but the effectiveness of booster doses is also important. We aimed to evaluate the role played by the type of the 3rd dose of vaccination by comparing the safety and efficacy of two common vaccination histories differing only in the 3rd received dose. METHODS We conducted a cross-sectional study on patients with respiratory symptoms suspected of having SARS-CoV-2 infection using Real-time PCR. We also collected information on the age, gender, and type of vaccine received for the third dose. RESULTS Out of 346 cases with respiratory symptoms, 120 cases tested positive for SARS-CoV-2 and had received two doses of Sinopharm and a different booster dose of either AZD1222 (AstraZeneca) or BIBP (Sinopharm). Among these 120 patients, vaccination with AZD1222 as a booster dose resulted in fewer symptoms compared to those vaccinated with three doses of BIBP. CONCLUSIONS Our study demonstrates that booster doses can help reduce hospitalization and the severity of infection, and it appears that a combination of different vaccines may be effective against severe COVID-19 infection.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nooshin Eyvazzadeh
- Department of Medical Bacteriology, Faculty of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Amirhossein Gharehkhani
- Department of Medical Laboratory Science, Faculty of Para Medicine, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Ayeh Khorshidian
- Department of Biology, Faculty of Basic Sciences, Gonbad Kavous University, Gonbad Kavous, Iran
| | - Siavash Chalabiani
- Department of Microbiology, Faculty of Biological Sciences, Islamic Azad University of Qom, Qom, Iran
| | - Elnaz Khodadoust Soufiani
- Department of Medical Laboratory Science, Faculty of Para Medicine, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Niloofar Khakpoor
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benyamin Shamsodini
- Department of Microbiology, Faculty of Basic Sciences, Islamic Azad University of Shahrekord, Shahrekord, Iran
| | - Taranom Beheshti
- Department of Genetics, Faculty of Advanced Sciences and Technology, Islamic Azad University of Tehran, Tehran, Iran
| | - Raha Taheri Bavili Olyaei
- Department of Medical Laboratory Science, Faculty of Medical Sciences, Islamic Azad University, Tabriz Medical Branch, Tabriz, Iran
| | - Anahita Soleimani
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Sarab University, Sarab, Iran
| | - Fatemeh Melyani
- Department of Medical Laboratory Sciences, Faculty of Para Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Ghazal Mashhadi Hossein
- Department of Microbiology, Faculty of Basic Sciences, Islamic Azad University of Arak, Arak, Iran
| |
Collapse
|
10
|
Bhattacharjee B, Ikbal AMA, Farooqui A, Sahu RK, Ruhi S, Syed A, Miatmoko A, Khan D, Khan J. Superior possibilities and upcoming horizons for nanoscience in COVID-19: noteworthy approach for effective diagnostics and management of SARS-CoV-2 outbreak. CHEMICKE ZVESTI 2023; 77:1-24. [PMID: 37362791 PMCID: PMC10072050 DOI: 10.1007/s11696-023-02795-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/18/2023] [Indexed: 04/07/2023]
Abstract
The outbreak of COVID-19 has caused great havoc and affected many parts of the world. It has imposed a great challenge to the medical and health fraternity with its ability to continue mutating and increasing the transmission rate. Some challenges include the availability of current knowledge of active drugs against the virus, mode of delivery of the medicaments, its diagnosis, which are relatively limited and do not suffice for further prognosis. One recently developed drug delivery system called nanoparticles is currently being utilized in combating COVID-19. This article highlights the existing methods for diagnosis of COVID-19 such as computed tomography scan, reverse transcription-polymerase chain reaction, nucleic acid sequencing, immunoassay, point-of-care test, detection from breath, nanotechnology-based bio-sensors, viral antigen detection, microfluidic device, magnetic nanosensor, magnetic resonance platform and internet-of-things biosensors. The latest detection strategy based on nanotechnology, biosensor, is said to produce satisfactory results in recognizing SARS-CoV-2 virus. It also highlights the successes in the research and development of COVID-19 treatments and vaccines that are already in use. In addition, there are a number of nanovaccines and nanomedicines currently in clinical trials that have the potential to target COVID-19.
Collapse
Affiliation(s)
- Bedanta Bhattacharjee
- Girijananda Chowdhury Institute of Pharmaceutical Science, Tezpur, Assam 784501 India
| | - Abu Md Ashif Ikbal
- Department of Pharmaceutical Sciences, Assam University (A Central University), Silchar, 788011 India
| | - Atika Farooqui
- The Deccan College of Medical Sciences, Kanchan Bagh, Hyderabad, Telangana 500058 India
| | - Ram Kumar Sahu
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Chauras Campus, Tehri Garhwal, Uttarakhand 249161 India
| | - Sakina Ruhi
- Department of Biochemistry, IMS, Management and Science University, University Drive, Off Persiaran Olahraga, 40100 Shah Alam, Selangor Malaysia
| | - Ayesha Syed
- International Medical School, Management and Science University, University Drive, Off Persiaran Olahraga, 40100 Shah Alam, Selangor Malaysia
| | - Andang Miatmoko
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, East Java 60115 Indonesia
| | - Danish Khan
- Panineeya Institute of Dental Science and Research Centre, Kalonji Narayana Rao University of Health Sciences, Warangal, Telangana 506007 India
| | - Jiyauddin Khan
- School of Pharmacy, Management and Science University, 40100 Shah Alam, Selangor Malaysia
| |
Collapse
|
11
|
Chavda VP, Jogi G, Dave S, Patel BM, Vineela Nalla L, Koradia K. mRNA-Based Vaccine for COVID-19: They Are New but Not Unknown! Vaccines (Basel) 2023; 11:507. [PMID: 36992091 PMCID: PMC10052021 DOI: 10.3390/vaccines11030507] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
mRNA vaccines take advantage of the mechanism that our cells use to produce proteins. Our cells produce proteins based on the knowledge contained in our DNA; each gene encodes a unique protein. The genetic information is essential, but cells cannot use it until mRNA molecules convert it into instructions for producing specific proteins. mRNA vaccinations provide ready-to-use mRNA instructions for constructing a specific protein. BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna) both are newly approved mRNA-based COVID-19 vaccines that have shown excellent protection and efficacy. In total, there are five more mRNA-based vaccine candidates for COVID-19 under different phases of clinical development. This review is specifically focused on mRNA-based vaccines for COVID-19 covering its development, mechanism, and clinical aspects.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad 380009, India
| | - Gargi Jogi
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad 380009, India
| | - Srusti Dave
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | - Bhoomika M. Patel
- School of Medico-legal Studies, National Forensic Sciences University, Gandhinagar 382007, India
| | - Lakshmi Vineela Nalla
- Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India
| | - Krishna Koradia
- Department of Pharmaceutics, Saurashtra University, Rajkot 360005, India
| |
Collapse
|
12
|
de Matos PH, da Silva TP, Mansano AB, Gancedo NC, Tonin FS, Pelloso FC, Petruco MV, de Melo EB, Fernandez-Llimos F, Sanches ACC, de Mello JCP, Chierrito D, de Medeiros Araújo DC. Bioactive compounds as potential angiotensin-converting enzyme II inhibitors against COVID-19: a scoping review. Inflamm Res 2022; 71:1489-1500. [PMID: 36307652 PMCID: PMC9616414 DOI: 10.1007/s00011-022-01642-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/29/2022] [Accepted: 09/10/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE AND DESIGN The current study aimed to summarize the evidence of compounds contained in plant species with the ability to block the angiotensin-converting enzyme 2 (ACE-II), through a scoping review. METHODS PubMed and Scopus electronic databases were used for the systematic search and a manual search was performed RESULTS: Studies included were characterized as in silico. Among the 200 studies retrieved, 139 studies listed after the exclusion of duplicates and 74 were included for the full read. Among them, 32 studies were considered eligible for the qualitative synthesis. The most evaluated class of secondary metabolites was flavonoids with quercetin and curcumin as most actives substances and terpenes (isothymol, limonin, curcumenol, anabsinthin, and artemisinin). Other classes that were also evaluated were alkaloid, saponin, quinone, substances found in essential oils, and primary metabolites as the aminoacid L-tyrosine and the lipidic compound 2-monolinolenin. CONCLUSION This review suggests the most active substance from each class of metabolites, which presented the strongest affinity to the ACE-II receptor, what contributes as a basis for choosing compounds and directing the further experimental and clinical investigation on the applications these compounds in biotechnological and health processes as in COVID-19 pandemic.
Collapse
Affiliation(s)
- Pedro Henrique de Matos
- Centro Universitário Ingá-UNINGÁ, Rodovia PR 317, 6114. Parque Industrial, 200, Maringá,, PR, 87035-510, Brazil
| | - Thalita Prates da Silva
- Departamento de Farmácia, Universidade Estadual de Maringá, Avenida Colombo, Maringá, 5790, Brazil
| | - Amanda Benites Mansano
- Departamento de Farmácia, Universidade Estadual de Maringá, Avenida Colombo, Maringá, 5790, Brazil
| | - Naiara Cássia Gancedo
- Departamento de Farmácia, Universidade Estadual de Maringá, Avenida Colombo, Maringá, 5790, Brazil
| | - Fernanda Stumpf Tonin
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Paraná, Avenida Prefeito Lothário Meissner 632, Curitiba, Brazil
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Fernando Castilho Pelloso
- Complexo Hospital de Clínicas, Universidade Federal Do Paraná, Rua General Carneiro, Curitiba, 181, Brazil
| | | | - Eduardo Borges de Melo
- Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Rua Universitário 2069, Cascavel, Brazil
| | - Fernando Fernandez-Llimos
- Departamento de Ciências do Medicamento, Universidade do Porto, Praça Gomes Teixeira, Porto, Portugal
| | | | | | - Danielly Chierrito
- Centro Universitário Ingá-UNINGÁ, Rodovia PR 317, 6114. Parque Industrial, 200, Maringá,, PR, 87035-510, Brazil
| | | |
Collapse
|
13
|
Dhama K, Dhawan M, Tiwari R, Emran TB, Mitra S, Rabaan AA, Alhumaid S, Alawi ZA, Al Mutair A. COVID-19 intranasal vaccines: current progress, advantages, prospects, and challenges. Hum Vaccin Immunother 2022; 18:2045853. [PMID: 35258416 PMCID: PMC8935456 DOI: 10.1080/21645515.2022.2045853] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Multiple vaccines have recently been developed, and almost all the countries are presently vaccinating their population to tackle the COVID-19 pandemic. Most of the COVID-19 vaccines in use are administered via intramuscular (IM) injection, eliciting protective humor and cellular immunity. COVID-19 intranasal (IN) vaccines are also being developed that have shown promising ability to induce a significant amount of antibody-mediated immune response and a robust cell-mediated immunity as well as hold the added ability to stimulate protective mucosal immunity along with the additional advantage of the ease of administration as compared to IM injected vaccines. By inducing secretory IgA antibody responses specifically in the nasal compartment, the intranasal SARS-CoV-2 vaccine can prevent virus infection, replication, shedding, and disease development, as well as possibly limits virus transmission. This article highlights the current progress, advantages, prospects, and challenges in developing intranasal COVID-19 vaccines for countering the ongoing pandemic.
Collapse
Affiliation(s)
- Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- The Trafford Group of Colleges, Manchester, UK
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, India
| | - Talha Bin Emran
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa, Saudi Arabia
| | - Zainab Al Alawi
- Division of Allergy and Immunology, College of Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa, Saudi Arabia
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, Australia
| |
Collapse
|
14
|
Valenzuela-Fernández A, Cabrera-Rodriguez R, Ciuffreda L, Perez-Yanes S, Estevez-Herrera J, González-Montelongo R, Alcoba-Florez J, Trujillo-González R, García-Martínez de Artola D, Gil-Campesino H, Díez-Gil O, Lorenzo-Salazar JM, Flores C, Garcia-Luis J. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Front Bioeng Biotechnol 2022; 10:1052436. [PMID: 36507266 PMCID: PMC9732709 DOI: 10.3389/fbioe.2022.1052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the associated coronavirus disease 2019 (COVID-19), which severely affect the respiratory system and several organs and tissues, and may lead to death, have shown how science can respond when challenged by a global emergency, offering as a response a myriad of rapid technological developments. Development of vaccines at lightning speed is one of them. SARS-CoV-2 outbreaks have stressed healthcare systems, questioning patients care by using standard non-adapted therapies and diagnostic tools. In this scenario, nanotechnology has offered new tools, techniques and opportunities for prevention, for rapid, accurate and sensitive diagnosis and treatment of COVID-19. In this review, we focus on the nanotechnological applications and nano-based materials (i.e., personal protective equipment) to combat SARS-CoV-2 transmission, infection, organ damage and for the development of new tools for virosurveillance, diagnose and immune protection by mRNA and other nano-based vaccines. All the nano-based developed tools have allowed a historical, unprecedented, real time epidemiological surveillance and diagnosis of SARS-CoV-2 infection, at community and international levels. The nano-based technology has help to predict and detect how this Sarbecovirus is mutating and the severity of the associated COVID-19 disease, thereby assisting the administration and public health services to make decisions and measures for preparedness against the emerging variants of SARS-CoV-2 and severe or lethal COVID-19.
Collapse
Affiliation(s)
- Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Romina Cabrera-Rodriguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Laura Ciuffreda
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Perez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Judith Estevez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | - Julia Alcoba-Florez
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departamento de Análisis Matemático, Facultad de Ciencias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Helena Gil-Campesino
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Oscar Díez-Gil
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Jonay Garcia-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
15
|
Miteva D, Peshevska-Sekulovska M, Snegarova V, Batselova H, Alexandrova R, Velikova T. Mucosal COVID-19 vaccines: Risks, benefits and control of the pandemic. World J Virol 2022; 11:221-236. [PMID: 36188733 PMCID: PMC9523321 DOI: 10.5501/wjv.v11.i5.221] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/14/2022] [Accepted: 08/10/2022] [Indexed: 02/05/2023] Open
Abstract
Based on mucosal immunization to promote both mucosal and systemic immune responses, next-generation coronavirus disease 2019 (COVID-19) vaccines would be administered intranasally or orally. The goal of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is to provide adequate immune protection and avoid severe disease and death. Mucosal vaccine candidates for COVID-19 including vector vaccines, recombinant subunit vaccines and live attenuated vaccines are under development. Furthermore, subunit protein vac-cines and virus-vectored vaccines have made substantial progress in preclinical and clinical settings, resulting in SARS-CoV-2 intranasal vaccines based on the previously successfully used nasal vaccines. Additional to their ability to trigger stable, protective immune responses at the sites of pathogenic infection, the development of 'specific' mucosal vaccines targeting coronavirus antigens could be an excellent option for preventing future pandemics. However, their efficacy and safety should be confirmed.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Department of Genetics, Sofia University “St. Kliment Ohridski,” Faculty of Biology, Sofia 1164, Bulgaria
| | - Monika Peshevska-Sekulovska
- Department of Gastroenterology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Violeta Snegarova
- Clinic of Internal Diseases, Naval Hospital - Varna, Military Medical Academy, Medical Faculty, Medical University, Varna 9000, Bulgaria
| | - Hristiana Batselova
- Department of Epidemiology and Disaster Medicine, Medical University, Plovdiv, University Hospital “St George”, Plovdiv 6000, Bulgaria
| | - Radostina Alexandrova
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia 1000, Bulgaria
| | - Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
16
|
Inchingolo AD, Malcangi G, Ceci S, Patano A, Corriero A, Vimercati L, Azzollini D, Marinelli G, Coloccia G, Piras F, Barile G, Settanni V, Mancini A, De Leonardis N, Garofoli G, Palmieri G, Isacco CG, Rapone B, Scardapane A, Curatoli L, Quaranta N, Ribezzi M, Massaro M, Jones M, Bordea IR, Tartaglia GM, Scarano A, Lorusso F, Macchia L, Larocca AMV, Aityan SK, Tafuri S, Stefanizzi P, Migliore G, Brienza N, Dipalma G, Favia G, Inchingolo F. Effectiveness of SARS-CoV-2 Vaccines for Short- and Long-Term Immunity: A General Overview for the Pandemic Contrast. Int J Mol Sci 2022; 23:8485. [PMID: 35955621 PMCID: PMC9369331 DOI: 10.3390/ijms23158485] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The recent COVID-19 pandemic produced a significant increase in cases and an emergency state was induced worldwide. The current knowledge about the COVID-19 disease concerning diagnoses, patient tracking, the treatment protocol, and vaccines provides a consistent contribution for the primary prevention of the viral infection and decreasing the severity of the SARS-CoV-2 disease. The aim of the present investigation was to produce a general overview about the current findings for the COVID-19 disease, SARS-CoV-2 interaction mechanisms with the host, therapies and vaccines' immunization findings. METHODS A literature overview was produced in order to evaluate the state-of-art in SARS-CoV-2 diagnoses, prognoses, therapies, and prevention. RESULTS Concerning to the interaction mechanisms with the host, the virus binds to target with its Spike proteins on its surface and uses it as an anchor. The Spike protein targets the ACE2 cell receptor and enters into the cells by using a special enzyme (TMPRSS2). Once the virion is quietly accommodated, it releases its RNA. Proteins and RNA are used in the Golgi apparatus to produce more viruses that are released. Concerning the therapies, different protocols have been developed in observance of the disease severity and comorbidity with a consistent reduction in the mortality rate. Currently, different vaccines are currently in phase IV but a remarkable difference in efficiency has been detected concerning the more recent SARS-CoV-2 variants. CONCLUSIONS Among the many questions in this pandemic state, the one that recurs most is knowing why some people become more seriously ill than others who instead contract the infection as if it was a trivial flu. More studies are necessary to investigate the efficiency of the treatment protocols and vaccines for the more recent detected SARS-CoV-2 variant.
Collapse
Affiliation(s)
- Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Sabino Ceci
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Assunta Patano
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Alberto Corriero
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70121 Bari, Italy; (A.C.); (M.R.); (N.B.)
| | - Luigi Vimercati
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Daniela Azzollini
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Grazia Marinelli
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Giovanni Coloccia
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Fabio Piras
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Giuseppe Barile
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Vito Settanni
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Nicole De Leonardis
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Grazia Garofoli
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Giulia Palmieri
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Ciro Gargiulo Isacco
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Biagio Rapone
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Arnaldo Scardapane
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Luigi Curatoli
- Department Neurosciences & Sensory Organs & Musculoskeletal System, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Nicola Quaranta
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
- Department Neurosciences & Sensory Organs & Musculoskeletal System, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Mario Ribezzi
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70121 Bari, Italy; (A.C.); (M.R.); (N.B.)
| | - Maria Massaro
- Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, 70124 Bari, Italy;
| | - Megan Jones
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, Faculty of Dentistry, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Gianluca Martino Tartaglia
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy;
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Luigi Macchia
- Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari Aldo Moro, 70124 Bari, Italy;
| | - Angela Maria Vittoria Larocca
- Hygiene Complex Operating Unit, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, Place Giulio Cesare 11 BARI CAP, 70124 Bari, Italy;
| | | | - Silvio Tafuri
- Department of Biomedical Science and Human Oncology, University of Bari, 70121 Bari, Italy;
| | - Pasquale Stefanizzi
- Interdisciplinary Department of Medicine, University Hospital of Bari, 70100 Bari, Italy; (P.S.); (G.M.)
| | - Giovanni Migliore
- Interdisciplinary Department of Medicine, University Hospital of Bari, 70100 Bari, Italy; (P.S.); (G.M.)
| | - Nicola Brienza
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70121 Bari, Italy; (A.C.); (M.R.); (N.B.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Gianfranco Favia
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, Section of Dental Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (S.C.); (A.P.); (L.V.); (D.A.); (G.M.); (G.C.); (F.P.); (G.B.); (V.S.); (A.M.); (N.D.L.); (G.G.); (G.P.); (C.G.I.); (B.R.); (A.S.); (N.Q.); (M.J.); (G.D.); (G.F.)
| |
Collapse
|
17
|
Gastrointestinal Involvement in SARS-CoV-2 Infection. Viruses 2022; 14:v14061188. [PMID: 35746659 PMCID: PMC9228950 DOI: 10.3390/v14061188] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 has evolved into a virus that primarily results in mild or asymptomatic disease, making its transmission more challenging to control. In addition to the respiratory tract, SARS-CoV-2 also infects the digestive tract. Some gastrointestinal symptoms occur with or before respiratory symptoms in patients with COVID-19. Respiratory infections are known to cause intestinal immune impairment and gastrointestinal symptoms. When the intestine is inflamed, cytokines affect the lung immune response and inflammation through blood circulation. The gastrointestinal microbiome may be a modifiable factor in determining the risk of SARS-CoV-2 infection and disease severity. The development of oral SARS-CoV-2 vaccine candidates and the maintenance of gut microbiota profiles may contribute to the early control of COVID-19 outbreaks. To this end, this review summarizes information on the gastrointestinal complications caused by SARS-CoV-2, SARS-CoV-2 infection, the gastrointestinal–lung axis immune response, potential control strategies for oral vaccine candidates and maintaining intestinal microbiota homeostasis.
Collapse
|
18
|
Jt S, M H, Wam B, Ac B, Sa N. Adenoviral vectors for cardiovascular gene therapy applications: a clinical and industry perspective. J Mol Med (Berl) 2022; 100:875-901. [PMID: 35606652 PMCID: PMC9126699 DOI: 10.1007/s00109-022-02208-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 11/29/2022]
Abstract
Abstract Despite the development of novel pharmacological treatments, cardiovascular disease morbidity and mortality remain high indicating an unmet clinical need. Viral gene therapy enables targeted delivery of therapeutic transgenes and represents an attractive platform for tackling acquired and inherited cardiovascular diseases in the future. Current cardiovascular gene therapy trials in humans mainly focus on improving cardiac angiogenesis and function. Encouragingly, local delivery of therapeutic transgenes utilising first-generation human adenovirus serotype (HAd)-5 is safe in the short term and has shown some efficacy in drug refractory angina pectoris and heart failure with reduced ejection fraction. Despite this success, systemic delivery of therapeutic HAd-5 vectors targeting cardiovascular tissues and internal organs is limited by negligible gene transfer to target cells, elimination by the immune system, liver sequestration, off-target effects, and episomal degradation. To circumvent these barriers, cardiovascular gene therapy research has focused on determining the safety and efficacy of rare alternative serotypes and/or genetically engineered adenoviral capsid protein-modified vectors following local or systemic delivery. Pre-clinical studies have identified several vectors including HAd-11, HAd-35, and HAd-20–42-42 as promising platforms for local and systemic targeting of vascular endothelial and smooth muscle cells. In the past, clinical gene therapy trials were often restricted by limited scale-up capabilities of gene therapy medicinal products (GTMPs) and lack of regulatory guidance. However, significant improvement of industrial GTMP scale-up and purification, development of novel producer cell lines, and issuing of GTMP regulatory guidance by national regulatory health agencies have addressed many of these challenges, creating a more robust framework for future adenoviral-based cardiovascular gene therapy. In addition, this has enabled the mass roll out of adenovirus vector-based COVID-19 vaccines. Key messages First-generation HAd-5 vectors are widely used in cardiovascular gene therapy. HAd-5-based gene therapy was shown to lead to cardiac angiogenesis and improved function. Novel HAd vectors may represent promising transgene carriers for systemic delivery. Novel methods allow industrial scale-up of rare/genetically altered Ad serotypes. National regulatory health agencies have issued guidance on GMP for GTMPs.
Collapse
Affiliation(s)
- Schwartze Jt
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| | - Havenga M
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bakker Wam
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bradshaw Ac
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nicklin Sa
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
19
|
Patenaude B, Ballreich J. Estimating & comparing greenhouse gas emissions for existing intramuscular COVID-19 vaccines and a novel thermostable oral vaccine. THE JOURNAL OF CLIMATE CHANGE AND HEALTH 2022; 6:100127. [PMID: 35262040 PMCID: PMC8894686 DOI: 10.1016/j.joclim.2022.100127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/08/2022] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Climate impacts are rarely considered in health impact and economic assessments of public health programs. This study estimates the greenhouse gas (GHG) emissions averted by a novel oral SARS-CoV-2 (COVID-19) vaccine compared with four existing intramuscular vaccines: AstraZeneca's COVISHIELD®, Pfizer/BioNTech's COMIRNATY®, Moderna's mRNA-1273, and Johnson & Johnson's Ad26.COV2.S COVID-19 vaccine. METHODS We estimated GHG emissions averted for five vaccine modalities across nine countries. GHG emissions averted were derived from differences in cold chain logistics, production of vaccine supplies, and medical waste disposal. Countryspecific data including population coverage and electricity production mix were included in GHG emissions calculations. Results are presented in averted GHG per vaccine course and country level based on modeled vaccination demand. FINDINGS Per course, an oral vaccine is estimated to avert between 0.007 and 0.024 kgCO2e compared with Johnson & Johnson, 0.013 to 0.048 kgCO2e compared with AstraZeneca, 0.23 to 0.108 kgCO2e compared with Moderna, and 0.134 to 0.466 kgCO2e compared with Pfizer/BioNTech. The total GHG averted varied across countries based upon predicted demand, mix of electrical production, and vaccination strategy with the largest emissions reductions projected for India and the United States. INTERPRETATION Our results demonstrate large potential GHG emissions reductions from the use of oral vs. intramuscular vaccines for mass COVID-19 vaccination programs. Up to 82.25 million kgCO2e could be averted from utilization of an oral vaccine in the United States alone, which is equivalent to eliminating 17,700 automobiles from the road for one year. FUNDING Funding was provided by Vaxart, Inc. Vaxart, Inc. is currently developing an oral COVID-19 vaccine, the characteristics of which were utilized to define the thermostable oral vaccine discussed in this study. Apart from providing data on the characteristics of the oral vaccine under development, the funders had no influence over the study design, methods, statistical analyses, results, framing of results, decision to submit the manuscript for publication, or choice of journal.
Collapse
Affiliation(s)
- Bryan Patenaude
- Johns Hopkins Bloomberg School of Public Health, Department of International Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Jeromie Ballreich
- Johns Hopkins Bloomberg School of Public Health, Department of Health Policy and Management, 624 North Broadway, Baltimore, MD 21205, United States
| |
Collapse
|
20
|
Zhang L, Yao L, Guo Y, Li X, Ma L, Sun R, Han X, Liu J, Huang J. Oral SARS-CoV-2 Spike Protein Recombinant Yeast Candidate Prompts Specific Antibody and Gut Microbiota Reconstruction in Mice. Front Microbiol 2022; 13:792532. [PMID: 35464985 PMCID: PMC9022078 DOI: 10.3389/fmicb.2022.792532] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
A recent study showed that patients with coronavirus disease 2019 (COVID-19) have gastrointestinal symptoms and intestinal flora dysbiosis. Yeast probiotics shape the gut microbiome and improve immune homeostasis. In this study, an oral candidate of yeast-derived spike protein receptor-binding domain (RBD) and fusion peptide displayed on the surface of the yeast cell wall was generated. The toxicity and immune efficacy of oral administration were further performed in Institute of Cancer Research (ICR) mice. No significant difference in body weights, viscera index, and other side effects were detected in the oral-treated group. The detectable RBD-specific immunoglobulin G (IgG) and immunoglobulin A (IgA) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and more complex microbiota were detected from oral administration mice compared with those of the control group. Interestingly, the recombinant yeast was identified in female fetal of the high-dose group. These results revealed that the displaying yeast could fulfill the agent-driven immunoregulation and gut microbiome reconstitution. The findings will shed light on new dimensions against SARS-CoV-2 infection with the synergistic oral agents as promising non-invasive immunization and restoring gut flora.
Collapse
Affiliation(s)
- Lilin Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Lan Yao
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Yanyu Guo
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoyang Li
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Li Ma
- Tianjin Institute of Pharmaceutical Research Co., Ltd., Tianjin, China
| | - Ruiqi Sun
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Xueqing Han
- Chinese Academy of Inspection and Quarantine, Beijing, China
| | - Jing Liu
- Tianjin Institute of Pharmaceutical Research Co., Ltd., Tianjin, China
- Jing Liu,
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, China
- *Correspondence: Jinhai Huang,
| |
Collapse
|
21
|
Kar S, Devnath P, Emran TB, Tallei TE, Mitra S, Dhama K. Oral and intranasal vaccines against SARS-CoV-2: Current progress, prospects, advantages, and challenges. Immun Inflamm Dis 2022; 10:e604. [PMID: 35349752 PMCID: PMC8959423 DOI: 10.1002/iid3.604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a deadly pandemic in the 21st century, resulting in many deaths, economic loss, and international immobility. Vaccination represents the only mechanism to defeat this virus. Several intramuscular vaccines have been approved and are currently used worldwide. MAIN BODY However, global mass vaccination has not been achieved owing to several limitations, including the need for expertise to administer the injection-based vaccine, improper distribution of the vaccine, and lack of cold chain facilities, particularly in resource-poor, low-income countries. Mucosal vaccines are typically administered either orally or nasally, and several studies have shown promising results for developing these vaccines against SARS-CoV-2 that might serve as viable alternatives to current vaccines. SARS-CoV-2 invades the human body via oral and nasal mucosal surfaces; thus, an oral or nasal vaccine can trigger the immune system to inhibit the virus at the mucosal level, preventing further transmission via a strong mucosal and systematic immune response. Although several approaches toward developing a mucosal vaccine are currently being tested, additional attention is required. CONCLUSION In this article, the current approaches used to develop effective oral and nasal mucosal vaccines against SARS-CoV-2 and their benefits, prospects, and challenges have been summarized.
Collapse
Affiliation(s)
- Sanchita Kar
- Department of Infectious DiseaseInstitute of Developing Science and Health Initiatives, ECB ChattarDhakaBangladesh
- Department of MicrobiologyUniversity of ChittagongChittagongBangladesh
| | - Popy Devnath
- Department of MicrobiologyNoakhali Science and Technology UniversityNoakhaliBangladesh
| | - Talha B. Emran
- Department of PharmacyBGC Trust University BangladeshChittagongBangladesh
| | - Trina E. Tallei
- Department of Biology, Faculty of Mathematics and Natural SciencesSam Ratulangi UniversityManadoNorth SulawesiIndonesia
- Division of Sustainable Use of Wallacea AreaThe University Centre of Excellence for Biotechnology and Conservation of Wallacea, Institute for Research and Community Services, Sam Ratulangi UniversityManadoNorth SulawesiIndonesia
| | - Saikat Mitra
- Department of Pharmacy, Faculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Kuldeep Dhama
- Division of PathologyICAR‐Indian Veterinary Research Institute, IzatnagarBareillyUttar PradeshIndia
| |
Collapse
|
22
|
The Prevalence of COVID-19 Infection in Students and Staff at a Private University in Thailand by Rapid SARS-CoV-2 Antigen Detection Assay. JOURNAL OF ENVIRONMENTAL AND PUBLIC HEALTH 2022; 2022:2350522. [PMID: 35256886 PMCID: PMC8897746 DOI: 10.1155/2022/2350522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022]
Abstract
The COVID-19 pandemic led to the suspension of all university courses which was followed directly by the implementation of online learning in Thailand. However, online learning was not suitable for all of Thailand. Rangsit University is a famous private university in Thailand and has been affected by this crisis, so it attempted to eliminate online learning by offering vaccination and antigen rapid screening tests to the students and staff who had to attend the university from July to September 2021. 93.71% of the students and staff from Rangsit University who attended the university from July to September 2021 were vaccinated. Only 1.18% of the students and staff were infected. The vaccines used were CoronaVac and AstraZeneca at 66.02% and 33.98%, respectively. The percentage of individuals that were infected after vaccination did not differ between the two vaccines. The percentage of people infected was 0.31% for CoronaVac and 0.29% for AstraZeneca. Other important factors that influenced the infection rate were the initial symptoms and the environment. Individuals who had initial symptoms and had visited areas with high-risk factors had a high possibility of becoming infected. This research is intended to be useful for risk management during the COVID-19 crisis.
Collapse
|
23
|
Singh DD, Sharma A, Lee HJ, Yadav DK. SARS-CoV-2: Recent Variants and Clinical Efficacy of Antibody-Based Therapy. Front Cell Infect Microbiol 2022; 12:839170. [PMID: 35237535 PMCID: PMC8883582 DOI: 10.3389/fcimb.2022.839170] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
Multiple variants of SARS-CoV-2 have emerged and are now prevalent at the global level. Currently designated variants of concern (VOCs) are B.1.1.7, B1.351, P.1, B.1.617.2 variants and B.1.1.529. Possible options for VOC are urgently required as they carry mutations in the virus spike protein that allow them to spread more easily and cause more serious illness. The primary targets for most therapeutic methods against SARS-CoV-2 are the S (Spike) protein and RBD (Receptor-Binding Domain), which alter the binding to ACE2 (Angiotensin-Converting Enzyme 2). The most popular of these strategies involves the use of drug development targeting the RBD and the NTD (N-terminal domain) of the spike protein and multiple epitopes of the S protein. Various types of mutations have been observed in the RBDs of B.1.1.7, B1.351, P. and B.1.620. The incidence of RBD mutations increases the binding affinity to the ACE2 receptor. The high binding affinity of RBD and ACE2 has provided a structural basis for future evaluation of antibodies and drug development. Here we discuss the variants of SARS-CoV-2 and recent updates on the clinical evaluation of antibody-based treatment options. Presently, most of the antibody-based treatments have been effective in patients with SARS-CoV-2. However, there are still significant challenges in verifying independence, and the need for further clinical evaluation.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Anshul Sharma
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do, South Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do, South Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do, South Korea
- Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, South Korea
- *Correspondence: Hae-Jeung Lee, ; Dharmendra K. Yadav,
| | - Dharmendra K. Yadav
- Department of Pharmacy, Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Incheon, South Korea
- *Correspondence: Hae-Jeung Lee, ; Dharmendra K. Yadav,
| |
Collapse
|
24
|
Kudlay D, Svistunov A. COVID-19 Vaccines: An Overview of Different Platforms. Bioengineering (Basel) 2022; 9:bioengineering9020072. [PMID: 35200425 PMCID: PMC8869214 DOI: 10.3390/bioengineering9020072] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 12/29/2022] Open
Abstract
Vaccination is one of the key strategies to stop the COVID-19 pandemic. This review aims to evaluate the current state of vaccine development and to determine the issues that merit additional research. We conducted a literature review of the development of COVID-19 vaccines, their effectiveness, and their use in special patient groups. To date, 140 vaccines are in clinical development. Vector, RNA, subunit, and inactivated vaccines, as well as DNA vaccines, have been approved for human use. Vector vaccines have been well studied prior to the COVID-19 pandemic; however, their long-term efficacy and approaches to scaling up their production remain questionable. The main challenge for RNA vaccines is to improve their stability during production, storage, and transportation. For inactivated vaccines, the key issue is to improve their immunogenicity and effectiveness. To date, it has been shown that the immunogenicity of COVID-19 vaccines directly correlates with their clinical efficacy. In view of the constant mutation, the emerging new SARS-CoV-2 variants have been shown to be able to partially escape post-vaccination immune response; however, most vaccines remain sufficiently effective regardless of the variant of the virus. One of the promising strategies to improve the effectiveness of vaccination, which is being studied, is the use of different platforms within a single vaccination course. Despite significant progress in the development and study of COVID-19 vaccines, there are many issues that require further research.
Collapse
Affiliation(s)
- Dmitry Kudlay
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Building 2, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-(499)-248-05-53
| | - Andrey Svistunov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Building 2, 119991 Moscow, Russia;
| |
Collapse
|
25
|
Cytotoxic T-Cell-Based Vaccine against SARS-CoV-2: A Hybrid Immunoinformatic Approach. Vaccines (Basel) 2022; 10:vaccines10020218. [PMID: 35214676 PMCID: PMC8878688 DOI: 10.3390/vaccines10020218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
This paper presents an alternative vaccination platform that provides long-term cellular immune protection mediated by cytotoxic T-cells. The immune response via cellular immunity creates superior resistance to viral mutations, which are currently the greatest threat to the global vaccination campaign. Furthermore, we also propose a safer, more facile, and physiologically appropriate immunization method using either intranasal or oral administration. The underlying technology is an adaptation of synthetic long peptides (SLPs) previously used in cancer immunotherapy. The overall quality of the SLP constructs was validated using in silico methods. SLPs comprising HLA class I and class II epitopes were designed to stimulate antigen cross-presentation and canonical class II presentation by dendritic cells. The desired effect is a cytotoxic T cell-mediated prompt and specific immune response against the virus-infected epithelia and a rapid and robust virus clearance. Epitopes isolated from COVID-19 convalescent patients were screened for HLA class I and class II binding (NetMHCpan and NetMHCIIpan) and highest HLA population coverage (IEDB Population Coverage). 15 class I and 4 class II epitopes were identified and used for this SLP design. The constructs were characterized based on their toxicity (ToxinPred), allergenicity (AllerCatPro), immunogenicity (VaxiJen 2.0), and physico-chemical parameters (ProtParam). Based on in silico predictions, out of 60 possible SLPs, 36 candidate structures presented a high probability to be immunogenic, non-allergenic, non-toxic, and stable. 3D peptide folding followed by 3D structure validation (PROCHECK) and molecular docking studies (HADDOCK 2.4) with Toll-like receptors 2 and 4 provided positive results, suggestive for favorable antigen presentation and immune stimulation.
Collapse
|
26
|
Drozdzik A, Drozdzik M. Oral Pathology in COVID-19 and SARS-CoV-2 Infection-Molecular Aspects. Int J Mol Sci 2022; 23:1431. [PMID: 35163355 PMCID: PMC8836070 DOI: 10.3390/ijms23031431] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
This review article was designed to evaluate the existing evidence related to the molecular processes of SARS-CoV-2 infection in the oral cavity. The World Health Organization stated that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission is produced by respiratory droplets and aerosols from the oral cavity of infected patients. The oral cavity structures, keratinized and non-keratinized mucosa, and salivary glands' epithelia express SARS-CoV-2 entry and transmission factors, especially angiotensin converting enzyme Type 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). Replication of the virus in cells leads to local and systemic infection spread, and cellular damage is associated with clinical signs and symptoms of the disease in the oral cavity. Saliva, both the cellular and acellular fractions, holds the virus particles and contributes to COVID-19 transmission. The review also presents information about the factors modifying SARS-CoV-2 infection potential and possible local pharmacotherapeutic interventions, which may confine SARS-CoV-2 virus entry and transmission in the oral cavity. The PubMed and Scopus databases were used to search for suitable keywords such as: SARS-CoV-2, COVID-19, oral virus infection, saliva, crevicular fluid, salivary gland, tongue, oral mucosa, periodontium, gingiva, dental pulp, ACE2, TMPRSS2, Furin, diagnosis, topical treatment, vaccine and related words in relevant publications up to 28 December 2021. Data extraction and quality evaluation of the articles were performed by two reviewers, and 63 articles were included in the final review.
Collapse
Affiliation(s)
- Agnieszka Drozdzik
- Department of Integrated Dentistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp 72, 70-111 Szczecin, Poland;
| | - Marek Drozdzik
- Department of Pharmacology, Pomeranian Medical University in Szczecin, Powstancow Wlkp 72, 70-111 Szczecin, Poland
| |
Collapse
|
27
|
Focosi D, Maggi F, Casadevall A. Mucosal Vaccines, Sterilizing Immunity, and the Future of SARS-CoV-2 Virulence. Viruses 2022; 14:187. [PMID: 35215783 PMCID: PMC8878800 DOI: 10.3390/v14020187] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
Sterilizing immunity after vaccination is desirable to prevent the spread of infection from vaccinees, which can be especially dangerous in hospital settings while managing frail patients. Sterilizing immunity requires neutralizing antibodies at the site of infection, which for respiratory viruses such as SARS-CoV-2 implies the occurrence of neutralizing IgA in mucosal secretions. Systemic vaccination by intramuscular delivery induces no or low-titer neutralizing IgA against vaccine antigens. Mucosal priming or boosting, is needed to provide sterilizing immunity. On the other side of the coin, sterilizing immunity, by zeroing interhuman transmission, could confine SARS-CoV-2 in animal reservoirs, preventing spontaneous attenuation of virulence in humans as presumably happened with the endemic coronaviruses. We review here the pros and cons of each vaccination strategy, the current mucosal SARS-CoV-2 vaccines under development, and their implications for public health.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy;
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, MD 21218, USA;
| |
Collapse
|
28
|
Karczmarzyk K, Kęsik-Brodacka M. Attacking the Intruder at the Gate: Prospects of Mucosal Anti SARS-CoV-2 Vaccines. Pathogens 2022; 11:pathogens11020117. [PMID: 35215061 PMCID: PMC8876505 DOI: 10.3390/pathogens11020117] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/04/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
The sudden outbreak of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic in December 2019 caused crises and health emergencies worldwide. The rapid spread of the virus created an urgent need for the development of an effective vaccine and mass immunization to achieve herd immunity. Efforts of scientific teams at universities and pharmaceutical companies around the world allowed for the development of various types of preparations and made it possible to start the vaccination process. However, it appears that the developed vaccines are not effective enough and do not guarantee long-lasting immunity, especially for new variants of SARS-CoV-2. Considering this problem, it is promising to focus on developing a Coronavirus Disease 2019 (COVID-19) mucosal vaccine. Such a preparation applied directly to the mucous membranes of the upper respiratory tract might provide an immune barrier at the primary point of virus entry into the human body while inducing systemic immunity. A number of such preparations against SARS-CoV-2 are already in various phases of preclinical and clinical trials, and several of them are very close to being accepted for general use, constituting a milestone toward pandemic containment.
Collapse
Affiliation(s)
- Kacper Karczmarzyk
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
- Correspondence:
| | | |
Collapse
|
29
|
Hafiz I, Illian DN, Meila O, Utomo ARH, Susilowati A, Susetya IE, Desrita D, Siregar GA, Basyuni M. Effectiveness and Efficacy of Vaccine on Mutated SARS-CoV-2 Virus and Post Vaccination Surveillance: A Narrative Review. Vaccines (Basel) 2022; 10:82. [PMID: 35062743 PMCID: PMC8777947 DOI: 10.3390/vaccines10010082] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/19/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023] Open
Abstract
The ongoing COVID-19 pandemic, as a result of the SARS-CoV-2 virus, since December 2019, is a major health problem and concern worldwide. The pandemic has impacted various fields, from the social to the development of health science and technology. The virus has been mutating and thus producing several new variants, rushing research in the field of molecular biology to develop rapidly to overcome the problems that occur. Vaccine clinical studies are developing promptly with the aim of obtaining vaccines that are effective in suppressing the spread of the virus; however, the development of viral mutations raises concerns about the decreasing effectiveness of the resulting vaccine, which also results in the need for more in-depth studies. There have been 330 vaccines developed, including 136 clinical developments and 194 pre-clinical developments. The SARS-CoV-2 variant continues to evolve today, and it poses a challenge in testing the effectiveness of existing vaccines. This is a narrative review describing the emergence of the COVID-19 pandemic, development of vaccine platforms, identification of concerning mutations and virus variants in various countries of the world, and real-world monitoring of post-vaccination effectiveness and surveillance.
Collapse
Affiliation(s)
- Ihsanul Hafiz
- Department of Pharmacology, Faculty of Pharmacy and Health, Institut Kesehatan Helvetia, Medan 20124, Indonesia;
- Doctoral Program in Pharmacy, Department of Pharmaceutical Biology, School of Pharmacy, Institut Teknologi Bandung, Bandung 40132, Indonesia
| | - Didi Nurhadi Illian
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia; (D.N.I.); (O.M.)
| | - Okpri Meila
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia; (D.N.I.); (O.M.)
- Doctoral Program of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, Universitas Indonesia, Depok 16424, Indonesia
| | | | - Arida Susilowati
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Forestry, Faculty of Forestry, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Ipanna Enggar Susetya
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Aquatic Resource Management, Faculty of Agriculture, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Desrita Desrita
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Aquatic Resource Management, Faculty of Agriculture, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Gontar Alamsyah Siregar
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Mohammad Basyuni
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Forestry, Faculty of Forestry, Universitas Sumatera Utara, Medan 20155, Indonesia
| |
Collapse
|
30
|
Forchette L, Sebastian W, Liu T. A Comprehensive Review of COVID-19 Virology, Vaccines, Variants, and Therapeutics. Curr Med Sci 2021; 41:1037-1051. [PMID: 34241776 PMCID: PMC8267225 DOI: 10.1007/s11596-021-2395-1] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative pathogen of the coronavirus disease 2019 (COVID-19), has caused more than 179 million infections and 3.8 million deaths worldwide. Throughout the past year, multiple vaccines have already been developed and used, while some others are in the process of being developed. However, the emergence of new mutant strains of SARS-CoV-2 that have demonstrated immune-evading characteristics and an increase in infective capabilities leads to potential ineffectiveness of the vaccines against these variants. The purpose of this review article is to highlight the current understanding of the immunological mechanisms of the virus and vaccines, as well as to investigate some key variants and mutations of the virus driving the current pandemic and their impacts on current management guidelines. We also discussed new technologies being developed for the prevention, treatment, and detection of SARS-CoV-2. In this paper, we thoroughly reviewed and provided crucial information on SARS-CoV-2 virology, vaccines and drugs being used and developed for its prevention and treatment, as well as important variant strains. Our review paper will be beneficial to health care professionals and researchers so they can have a better understanding of the basic sciences, prevention, and clinical treatment of COVID-19 during the pandemic. This paper consists of the most updated information that has been available as of June 21, 2021.
Collapse
Affiliation(s)
- Lauren Forchette
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, 400 Lee Street North, Lewisburg, West Virginia 24901 USA
| | - William Sebastian
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, 400 Lee Street North, Lewisburg, West Virginia 24901 USA
| | - Tuoen Liu
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, 400 Lee Street North, Lewisburg, West Virginia 24901 USA
| |
Collapse
|
31
|
Kandimalla R, Chakraborty P, Vallamkondu J, Chaudhary A, Samanta S, Reddy PH, De Feo V, Dewanjee S. Counting on COVID-19 Vaccine: Insights into the Current Strategies, Progress and Future Challenges. Biomedicines 2021; 9:1740. [PMID: 34829969 PMCID: PMC8615473 DOI: 10.3390/biomedicines9111740] [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: 09/24/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/27/2022] Open
Abstract
The emergence of a novel coronavirus viz., severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019 and its subsequent substantial spread produced the coronavirus disease 2019 (COVID-19) pandemic worldwide. Given its unprecedented infectivity and pathogenicity, the COVID-19 pandemic had a devastating impact on human health, and its clinical management has been a great challenge, which has led to the development and speedy trials of several vaccine candidates against SARS-CoV-2 at an exceptional pace. As a result, several COVID-19 vaccines were made commercially available in the first half of 2021. Although several COVID-19 vaccines showed promising results, crucial insights into their epidemiology, protective mechanisms, and the propensities of reinfection are not largely reviewed. In the present report, we provided insights into the prospects of vaccination against COVID-19 and assessed diverse vaccination strategies including DNA, mRNA, protein subunits, vector-based, live attenuated, and inactivated whole/viral particle-based vaccines. Next, we reviewed major aspects of various available vaccines approved by the World Health Organization and by the local administrations to use against COVID-19. Moreover, we comprehensively assessed the success of these approved vaccines and also their untoward effects, including the possibility of reinfection. We also provided an update on the vaccines that are under development and could be promising candidates in the future. Conclusively, we provided insights into the COVID-19 vaccine epidemiology, their potency, and propensity for SARS-CoV-2 reinfection, while a careful review of their current status, strategies, success, and future challenges was also presented.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, Telangana, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | | | - Anupama Chaudhary
- Orinin-BioSystems, LE-52, Lotus Road 4, CHD City, Karnal 132001, Haryana, India;
| | - Sonalinandini Samanta
- Department of Dermatology (Skin & Venereology), ESIC Medical College & Hospital, Patna 801103, Bihar, India;
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
- Department of Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| |
Collapse
|
32
|
Side Effects of COVID-19 Pfizer-BioNTech mRNA Vaccine in Children Aged 12-18 Years in Saudi Arabia. Vaccines (Basel) 2021; 9:vaccines9111297. [PMID: 34835228 PMCID: PMC8621258 DOI: 10.3390/vaccines9111297] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/23/2021] [Accepted: 10/27/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Massive vaccination campaigns have been undertaken globally to combat the spread of the Coronavirus Disease 2019 (COVID-19). While most COVID-19 vaccines have shown excellent efficacy and safety profiles in clinical studies, real-world monitoring of vaccine safety is still important. In this study, we aimed to investigate the early side effects of Pfizer-BioNTech (BNT162b2) mRNA vaccine in children between 12–18 years old in Saudi Arabia. Method: To investigate the side effects in children in this age range following the administration of either one or two doses of Pfizer-BioNTech (BNT162b2) mRNA vaccine, we conducted a retrospective, cross-sectional study using a self-administered online survey. General and demographic data were collected, and vaccine-associated side effects following vaccination were evaluated. Results: The study recruited a total of 965 eligible participants. Overall, 571 (60%) of the study participants reported at least one side effect following Pfizer-BioNTech (BNT162b2) mRNA vaccination. The most frequently reported side effects were pain or redness at the site of injection (90%), fatigue (67%), fever (59%), headache (55%), nausea or vomiting (21%), and chest pain and shortness of breath (20%). Joint or bone pain were reported less frequently among our participants (2%). Our data showed that more female participants reported side effects compared to male participants, with 52% and 48%, respectively. Side effects were more common after the second dose compared to the first dose in our study cohort. Conclusions: While 60% of the children (12–18 years old) who received Pfizer-BioNTech (BNT162b2) mRNA vaccine reported side effects, our data showed that these side effects were not different from those that were reported in the clinical trials which lasted only for a few days. Side effects were more common after the second dose. Larger epidemiological and molecular studies are needed to evaluate the safety and the effectiveness of COVID-19 vaccine in protection of children against SARS-CoV-2 reinfections.
Collapse
|
33
|
Keikha R, Hashemi-Shahri SM, Jebali A. The evaluation of novel oral vaccines based on self-amplifying RNA lipid nanparticles (saRNA LNPs), saRNA transfected Lactobacillus plantarum LNPs, and saRNA transfected Lactobacillus plantarum to neutralize SARS-CoV-2 variants alpha and delta. Sci Rep 2021; 11:21308. [PMID: 34716391 PMCID: PMC8556360 DOI: 10.1038/s41598-021-00830-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/19/2021] [Indexed: 12/02/2022] Open
Abstract
The aim of this study was to present and evaluate novel oral vaccines, based on self-amplifying RNA lipid nanparticles (saRNA LNPs), saRNA transfected Lactobacillus plantarum LNPs, and saRNA transfected Lactobacillus plantarum, to neutralize severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) variants alpha and delta. After invitro evaluation of the oral vaccines on HEK293T/17 cells, we found that saRNA LNPs, saRNA transfected Lactobacillus plantarum LNPs, and saRNA transfected Lactobacillus plantarum could express S-protein at both mRNA and protein levels. In the next step, BALB/c mice were orally vaccinated with saRNA LNPs, saRNA transfected Lactobacillus plantarum LNPs, and saRNA transfected Lactobacillus plantarum at weeks 1 and 3. Importantly, a high titer of IgG and IgA was observed by all of them, sharply in week 6 (P < 0.05). In all study groups, their ratio of IgG2a/IgG1 was upper 1, indicating Th1-biased responses. Wild-type viral neutralization assay showed that the secreted antibodies in vaccinated mice and recovered COVID-19 patients could neutralize SARS-COV-2 variants alpha and delta. After oral administration of oral vaccines, biodistribution assay was done. It was found that all of them had the same biodistribution pattern. The highest concentration of S-protein was seen in the small intestine, followed by the large intestine and liver.
Collapse
MESH Headings
- Administration, Oral
- Adult
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- COVID-19/blood
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/pharmacokinetics
- Female
- HEK293 Cells
- Humans
- Immunoglobulin A/blood
- Immunoglobulin A/immunology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Intestine, Small/metabolism
- Lactobacillus plantarum/genetics
- Lactobacillus plantarum/metabolism
- Lipids/chemistry
- Male
- Mice
- Mice, Inbred BALB C
- Middle Aged
- Models, Animal
- Nanoparticles/chemistry
- Neutralization Tests
- RNA, Messenger/genetics
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Tissue Distribution
- Transfection/methods
- Vaccination/methods
- Vaccines, Synthetic/administration & dosage
- mRNA Vaccines
Collapse
Affiliation(s)
- Reza Keikha
- Infectious Diseases and Tropical Medicine Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Pathology, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Seyed Mohammad Hashemi-Shahri
- Infectious Diseases and Tropical Medicine Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ali Jebali
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
34
|
Marinović B, Miše J, Jukić IL, Bukvić Mokos Z. Pemphigus-The Crux of Clinics, Research, and Treatment during the COVID-19 Pandemic. Biomedicines 2021; 9:1555. [PMID: 34829784 PMCID: PMC8615103 DOI: 10.3390/biomedicines9111555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Pemphigus is a rare autoimmune disease characterised by the production of pathogenic autoantibodies in response to different desmosome proteins. The pathophysiological process leads to the development of blisters and erosions on mucosal and/or skin surfaces. The classical clinical variants of pemphigus are pemphigus vulgaris and pemphigus foliaceus. A diagnostic delay is very common in pemphigus, especially among patients with mucosal involvement. However, in recent years we have witnessed considerably fewer patients with extensive mucocutaneous manifestations, since patients with oral lesions are referred to dermatologists to start the treatment much sooner than they had been previously. Among non-classical variants of pemphigus, unusual cases with discrepancies between autoantibody profiles and clinics challenge the "desmoglein compensation theory". The identification of several other autoantigens that perform a role in the pathogenesis of different variants of pemphigus will progress immunodermatology towards an approach that will determine personalized pemphigus subtypes for each patient. Comorbidities among patients are primarily associated with the prolonged use of corticosteroids and other immunosuppressive agents. The SARS-CoV-2 pandemic raised concerns regarding the immunosuppressive effects of treatment and the risk of a more complicated COVID-19 infection, as well as on the ability to develop an adequate vaccine response.
Collapse
Affiliation(s)
- Branka Marinović
- Department of Dermatology and Venereology, University Hospital Centre Zagreb, School of Medicine University of Zagreb, Šalata 4, 10000 Zagreb, Croatia; (B.M.); (I.L.J.)
| | - Joško Miše
- Department of Dermatology and Venereology, University Hospital Centre Zagreb, European Reference Network (ERN)-Skin Reference Centre, Kišpatićeva 12, 10000 Zagreb, Croatia;
| | - Ines Lakoš Jukić
- Department of Dermatology and Venereology, University Hospital Centre Zagreb, School of Medicine University of Zagreb, Šalata 4, 10000 Zagreb, Croatia; (B.M.); (I.L.J.)
| | - Zrinka Bukvić Mokos
- Department of Dermatology and Venereology, University Hospital Centre Zagreb, School of Medicine University of Zagreb, Šalata 4, 10000 Zagreb, Croatia; (B.M.); (I.L.J.)
| |
Collapse
|
35
|
Wang S, Li L, Yan F, Gao Y, Yang S, Xia X. COVID-19 Animal Models and Vaccines: Current Landscape and Future Prospects. Vaccines (Basel) 2021; 9:1082. [PMID: 34696190 PMCID: PMC8537799 DOI: 10.3390/vaccines9101082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022] Open
Abstract
The worldwide pandemic of coronavirus disease 2019 (COVID-19) has become an unprecedented challenge to global public health. With the intensification of the COVID-19 epidemic, the development of vaccines and therapeutic drugs against the etiological agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also widespread. To prove the effectiveness and safety of these preventive vaccines and therapeutic drugs, available animal models that faithfully recapitulate clinical hallmarks of COVID-19 are urgently needed. Currently, animal models including mice, golden hamsters, ferrets, nonhuman primates, and other susceptible animals have been involved in the study of COVID-19. Moreover, 117 vaccine candidates have entered clinical trials after the primary evaluation in animal models, of which inactivated vaccines, subunit vaccines, virus-vectored vaccines, and messenger ribonucleic acid (mRNA) vaccines are promising vaccine candidates. In this review, we summarize the landscape of animal models for COVID-19 vaccine evaluation and advanced vaccines with an efficacy range from about 50% to more than 95%. In addition, we point out future directions for COVID-19 animal models and vaccine development, aiming at providing valuable information and accelerating the breakthroughs confronting SARS-CoV-2.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Ling Li
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao 266000, China;
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| |
Collapse
|
36
|
Jeeva S, Kim KH, Shin CH, Wang BZ, Kang SM. An Update on mRNA-Based Viral Vaccines. Vaccines (Basel) 2021; 9:965. [PMID: 34579202 PMCID: PMC8473183 DOI: 10.3390/vaccines9090965] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022] Open
Abstract
With the success of COVID-19 vaccines, newly created mRNA vaccines against other infectious diseases are beginning to emerge. Here, we review the structural elements required for designing mRNA vaccine constructs for effective in vitro synthetic transcription reactions. The unprecedently speedy development of mRNA vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was enabled with previous innovations in nucleoside modifications during in vitro transcription and lipid nanoparticle delivery materials of mRNA. Recent updates are briefly described in the status of mRNA vaccines against SARS-CoV-2, influenza virus, and other viral pathogens. Unique features of mRNA vaccine platforms and future perspectives are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (S.J.); (K.-H.K.); (C.H.S.); (B.-Z.W.)
| |
Collapse
|
37
|
Could Probiotics and Postbiotics Function as "Silver Bullet" in the Post-COVID-19 Era? Probiotics Antimicrob Proteins 2021; 13:1499-1507. [PMID: 34386940 PMCID: PMC8360758 DOI: 10.1007/s12602-021-09833-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 12/14/2022]
Abstract
We are currently experiencing the realities of the most severe pandemic within living memory, with major impacts on the health and economic well-being of our planet. The scientific community has demonstrated an unprecedented mobilization capability, with the rapid development of vaccines and drugs targeting the protection of human life and palliative measures for infected individuals. However, are we adequately prepared for ongoing defense against COVID-19 and its variants in the post-pandemic world? Moreover, are we equipped to provide a satisfactory quality of life for individuals who are recovering from COVID-19 disease? What are the possibilities for the acceleration of the recovery process? Here, we give special consideration to the potential and already-demonstrated role of probiotics and traditional medical approaches to the management of current and potential future encounters with our major virus adversaries.
Collapse
|
38
|
Jicsinszky L, Martina K, Cravotto G. Cyclodextrins in the antiviral therapy. J Drug Deliv Sci Technol 2021; 64:102589. [PMID: 34035845 PMCID: PMC8135197 DOI: 10.1016/j.jddst.2021.102589] [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: 03/01/2021] [Revised: 04/30/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023]
Abstract
The main antiviral drug-cyclodextrin interactions, changes in physicochemical and physiological properties of the most commonly used virucides are summarized. The potential complexation of antiviral molecules against the SARS-Cov2 also pointed out the lack of detailed information in designing effective and general medicines against viral infections. The principal problem of the current molecules is the 3D structures of the currently active compounds. Improving the solubility or bioavailability of antiviral molecules is possible, however, there is no universal solution, and the complexation experiments dominantly use the already approved cyclodextrin derivatives. This review discusses the basic properties of the different cyclodextrin derivatives, their potential in antiviral formulations, and the prevention and treatment of viral infections. The biologically active new cyclodextrin derivatives are also discussed.
Collapse
Affiliation(s)
- László Jicsinszky
- Dept. of Drug Science and Technology, University of Turin, Via Giuria 9, 10125, Torino, Italy
| | - Katia Martina
- Dept. of Drug Science and Technology, University of Turin, Via Giuria 9, 10125, Torino, Italy
| | - Giancarlo Cravotto
- Dept. of Drug Science and Technology, University of Turin, Via Giuria 9, 10125, Torino, Italy
| |
Collapse
|
39
|
Shin CH, Kim KH, Jeeva S, Kang SM. Towards Goals to Refine Prophylactic and Therapeutic Strategies Against COVID-19 Linked to Aging and Metabolic Syndrome. Cells 2021; 10:1412. [PMID: 34204163 PMCID: PMC8227274 DOI: 10.3390/cells10061412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) gave rise to the coronavirus disease 2019 (COVID-19) pandemic. A strong correlation has been demonstrated between worse COVID-19 outcomes, aging, and metabolic syndrome (MetS), which is primarily derived from obesity-induced systemic chronic low-grade inflammation with numerous complications, including type 2 diabetes mellitus (T2DM). The majority of COVID-19 deaths occurs in people over the age of 65. Individuals with MetS are inclined to manifest adverse disease consequences and mortality from COVID-19. In this review, we examine the prevalence and molecular mechanisms underlying enhanced risk of COVID-19 in elderly people and individuals with MetS. Subsequently, we discuss current progresses in treating COVID-19, including the development of new COVID-19 vaccines and antivirals, towards goals to elaborate prophylactic and therapeutic treatment options in this vulnerable population.
Collapse
Affiliation(s)
- Chong-Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (S.J.)
| | | | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (S.J.)
| |
Collapse
|
40
|
Abu-Hammad O, Alduraidi H, Abu-Hammad S, Alnazzawi A, Babkair H, Abu-Hammad A, Nourwali I, Qasem F, Dar-Odeh N. Side Effects Reported by Jordanian Healthcare Workers Who Received COVID-19 Vaccines. Vaccines (Basel) 2021; 9:vaccines9060577. [PMID: 34205917 PMCID: PMC8228820 DOI: 10.3390/vaccines9060577] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/17/2021] [Accepted: 05/22/2021] [Indexed: 12/04/2022] Open
Abstract
Background Distribution of COVID-19 vaccines has been surrounded by suspicions and rumors making it necessary to provide the public with accurate reports from trustworthy experts such as healthcare professionals. Methods We distributed a questionnaire in Jordan among physicians, dentists and nurses who received a COVID-19 vaccine to explore the side effects (SE) they encountered after the first or the second dose of one of three vaccines namely: AstraZeneca Vaxzevria (AZ), Pfizer-BioNTeck (PB), and SinoPharm (SP) vaccines. Results A total of 409 professionals participated. Approximately 18% and 31% of participants reported no SE after the first dose and second dose, respectively. The remainder had mostly local side effects related to injection site (74%). Systemic side effects in the form of fatigue (52%), myalgia (44%), headache (42%), and fever (35%) prevailed mainly after the first dose. These were significantly associated with AZ vaccine, and age ≤ 45 years (p = 0.000 and 0.01, respectively). No serious SE were reported. Conclusions We can conclude that SE of COVID-19 vaccines distributed in Jordan are within the common range known so far for these vaccines. Further studies are needed to include larger sample size and longer follow-up period to monitor possible serious and long-term SE of the vaccines.
Collapse
Affiliation(s)
- Osama Abu-Hammad
- College of Dentistry, Taibah University, Al Madinah Al Munawara 43353, Saudi Arabia; (O.A.-H.); (A.A.); (H.B.); (I.N.)
- School of Dentistry, University of Jordan, Amman 11942, Jordan
| | - Hamza Alduraidi
- School of Nursing, University of Jordan, Amman 11942, Jordan;
| | | | - Ahmed Alnazzawi
- College of Dentistry, Taibah University, Al Madinah Al Munawara 43353, Saudi Arabia; (O.A.-H.); (A.A.); (H.B.); (I.N.)
| | - Hamzah Babkair
- College of Dentistry, Taibah University, Al Madinah Al Munawara 43353, Saudi Arabia; (O.A.-H.); (A.A.); (H.B.); (I.N.)
| | | | - Ibrahim Nourwali
- College of Dentistry, Taibah University, Al Madinah Al Munawara 43353, Saudi Arabia; (O.A.-H.); (A.A.); (H.B.); (I.N.)
| | - Farah Qasem
- University of Jordan Hospital, Amman 11942, Jordan;
| | - Najla Dar-Odeh
- College of Dentistry, Taibah University, Al Madinah Al Munawara 43353, Saudi Arabia; (O.A.-H.); (A.A.); (H.B.); (I.N.)
- School of Dentistry, University of Jordan, Amman 11942, Jordan
- Correspondence:
| |
Collapse
|
41
|
Sampath V, Rabinowitz G, Shah M, Jain S, Diamant Z, Jesenak M, Rabin R, Vieths S, Agache I, Akdis M, Barber D, Breiteneder H, Chinthrajah S, Chivato T, Collins W, Eiwegger T, Fast K, Fokkens W, O'Hehir RE, Ollert M, O'Mahony L, Palomares O, Pfaar O, Riggioni C, Shamji MH, Sokolowska M, Jose Torres M, Traidl-Hoffmann C, van Zelm M, Wang DY, Zhang L, Akdis CA, Nadeau KC. Vaccines and allergic reactions: The past, the current COVID-19 pandemic, and future perspectives. Allergy 2021; 76:1640-1660. [PMID: 33811364 PMCID: PMC8251022 DOI: 10.1111/all.14840] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 12/11/2022]
Abstract
Vaccines are essential public health tools with a favorable safety profile and prophylactic effectiveness that have historically played significant roles in reducing infectious disease burden in populations, when the majority of individuals are vaccinated. The COVID-19 vaccines are expected to have similar positive impacts on health across the globe. While serious allergic reactions to vaccines are rare, their underlying mechanisms and implications for clinical management should be considered to provide individuals with the safest care possible. In this review, we provide an overview of different types of allergic adverse reactions that can potentially occur after vaccination and individual vaccine components capable of causing the allergic adverse reactions. We present the incidence of allergic adverse reactions during clinical studies and through post-authorization and post-marketing surveillance and provide plausible causes of these reactions based on potential allergenic components present in several common vaccines. Additionally, we review implications for individual diagnosis and management and vaccine manufacturing overall. Finally, we suggest areas for future research.
Collapse
Affiliation(s)
- Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Grace Rabinowitz
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Mihir Shah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Surabhi Jain
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Zuzana Diamant
- Departmentt of Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Leuven, Belgium
- Department of Respiratory Medicine & Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden
- Department of Clinical Pharmacy &Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Milos Jesenak
- Department of Pediatrics and Department of Clinical Immunology and Allergology, Jessenius Faculty of Medicine in Martin, Center for Vaccination in Special Situations, University Hospital in Martin, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ronald Rabin
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Stefan Vieths
- Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | | | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Zurich, Switzerland
| | - Domingo Barber
- Departamento de CienciasMédicasBásicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, España
- Instituto de Salud Carlos III, RETIC ARADYAL, Madrid, Spain
| | - Heimo Breiteneder
- Division of Medical Biotechnology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Tomas Chivato
- School of Medicine, University CEU San Pablo, Madrid, Spain
| | - William Collins
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Hospital Medicine, Stanford University, Stanford, CA, USA
| | - Thomas Eiwegger
- Division of Immunology and Allergy, Food Allergy and Anaphylaxis Program, The Hospital for Sick Children, Toronto, ON, Canada
- Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Katharine Fast
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Wytske Fokkens
- Department of Otorhinolaryngology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Robyn E O'Hehir
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University, and Alfred Health, Melbourne, Vic, Australia
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Carmen Riggioni
- Department of Paediatrics, Allergy and Clinical Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Mohamed H Shamji
- Department of National Heart and Lung Institute, Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Imperial College London, London, UK
- Centre in Allergic Mechanisms of Asthma, London, UK
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Zurich, Switzerland
| | - Maria Jose Torres
- Allergy Unit, Malaga Regional University Hospital-UMA-ARADyAL, Málaga, Spain
| | - Claudia Traidl-Hoffmann
- Department of Environmental Medicine, Medical Faculty, University Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany
| | - Menno van Zelm
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, VIC, Australia
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Zurich, Switzerland
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
42
|
Beeraka NM, Tulimilli SV, Karnik M, Sadhu SP, Pragada RR, Aliev G, Madhunapantula SV. The Current Status and Challenges in the Development of Vaccines and Drugs against Severe Acute Respiratory Syndrome-Corona Virus-2 (SARS-CoV-2). BIOMED RESEARCH INTERNATIONAL 2021; 2021:8160860. [PMID: 34159203 PMCID: PMC8168478 DOI: 10.1155/2021/8160860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 04/16/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection causes coronavirus disease-19 (COVID-19), which is characterized by clinical manifestations such as pneumonia, lymphopenia, severe acute respiratory distress, and cytokine storm. S glycoprotein of SARS-CoV-2 binds to angiotensin-converting enzyme II (ACE-II) to enter into the lungs through membrane proteases consequently inflicting the extensive viral load through rapid replication mechanisms. Despite several research efforts, challenges in COVID-19 management still persist at various levels that include (a) availability of a low cost and rapid self-screening test, (b) lack of an effective vaccine which works against multiple variants of SARS-CoV-2, and (c) lack of a potent drug that can reduce the complications of COVID-19. The development of vaccines against SARS-CoV-2 is a complicated process due to the emergence of mutant variants with greater virulence and their ability to invoke intricate lung pathophysiology. Moreover, the lack of a thorough understanding about the virus transmission mechanisms and complete pathogenesis of SARS-CoV-2 is making it hard for medical scientists to develop a better strategy to prevent the spread of the virus and design a clinically viable vaccine to protect individuals from being infected. A recent report has tested the hypothesis of T cell immunity and found effective when compared to the antibody response in agammaglobulinemic patients. Understanding SARS-CoV-2-induced changes such as "Th-2 immunopathological variations, mononuclear cell & eosinophil infiltration of the lung and antibody-dependent enhancement (ADE)" in COVID-19 patients provides key insights to develop potential therapeutic interventions for immediate clinical management. Therefore, in this review, we have described the details of rapid detection methods of SARS-CoV-2 using molecular and serological tests and addressed different therapeutic modalities used for the treatment of COVID-19 patients. In addition, the current challenges against the development of vaccines for SARS-CoV-2 are also briefly described in this article.
Collapse
Affiliation(s)
- Narasimha M. Beeraka
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow 119991, Russia
| | - SubbaRao V. Tulimilli
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
| | - Medha Karnik
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
| | - Surya P. Sadhu
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, 530003 Andhra Pradesh, India
| | - Rajeswara Rao Pragada
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, 530003 Andhra Pradesh, India
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow 119991, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region 142432, Russia
- Research Institute of Human Morphology, 3Tsyurupy Street, Moscow 117418, Russia
- GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
| |
Collapse
|
43
|
Kim KH, Bhatnagar N, Jeeva S, Oh J, Park BR, Shin CH, Wang BZ, Kang SM. Immunogenicity and Neutralizing Activity Comparison of SARS-CoV-2 Spike Full-Length and Subunit Domain Proteins in Young Adult and Old-Aged Mice. Vaccines (Basel) 2021; 9:316. [PMID: 33805473 PMCID: PMC8066235 DOI: 10.3390/vaccines9040316] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/30/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to be expanding the pandemic disease across the globe. Although SARS-CoV-2 vaccines were rapidly developed and approved for emergency use of vaccination in humans, supply and production difficulties are slowing down the global vaccination program. The efficacy of many different versions of vaccine candidates and adjuvant effects remain unknown, particularly in the elderly. In this study, we compared the immunogenic properties of SARS-CoV-2 full-length spike (S) ectodomain in young adult and aged mice, S1 with receptor binding domain, and S2 with fusion domain. Full-length S was more immunogenic and effective in inducing IgG antibodies after low dose vaccination, compared to the S1 subunit. Old-aged mice induced SARS-CoV-2 spike-specific IgG antibodies with neutralizing activity after high dose S vaccination. With an increased vaccine dose, S1 was highly effective in inducing neutralizing and receptor-binding inhibiting antibodies, although both S1 and S2 subunit domain vaccines were similarly immunogenic. Adjuvant effects were significant for effective induction of IgG1 and IgG2a isotypes, neutralizing and receptor-binding inhibiting antibodies, and antibody-secreting B cell and interferon-γ secreting T cell immune responses. Results of this study provide information in designing SARS-CoV-2 spike vaccine antigens and effective vaccination in the elderly.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (K.-H.K.); (N.B.); (S.J.); (J.O.); (B.R.P.); (C.H.S.); (B.-Z.W.)
| |
Collapse
|