1
|
Ito T, Suzuki T, Sakai Y, Nishioka K, Itoh Y, Sakamoto K, Ikemura N, Matoba S, Kanda Y, Takagi J, Okamoto T, Tahara K, Hoshino A. Engineered ACE2 decoy in dry powder form for inhalation: A novel therapy for SARS-CoV-2 variants. Mol Ther Methods Clin Dev 2025; 33:101459. [PMID: 40276779 PMCID: PMC12019485 DOI: 10.1016/j.omtm.2025.101459] [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: 11/12/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
The persistent threat of SARS-CoV-2 and the emergence of new variants has prompted the development of a novel, easily administered modality that can overcome viral mutations. The engineered ACE2 decoy shows neutralizing activity comparable to monoclonal antibodies and is broadly effective against SARS-CoV-2 variants and ACE2-utilizing sarbecoviruses. In addition to intravenous administration, this decoy has shown antiviral efficacy through nebulized aerosol inhalation in murine and primate models, offering a dose-sparing advantage. Clinically, dry powder formulation is ideal for convenience and storage but poses challenges for protein biologics. This study developed a freeze-dried spray formulation of the ACE2 decoy for inhalation. The trehalose and leucine-based excipient maintained neutralizing activity and prevented aggregate formation. The dry powder showed aerodynamic distribution from bronchi to alveoli, aiding protection against SARS-CoV-2 infections. Neutralizing activity, structural stability, and powder dispersibility were preserved after 6 months of storage. In a mouse model of SARS-CoV-2 infection, significant reductions in viral replication and lung pathology were observed with intratracheal administration 24 h post-infection. The ACE2 decoy retained activity against recent JN.1 and current KP.3 strains, confirming its robust efficacy against viral mutations. This ACE2 decoy powder inhalant is a self-administered, next-generation treatment addressing the ongoing immune-evading evolution of SARS-CoV-2.
Collapse
Affiliation(s)
- Takaaki Ito
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Keisuke Nishioka
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kentarou Sakamoto
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Nariko Ikemura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kohei Tahara
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
- Laboratory of Nanofiber Technology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
2
|
Liu Y, Deng S, Ren S, Tam RCY, Liu S, Zhang AJ, To KKW, Yuen KY, Chen H, Wang P. Intranasal influenza virus-vectored vaccine offers protection against clade 2.3.4.4b H5N1 infection in small animal models. Nat Commun 2025; 16:3133. [PMID: 40169649 PMCID: PMC11962148 DOI: 10.1038/s41467-025-58504-z] [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: 08/22/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
The highly pathogenic avian influenza (HPAI) H5N1 virus has been endemic in aquatic birds since 1997, causing outbreaks in domestic poultry and occasional human infections worldwide. Recently, the cross-species transmission of a new reassortant variant from clade 2.3.4.4b of H5N1 to cattle in the US has heightened concerns regarding the expansion of host range and potential human infection. As eradicating the H5N1 virus from its reservoir is impossible, it is essential to prepare for a potential pandemic caused by an H5N1 derivative. Utilizing a deleted-NS1 live attenuated influenza viral vector vaccine system (DelNS1 LAIV), a system we have previously used in the development of a COVID-19 vaccine, we have rapidly developed an intranasal vaccine for cattle H5N1 and related clade 2.3.4.4b strains, based on publicly available sequences. Our research demonstrates that a single intranasal immunization can provide effective protection against lethal challenges from HPAI cattle or mink H5N1 variants, offering strong, sustained immunity after two months in female mouse and male hamster models. Immunogenicity analysis reveals that intranasal vaccination with DelNS1 LAIV induces robust neutralizing antibody, mucosal IgA and T cell responses in mice. It is crucial to further evaluate the DelNS1-H5N1 LAIV system to prepare for potential future H5N1 outbreaks in humans.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shaofeng Deng
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shuang Ren
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Rachel Chun-Yee Tam
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Siwen Liu
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anna Jinxia Zhang
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Pandemic Research Alliance Unit, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Pandemic Research Alliance Unit, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Honglin Chen
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Pui Wang
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| |
Collapse
|
3
|
Ngoufack Jagni Semengue E, Takou D, Potesta M, Ndjeyep Djupsa SC, Montesano C, Chenwi CA, Beloumou G, Nka AD, Kengni Ngueko AM, Molimbou E, Etame NK, Gouissi Anguechia DH, Mundo Nayang AR, Tueguem PP, Ndomgue T, Tambe Ayuk Ngwese D, Moko Fotso LG, Tommo Tchouaket CM, Ka'e AC, Fainguem N, Abega Abega CA, Mandeng N, Epee E, Esso L, Etoundi Mballa G, Santoro MM, Z-K Bissek AC, Otokoye Otshudiema J, Alteri C, Boum Ii Y, Marcelin AG, Ceccherini-Silberstein F, Ndjolo A, Perno CF, Kaseya J, Colizzi V, Ndembi N, Fokam J. Disparities in anti-SARS-CoV-2 reactivity according to vaccines administered in the era of omicron in Cameroon: Lessons for future outbreak response. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004312. [PMID: 40106487 PMCID: PMC11922206 DOI: 10.1371/journal.pgph.0004312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
With the advent of COVID-19, anti-SARS-CoV-2 vaccines were a global health priority, but evidence on their significance within tropical settings remained limited. We sought to assess the distribution of anti-SARS-CoV-2 antibodies according to vaccine status and types of vaccines administered in Cameroon during Omicron waves. A community based cross-sectional sero-survey was conducted from February-15 through July-31 2022 among individuals tested for COVID-19 in Yaoundé-Cameroon. Sociodemographic data were collected from participants. Anti-SARS-CoV-2 antibodies (both IgG and IgM) were tested on plasma and statistical analyses were performed wherever appropriate. Logistic regression was done with p<0.05 considered statistically significant. Overall, 2449 participants were enrolled: median-age was 40 [31-49], 56.4% (1382/2449) men, 2.2% (54/2449) with flu-like symptoms and 19.6% (481/2449) reporting previous SARS-CoV-2 positivity. Regarding COVID-19 vaccination, 67.5% (1652/2449) had received at least one dose, 55.0% (909/1652) two-dose series and 37.1% (613/1652) received additional booster doses. Median duration from vaccination to phlebotomy was 5 [4-9] months. Seroprevalence of anti-SARS-CoV-2 antibodies was 81.1% (1987/2449). Following logistic regression, vaccine status (aOR=1.95), booster doses (aOR=1.36), post-vaccination time (≤5 months; aOR=1.64), Pfizer (aOR=2.07) and Moderna (aOR=1.52) vaccines, were all associated with a high prevalence of anti-SARS-CoV-2 antibodies (all p<0.05). This high seroprevalence of anti-SARS-CoV-2 antibodies suggests a certain degree of immunity/protection at community-level in Cameroon during Omicron waves, with Pfizer and Moderna inducing greater immunogenicity. However, rapid antibody waning (~5 months) calls for vaccine updates with novel variants (arising from a rapidly evolving virus) that could compromise already acquired immunity.
Collapse
Affiliation(s)
| | - Desire Takou
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Marina Potesta
- Department of Biology, Faculty of Sciences, University of Rome "Tor Vergata", Rome, Italy
| | | | - Carla Montesano
- Department of Biology, Faculty of Sciences, University of Rome "Tor Vergata", Rome, Italy
- Faculty of Science and Technology, Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Collins Ambes Chenwi
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Grace Beloumou
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Alex Durand Nka
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Aurelie Minelle Kengni Ngueko
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Evariste Molimbou
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Science and Technology, Evangelic University of Cameroon, Bandjoun, Cameroon
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Naomi-Karell Etame
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Davy-Hyacinthe Gouissi Anguechia
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Audrey Rachel Mundo Nayang
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Pamela Patricia Tueguem
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Therese Ndomgue
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Department of Biology, Faculty of Sciences, University of Rome "Tor Vergata", Rome, Italy
| | - Derrick Tambe Ayuk Ngwese
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Larissa Gaëlle Moko Fotso
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Carlos Michel Tommo Tchouaket
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- School of Health Sciences, Catholic University of Central Africa, Yaoundé, Cameroon
| | - Aude Christelle Ka'e
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Nadine Fainguem
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Cyrille Alain Abega Abega
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | - Nadia Mandeng
- Faculty of Health Sciences, University of Bamenda, Bamenda, Cameroon
- National Public Health Emergency Operations Coordination Centre, Ministry of Public Health, Yaoundé, Cameroon
- Department of Disease, Epidemic and Pandemic Control, Ministry of Public Health, Yaounde, Cameroon
| | - Emilienne Epee
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National Public Health Emergency Operations Coordination Centre, Ministry of Public Health, Yaoundé, Cameroon
- Department of Disease, Epidemic and Pandemic Control, Ministry of Public Health, Yaounde, Cameroon
| | - Linda Esso
- National Public Health Emergency Operations Coordination Centre, Ministry of Public Health, Yaoundé, Cameroon
- Department of Disease, Epidemic and Pandemic Control, Ministry of Public Health, Yaounde, Cameroon
| | - Georges Etoundi Mballa
- National Public Health Emergency Operations Coordination Centre, Ministry of Public Health, Yaoundé, Cameroon
- Department of Disease, Epidemic and Pandemic Control, Ministry of Public Health, Yaounde, Cameroon
| | - Maria Mercedes Santoro
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | | | - John Otokoye Otshudiema
- COVID-19 Incident Management Team, World Health Organization, Country Office - Yaoundé, Yaoundé, Cameroon
| | - Claudia Alteri
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Yap Boum Ii
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National Public Health Emergency Operations Coordination Centre, Ministry of Public Health, Yaoundé, Cameroon
- Department of Disease, Epidemic and Pandemic Control, Ministry of Public Health, Yaounde, Cameroon
| | | | | | - Alexis Ndjolo
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Carlo-Federico Perno
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Bambino Gesu' Children's Research Hospital, Rome, Italy
| | - Jean Kaseya
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Vittorio Colizzi
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- Faculty of Science and Technology, Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Nicaise Ndembi
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
- Institute of Human Virology, University of Maryland School of Medicine, College Park, Maryland, United States of America
| | - Joseph Fokam
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
- National Public Health Emergency Operations Coordination Centre, Ministry of Public Health, Yaoundé, Cameroon
- Faculty of Health Sciences, University of Buea, Buea, Cameroon
| |
Collapse
|
4
|
Awakoaiye B, Dangi T, Penaloza-MacMaster P. Effects of protein boosters on antibody responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.637239. [PMID: 39990358 PMCID: PMC11844364 DOI: 10.1101/2025.02.08.637239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
SARS-CoV-2 has infected a large fraction of the human population. Currently, most individuals have developed immunity either through vaccination or natural infection. Despite this, SARS-CoV-2 booster immunizations are still recommended to reduce the risk of reinfections, but there is still limited understanding on how different booster vaccine platforms influence antibody responses. We conducted immunological studies in mice to evaluate the boosting effects of different vaccine platforms on antibody responses. C57BL/6 mice were first primed with an adenovirus serotype 5 (Ad5) vaccine expressing the SARS-CoV-2 spike protein. The mice were then boosted with the same Ad5-based vaccine (homologous boosting) or with a protein-based vaccine (heterologous boosting). Interestingly, the heterologous regimen (Ad5 prime + protein boost) elicited superior antibody responses, relative to the homologous regimen (Ad5 prime + Ad5 boost). Similar potentiation of antibody responses was reported when mice were primed with poxvirus or rhabdovirus vectors and then boosted with protein. These findings highlight a potential advantage of protein booster immunizations to potentiate humoral immunity.
Collapse
Affiliation(s)
| | - Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Wang M, Liu C, Fan Q, Sun Y, Tang S, Guo H, Zhou B, Wang H, Ge X, Zhang Z, Ju B. Rapid clonal expansion and somatic hypermutation contribute to the fate of SARS-CoV-2 broadly neutralizing antibodies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:278-289. [PMID: 40073246 DOI: 10.1093/jimmun/vkae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 12/09/2024] [Indexed: 03/14/2025]
Abstract
Several vaccines and immunization strategies, including inactivated vaccines, have proven effective in eliciting antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), providing an opportunity to characterize the antibody response. In this study, we investigated the monoclonal antibody responses elicited by wild-type SARS-CoV-2 inactivated vaccination compared to those elicited by natural infection and mRNA vaccination. The analysis showed that antibodies encoded by biased germline genes were shared between SARS-CoV-2 vaccinated and naturally infected individuals. Among the 35 shared clonotypes identified, besides the well-known IGHV3-53 and IGHV1-58, we identified a class of IGHV4-59 antibodies characterized by rapid response and neutralizing activity, elicited by 3 doses of inactivated vaccine. Members of this lineage exhibited similar sensitivity against wild-type SARS-CoV-2, whereas different neutralizing activities against SARS-CoV-2 variants, especially against various Omicron subvariants, BA.1, BA.2, BA.2.12.1, BA.4/5, and BA.2.75. Structural analysis of BA.1 spike complexed with VacBB-639 revealed that the IGHV4-59-lineage antibodies belonged to the Class 2/3 group. Using sequence alignment, site-mutation assays, and functional verification, we identified two substitutions, N60K in HFR3 and S56G in HCDR2, contributing to opposite neutralization changes of IGHV4-59-lineage antibodies against these Omicron subvariants. These results demonstrate the importance of somatic hypermutation in the evolution of prototypical antigen-elicited antibodies in terms of their neutralization breadth and potency against SARS-CoV-2 Omicron variants.
Collapse
Affiliation(s)
- Miao Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Congcong Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Qing Fan
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Yuehong Sun
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Shilong Tang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Huimin Guo
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Bing Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Haiyan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xiangyang Ge
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong Province, China
- Shenzhen Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Shenzhen, Guangdong Province, China
| | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong Province, China
| |
Collapse
|
6
|
Ovchinnikov V, Karplus M. Phenomenological Modeling of Antibody Response from Vaccine Strain Composition. Antibodies (Basel) 2025; 14:6. [PMID: 39846614 PMCID: PMC11755667 DOI: 10.3390/antib14010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
The elicitation of broadly neutralizing antibodies (bnAbs) is a major goal of vaccine design for highly mutable pathogens, such as influenza, HIV, and coronavirus. Although many rational vaccine design strategies for eliciting bnAbs have been devised, their efficacies need to be evaluated in preclinical animal models and in clinical trials. To improve outcomes for such vaccines, it would be useful to develop methods that can predict vaccine efficacies against arbitrary pathogen variants. As a step in this direction, here, we describe a simple biologically motivated model of antibody reactivity elicited by nanoparticle-based vaccines using only antigen amino acid sequences, parametrized with a small sample of experimental antibody binding data from influenza or SARS-CoV-2 nanoparticle vaccinations. Results: The model is able to recapitulate the experimental data to within experimental uncertainty, is relatively insensitive to the choice of the parametrization/training set, and provides qualitative predictions about the antigenic epitopes exploited by the vaccine, which are testable by experiment. For the mosaic nanoparticle vaccines considered here, model results suggest indirectly that the sera obtained from vaccinated mice contain bnAbs, rather than simply different strain-specific Abs. Although the present model was motivated by nanoparticle vaccines, we also apply it to a mutlivalent mRNA flu vaccination study, and demonstrate good recapitulation of experimental results. This suggests that the model formalism is, in principle, sufficiently flexible to accommodate different vaccination strategies. Finally, we show how the model could be used to rank the efficacies of vaccines with different antigen compositions. Conclusions: Overall, this study suggests that simple models of vaccine efficacy parametrized with modest amounts of experimental data could be used to compare the effectiveness of designed vaccines.
Collapse
Affiliation(s)
- Victor Ovchinnikov
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Martin Karplus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Laboratoire de Chimie Biophysique, ISIS, Université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
7
|
Li Y, Feng Y, Li W, Zhang Y, Sun Y, Wang S, Curtiss R, Shi H. A Purely Biomanufactured System for Delivering Nanoparticles and STING Agonists. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408539. [PMID: 39588587 PMCID: PMC11744655 DOI: 10.1002/advs.202408539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/24/2024] [Indexed: 11/27/2024]
Abstract
Nanovaccines have significantly contributed in the prevention and treatment of diseases. However, most of these technologies rely on chemical or hybrid semibiological synthesis methods, which limit the manufacturing performance advantages and improved inoculation outcomes compared with traditional vaccines. Herein, a universal and purely biological nanovaccine system is reported. This system integrates three modules: (1) self-assembling nanoparticles, (2) self-catalyzed synthesis of small-molecule stimulator of interferon gene (STING) agonists, and (3) delivery vectors that target the cytosolic surveillance system. Various nanoparticles are efficiently self-assembled using this system. After confirming the excellent immunostimulatory and lymph node targeting of this system, its broad-spectrum antiviral efficacy is further demonstrated. By leveraging the comprehensive biosynthetic capabilities of bacterial cells, this system can efficiently combine various adjuvant-active modular components and antigenic cargo, thereby providing a highly diversified and potent vaccine platform.
Collapse
Affiliation(s)
- Yu‐an Li
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouChina
| | - Yi Feng
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouChina
| | - Wenjing Li
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouChina
| | - Yuqin Zhang
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouChina
| | - Yanni Sun
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouChina
| | - Shifeng Wang
- Department of Infectious Diseases and ImmunologyCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL32611‐0880USA
| | - Roy Curtiss
- Department of Infectious Diseases and ImmunologyCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL32611‐0880USA
| | - Huoying Shi
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouChina
- Joint International Research Laboratory of Agriculture and Agri‐Product SafetyYangzhou University (JIRLAAPS)YangzhouChina
| |
Collapse
|
8
|
Athavale A, Gaur A, Ahmed N, Subramaniam A, Dandotiya J, Raj S, Upadhyay SK, Samal S, Pandey AK, Rai RC, Awasthi A. Receptor Binding Domain-Specific B Cell Memory Responses Among Individuals Vaccinated Against SARS-CoV-2. Vaccines (Basel) 2024; 12:1396. [PMID: 39772064 PMCID: PMC11680197 DOI: 10.3390/vaccines12121396] [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: 09/19/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The COVID-19 pandemic prompted unprecedented vaccine development efforts against SARS-CoV-2. India, which was one of the countries most impacted by COVID-19, developed its indigenous vaccine in addition to utilizing the ones developed by other countries. While antibody levels and neutralizing antibody titres are considered initial correlates of immune protection, long-term protection from the pathogen relies on memory B and T cells and their recall responses. In this regard, global research has primarily focused on mRNA-based vaccines. The studies on immune memory response, particularly B cell memory response induced by the vaccines given to Indians, remain relatively obscure. Methods: We assessed Receptor Binding Domain-specific memory B cells in the peripheral circulation and their ability to secrete antigen-specific antibodies among Indians vaccinated with Covaxin (BBV152), Covishield (AZD1222), Corbevax (BECOV2D), and Sputnik Light, as well as unvaccinated individuals. Results: Corbevax and Sputnik Light conferred better antibody-secreting cell (ASC) responses over time compared to other groups. Conclusions: These findings contribute to our understanding of vaccine-induced immune memory in the Indian population; providing insights that could inform future vaccine strategies.
Collapse
Affiliation(s)
- Atharv Athavale
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Anmol Gaur
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Nafees Ahmed
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Adarsh Subramaniam
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Jyotsna Dandotiya
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Sneha Raj
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | | | - Sweety Samal
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | | | - Ramesh Chandra Rai
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Amit Awasthi
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| |
Collapse
|
9
|
Yang CH, Shen KY, Ho HM, Huang CY, Cheng YJ, Pu CC, Chiu FF, Huang WC, Liao HC, Chen HW, Liao CL, Liu SJ, Huang MH. Boosting DNA vaccine power by lipid nanoparticles surface engineered with amphiphilic bioresorbable copolymer. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102261. [PMID: 39071950 PMCID: PMC11278320 DOI: 10.1016/j.omtn.2024.102261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/14/2024] [Indexed: 07/30/2024]
Abstract
Successful DNA vaccination generally requires the aid of either a viral vector within vaccine components or an electroporation device into the muscle or skin of the host. However, these systems come with certain obstacles, including limited transgene capacity, broad preexisting immunity in humans, and substantial cell death caused by high voltage pulses, respectively. In this study, we repurposed the use of an amphiphilic bioresorbable copolymer (ABC), called PLA-PEG, as a surface engineering agent that conciliates lipid nanoparticles (LNPs) between stability during preparation and biocompatibility post-vaccination. The LNP carrier can be loaded with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike-specific DNA; in this form, the DNA-LNP is immunogenic in hamsters and elicits protective immunity following DNA-LNP vaccination against heterologous virus challenge or as a hybrid-type vaccine booster against SARS-CoV-2 variants. The data provide comprehensive information on the relationships between LNP composition, manufacturing process, and vaccine efficacy. The outcomes of this study offer new insights into designing next-generation LNP formulations and pave the way for boosting vaccine power to combat existing and possible emerging infectious diseases/pathogens.
Collapse
Affiliation(s)
- Chung-Hsiang Yang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Kuan-Yin Shen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hui-Min Ho
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chiung-Yi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yu-Jhen Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chih-Chun Pu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Fang-Feng Chiu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Wan-Chun Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Hsi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
10
|
Gupta M, Bogatyreva K, Pienaar K, Vally H, Bennett CM. The timing of local SARS-Cov-2 outbreaks and vaccination coverage during the Delta wave in Melbourne. Aust N Z J Public Health 2024; 48:100164. [PMID: 38945056 DOI: 10.1016/j.anzjph.2024.100164] [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: 08/17/2023] [Revised: 05/01/2024] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
OBJECTIVE This article presents a longitudinal analysis of COVID-19 infection and vaccination coverage in Melbourne metropolitan local government areas (LGAs) during the 2021 Delta wave. METHODS COVID-19 vaccination and infection data from 12 July to 27 November 2021 were sourced from government websites. Summary statistics and associated 95% confidence intervals (95% CI) were compared by LGA ranked according to socioeconomic status: total "burden" (total infections per thousand), "peak" (highest weekly infection rate), "lag" (interval between peak and 70% double vaccination). RESULTS LGAs in the bottom five deciles for social advantage experienced higher infection rates (39.0 per thousand [95% CI: 38.5, 39.5] vs. 14.8 [14.7, 14.9]), and had lower two-dose vaccination coverage (23.8% [23.6, 23.9] vs. 32.7% [32.6, 32.7]) compared with LGAs in the top five deciles. LGAs that achieved 70% coverage two weeks or more after the infection peak experienced nearly twice the total infection burden (27.7 per 1000 [27.3, 28.0] compared with 14.9 [14.7, 15.0]) than LGAs with a shorter lag. CONCLUSIONS Exposure and transmission risk factors cluster within disadvantaged LGAs. The potential for large local outbreaks is heightened if vaccination uptake trails in these communities. IMPLICATIONS FOR PUBLIC HEALTH In a pandemic, decision-makers must prioritise disease control and harm reduction interventions for at-risk LGAs.
Collapse
Affiliation(s)
- Mehr Gupta
- Institute for Health Transformation, Deakin University, Waurn Ponds, Australia
| | - Kat Bogatyreva
- Institute for Health Transformation, Deakin University, Waurn Ponds, Australia
| | - Kiran Pienaar
- Department of Sociology, School of Humanities and Social Sciences, Deakin University, Burwood, Victoria, Australia
| | - Hassan Vally
- Institute for Health Transformation, Deakin University, Waurn Ponds, Australia; Centre for Innovation in Infectious Disease and Immunology Research, Deakin University, Waurn Ponds, Australia
| | - Catherine M Bennett
- Institute for Health Transformation, Deakin University, Waurn Ponds, Australia; Centre for Innovation in Infectious Disease and Immunology Research, Deakin University, Waurn Ponds, Australia.
| |
Collapse
|
11
|
Maurice RL. Post-COVID-19: Time to Change Our Way of Life for a Better Future. EPIDEMIOLOGIA 2024; 5:211-220. [PMID: 38804342 PMCID: PMC11130812 DOI: 10.3390/epidemiologia5020015] [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: 04/09/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Background and Objectives: From the year 1 anno Domini until 1855, with the third plague, major pandemics occurred on average every 348 years. Since then, they have occurred on average every 33 years, with coronavirus disease 2019 (COVID-19) now underway. Even though current technologies have greatly improved the way of life of human beings, COVID-19, with more than 700,000,000 cases and 6,950,000 deaths worldwide by the end of 2023, reminds us that much remains to be done. This report looks back at 18 months of COVID-19, from March 2020 to August 2021, with the aim of highlighting potential solutions that could help mitigate the impact of future pandemics. Materials and Methods: COVID-19 data, including case and death reports, were extracted daily from the Worldometer platform to build a database for the macroscopic analysis of the spread of the virus around the world. Demographic data were integrated into the COVID-19 database for a better understanding of the spatial spread of the SARS-CoV-2 virus in cities/municipalities. Without loss of generality, only data from the top 30 (out of 200 and above) countries ranked by total number of COVID-19 cases were analyzed. Statistics (regression, t-test (p < 0.05), correlation, mean ± std, etc.) were carried out with Excel software (Microsoft® Excel® 2013 (15.0.5579.1001)). Spectral analysis, using Matlab software (license number: 227725), was also used to try to better understand the temporal spread of COVID-19. Results: This study showed that COVID-19 mainly affects G20 countries and that cities/municipalities with high population density are a powerful activator of the spread of the virus. In addition, spectral analysis highlighted that the very first months of the spread of COVID-19 were the most notable, with a strong expansion of the SARS-CoV-2 virus. On the other hand, the following six months showed a certain level of stability, mainly due to multiple preventive measures such as confinement, the closure of non-essential services, the wearing of masks, distancing of 2 m, etc. Conclusion: Given that densely populated cities and municipal areas have largely favored the spread of the SARS-CoV-2 virus, it is believed that such a demographic context is becoming a societal problem that developed countries must address in a manner that is adequate and urgent. COVID-19 has made us understand that it is time to act both preventatively and curatively. With phenomenological evidence suggesting that the next pandemic could occur in less than 50 years, it may be time to launch new societal projects aimed at relieving congestion in densely populated regions.
Collapse
Affiliation(s)
- Roch Listz Maurice
- Groupe Biomédical Montérégie, Centre Intégré de Santé et des Services Sociaux de la Montérégie-Centre (CISSSMC), Brossard, QC J4W 3J8, Canada
| |
Collapse
|
12
|
Fu W, Guo M, Zhou X, Wang Z, Sun J, An Y, Guan T, Hu M, Li J, Chen Z, Ye J, Gao X, Gao GF, Dai L, Wang Y, Chen C. Injectable Hydrogel Mucosal Vaccine Elicits Protective Immunity against Respiratory Viruses. ACS NANO 2024; 18:11200-11216. [PMID: 38620102 DOI: 10.1021/acsnano.4c00155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Intranasal vaccines, eliciting mucosal immune responses, can prevent early invasion, replication, and transmission of pathogens in the respiratory tract. However, the effective delivery of antigens through the nasal barrier and boosting of a robust systematic and mucosal immune remain challenges in intranasal vaccine development. Here, we describe an intranasally administered self-healing hydrogel vaccine with a reversible strain-dependent sol-gel transition by precisely modulating the self-assembly processes between the natural drug rhein and aluminum ions. The highly bioadhesive hydrogel vaccine enhances antigen stability and prolongs residence time in the nasal cavity and lungs by confining the antigen to the surface of the nasal mucosa, acting as a "mucosal mask". The hydrogel also stimulates superior immunoenhancing properties, including antigen internalization, cross-presentation, and dendritic cell maturation. Furthermore, the formulation recruits immunocytes to the nasal mucosa and nasal-associated lymphoid tissue (NALT) while enhancing antigen-specific humoral, cellular, and mucosal immune responses. Our findings present a promising strategy for preparing intranasal vaccines for infectious diseases or cancer.
Collapse
Affiliation(s)
- Wenjiao Fu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Xuemei Zhou
- School of Life Sciences, Hebei University, Baoding 071002, People's Republic of China
| | - Zhenzhen Wang
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jiufeng Sun
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, People's Republic of China
| | - Yaling An
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Tong Guan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Mingdi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jiayang Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Ziwei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jinmin Ye
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Xingfa Gao
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Yaling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100021, People's Republic of China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100021, People's Republic of China
| |
Collapse
|
13
|
Hannula L, Kuivanen S, Lasham J, Kant R, Kareinen L, Bogacheva M, Strandin T, Sironen T, Hepojoki J, Sharma V, Saviranta P, Kipar A, Vapalahti O, Huiskonen JT, Rissanen I. Nanobody engineering for SARS-CoV-2 neutralization and detection. Microbiol Spectr 2024; 12:e0419922. [PMID: 38363137 PMCID: PMC10986514 DOI: 10.1128/spectrum.04199-22] [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: 10/17/2022] [Accepted: 01/03/2024] [Indexed: 02/17/2024] Open
Abstract
In response to the ongoing COVID-19 pandemic, the quest for coronavirus inhibitors has inspired research on a variety of small proteins beyond conventional antibodies, including robust single-domain antibody fragments, i.e., "nanobodies." Here, we explore the potential of nanobody engineering in the development of antivirals and diagnostic tools. Through fusion of nanobody domains that target distinct binding sites, we engineered multimodular nanobody constructs that neutralize wild-type SARS-CoV-2 and the Alpha and Delta variants at high potency, with IC50 values as low as 50 pM. Despite simultaneous binding to distinct epitopes, Beta and Omicron variants were more resistant to neutralization by the multimodular nanobodies, which highlights the importance of accounting for antigenic drift in the design of biologics. To further explore the applications of nanobody engineering in outbreak management, we present an assay based on fusions of nanobodies with fragments of NanoLuc luciferase that can detect sub-nanomolar quantities of the SARS-CoV-2 spike protein in a single step. Our work showcases the potential of nanobody engineering to combat emerging infectious diseases. IMPORTANCE Nanobodies, small protein binders derived from the camelid antibody, are highly potent inhibitors of respiratory viruses that offer several advantages over conventional antibodies as candidates for specific therapies, including high stability and low production costs. In this work, we leverage the unique properties of nanobodies and apply them as building blocks for new therapeutic and diagnostic tools. We report ultra-potent SARS-CoV-2 inhibition by engineered nanobodies comprising multiple modules in structure-guided combinations and develop nanobodies that carry signal molecules, allowing rapid detection of the SARS-CoV-2 spike protein. Our results highlight the potential of engineered nanobodies in the development of effective countermeasures, both therapeutic and diagnostic, to manage outbreaks of emerging viruses.
Collapse
Affiliation(s)
- Liina Hannula
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Suvi Kuivanen
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jonathan Lasham
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Ravi Kant
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Tropical Parasitology, Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| | - Lauri Kareinen
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Mariia Bogacheva
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Tomas Strandin
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tarja Sironen
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Vivek Sharma
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Petri Saviranta
- VTT Technical Research Centre of Finland Ltd., Espoo, Finland
| | - Anja Kipar
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Olli Vapalahti
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Juha T. Huiskonen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Ilona Rissanen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
14
|
Estapé Senti M, García Del Valle L, Schiffelers RM. mRNA delivery systems for cancer immunotherapy: Lipid nanoparticles and beyond. Adv Drug Deliv Rev 2024; 206:115190. [PMID: 38307296 DOI: 10.1016/j.addr.2024.115190] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
mRNA-based vaccines are emerging as a promising alternative to standard cancer treatments and the conventional vaccines. Moreover, the FDA-approval of three nucleic acid based therapeutics (Onpattro, BNT162b2 and mRNA-1273) has further increased the interest and trust on this type of therapeutics. In order to achieve a significant therapeutic efficacy, the mRNA needs from a drug delivery system. In the last years, several delivery platforms have been explored, being the lipid nanoparticles (LNPs) the most well characterized and studied. A better understanding on how mRNA-based therapeutics operate (both the mRNA itself and the drug delivery system) will help to further improve their efficacy and safety. In this review, we will provide an overview of what mRNA cancer vaccines are and their mode of action and we will highlight the advantages and challenges of the different delivery platforms that are under investigation.
Collapse
Affiliation(s)
- Mariona Estapé Senti
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Lucía García Del Valle
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Raymond M Schiffelers
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
15
|
Lee IJ, Lan YH, Wu PY, Wu YW, Chen YH, Tseng SC, Kuo TJ, Sun CP, Jan JT, Ma HH, Liao CC, Liang JJ, Ko HY, Chang CS, Liu WC, Ko YA, Chen YH, Sie ZL, Tsung SI, Lin YL, Wang IH, Tao MH. A receptor-binding domain-based nanoparticle vaccine elicits durable neutralizing antibody responses against SARS-CoV-2 and variants of concern. Emerg Microbes Infect 2023; 12:2149353. [PMID: 36395071 PMCID: PMC9793938 DOI: 10.1080/22221751.2022.2149353] [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] [Indexed: 11/18/2022]
Abstract
Numerous vaccines have been developed to address the current COVID-19 pandemic, but safety, cross-neutralizing efficacy, and long-term protectivity of currently approved vaccines are still important issues. In this study, we developed a subunit vaccine, ASD254, by using a nanoparticle vaccine platform to encapsulate the SARS-CoV-2 spike receptor-binding domain (RBD) protein. As compared with the aluminum-adjuvant RBD vaccine, ASD254 induced higher titers of RBD-specific antibodies and generated 10- to 30-fold more neutralizing antibodies. Mice vaccinated with ASD254 showed protective immune responses against SARS-CoV-2 challenge, with undetectable infectious viral loads and reduced typical lesions in lung. Besides, neutralizing antibodies in vaccinated mice lasted for at least one year and were effective against various SARS-CoV-2 variants of concern, including B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma), B.1.617.2 (Delta), and B.1.1.529 (Omicron). Furthermore, particle size, polydispersity index, and zeta-potential of ASD254 remained stable after 8-month storage at 4°C. Thus, ASD254 is a promising nanoparticle vaccine with good immunogenicity and stability to be developed as an effective vaccine option in controlling upcoming waves of COVID-19.
Collapse
Affiliation(s)
- I-Jung Lee
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Hua Lan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ping-Yi Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yan-Wei Wu
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hung Chen
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Che Tseng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Tzu-Jiun Kuo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Cheng-Pu Sun
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsiu-Hua Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chun-Che Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Ying Ko
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Shin Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Wen-Chun Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-An Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Zong-Lin Sie
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Szu-I Tsung
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan,Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - I-Hsuan Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Mi-Hua Tao
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan,Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan, Mi-Hua Tao Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei115, Taiwan
| |
Collapse
|
16
|
Ovchinnikov V, Karplus M. Free Energy Simulations of Receptor-Binding Domain Opening of the SARS-CoV-2 Spike Indicate a Barrierless Transition with Slow Conformational Motions. J Phys Chem B 2023; 127:8565-8575. [PMID: 37756691 PMCID: PMC10578350 DOI: 10.1021/acs.jpcb.3c05236] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/12/2023] [Indexed: 09/29/2023]
Abstract
Infection by sarbecoviruses begins with the attachment of the homotrimeric viral "spike" protein to the angiotensin-converting enzyme 2 receptor on the surface of mammalian cells. This requires one or more receptor-binding domains (RBDs) to be in the open (up) position. Here, we present the results of long molecular dynamics simulations with umbrella sampling (US) to compute a one-dimensional free energy profile of RBD opening/closing and the associated transition times. After ≃3.58μs of simulation time per US window (∼229 μs in total), which was required to approach trajectory decorrelation, the computed free energy profile was found to be without large barriers. This suggests that the RBD diffuses between the open and closed positions without significant energetic hindrance. This interpretation appears consistent with experiments but is at odds with some previous simulations. Modeling the RBD motion as diffusive dynamics along the computed free energy profile, we find that the overall time required for the transition is only about 2 μs, which is 5 orders of magnitude shorter than experimentally measured transition times. We speculate that the most likely reason for the transition time mismatch is our use of very short glycans, which was required to make the simulations performed here feasible. Despite the long simulation times, the final free energy profile is not fully converged with statistical errors of ≃1.16 kcal/mol, which were found to be consistent with the slow time decay in the autocorrelation of the conformational motions of the protein. The simulation lengths that would be required to obtain fully converged results remain unknown, but the present calculations would benefit from at least an order-of-magnitude extension.
Collapse
Affiliation(s)
- Victor Ovchinnikov
- Department
of Chemistry and Chemical Biology, Harvard
University, Cambridge, Massachusetts 02138, United States
| | - Martin Karplus
- Department
of Chemistry and Chemical Biology, Harvard
University, Cambridge, Massachusetts 02138, United States
- Laboratoire
de Chimie Biophysique, ISIS, Université
de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
17
|
Oubahmane M, Hdoufane I, Delaite C, Sayede A, Cherqaoui D, El Allali A. Design of Potent Inhibitors Targeting the Main Protease of SARS-CoV-2 Using QSAR Modeling, Molecular Docking, and Molecular Dynamics Simulations. Pharmaceuticals (Basel) 2023; 16:ph16040608. [PMID: 37111365 PMCID: PMC10146715 DOI: 10.3390/ph16040608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/08/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a serious global public health threat. The evolving strains of SARS-CoV-2 have reduced the effectiveness of vaccines. Therefore, antiviral drugs against SARS-CoV-2 are urgently needed. The main protease (Mpro) of SARS-CoV-2 is an extremely potent target due to its pivotal role in virus replication and low susceptibility to mutation. In the present study, a quantitative structure-activity relationship (QSAR) study was performed to design new molecules that might have higher inhibitory activity against SARS-CoV-2 Mpro. In this context, a set of 55 dihydrophenanthrene derivatives was used to build two 2D-QSAR models using the Monte Carlo optimization method and the Genetic Algorithm Multi-Linear Regression (GA-MLR) method. From the CORAL QSAR model outputs, the promoters responsible for the increase/decrease in inhibitory activity were extracted and interpreted. The promoters responsible for an increase in activity were added to the lead compound to design new molecules. The GA-MLR QSAR model was used to ensure the inhibitory activity of the designed molecules. For further validation, the designed molecules were subjected to molecular docking analysis and molecular dynamics simulations along with an absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis. The results of this study suggest that the newly designed molecules have the potential to be developed as effective drugs against SARS-CoV-2.
Collapse
Affiliation(s)
- Mehdi Oubahmane
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, BP 2390, Marrakech 40000, Morocco
| | - Ismail Hdoufane
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, BP 2390, Marrakech 40000, Morocco
| | - Christelle Delaite
- Laboratoire de Photochimie et d'Ingénierie Macromoléculaires (LPIM), Ecole Nationale Supérieure de Chimie de Mulhouse, Université de Haute-Alsace, 68100 Mulhouse, France
| | - Adlane Sayede
- University Artois, CNRS, Centrale Lille, University Lille, UMR 8181, Unité de Catalyse et Chimie du Solide (UCCS), F-62300 Lens, France
| | - Driss Cherqaoui
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, BP 2390, Marrakech 40000, Morocco
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir 43150, Morocco
| |
Collapse
|
18
|
Deng S, Liu Y, Tam RCY, Chen P, Zhang AJ, Mok BWY, Long T, Kukic A, Zhou R, Xu H, Song W, Chan JFW, To KKW, Chen Z, Yuen KY, Wang P, Chen H. An intranasal influenza virus-vectored vaccine prevents SARS-CoV-2 replication in respiratory tissues of mice and hamsters. Nat Commun 2023; 14:2081. [PMID: 37045873 PMCID: PMC10092940 DOI: 10.1038/s41467-023-37697-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
Current available vaccines for COVID-19 are effective in reducing severe diseases and deaths caused by SARS-CoV-2 infection but less optimal in preventing infection. Next-generation vaccines which are able to induce mucosal immunity in the upper respiratory to prevent or reduce infections caused by highly transmissible variants of SARS-CoV-2 are urgently needed. We have developed an intranasal vaccine candidate based on a live attenuated influenza virus (LAIV) with a deleted NS1 gene that encodes cell surface expression of the receptor-binding-domain (RBD) of the SARS-CoV-2 spike protein, designated DelNS1-RBD4N-DAF. Immune responses and protection against virus challenge following intranasal administration of DelNS1-RBD4N-DAF vaccines were analyzed in mice and compared with intramuscular injection of the BioNTech BNT162b2 mRNA vaccine in hamsters. DelNS1-RBD4N-DAF LAIVs induced high levels of neutralizing antibodies against various SARS-CoV-2 variants in mice and hamsters and stimulated robust T cell responses in mice. Notably, vaccination with DelNS1-RBD4N-DAF LAIVs, but not BNT162b2 mRNA, prevented replication of SARS-CoV-2 variants, including Delta and Omicron BA.2, in the respiratory tissues of animals. The DelNS1-RBD4N-DAF LAIV system warrants further evaluation in humans for the control of SARS-CoV-2 transmission and, more significantly, for creating dual function vaccines against both influenza and COVID-19 for use in annual vaccination strategies.
Collapse
Affiliation(s)
- Shaofeng Deng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Ying Liu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Rachel Chun-Yee Tam
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Pin Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Anna Jinxia Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Bobo Wing-Yee Mok
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Teng Long
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Anja Kukic
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Runhong Zhou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Haoran Xu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Wenjun Song
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kelvin Kai-Wang To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Zhiwei Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Pui Wang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
| | - Honglin Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
| |
Collapse
|
19
|
Moore KA, Leighton T, Ostrowsky JT, Anderson CJ, Danila RN, Ulrich AK, Lackritz EM, Mehr AJ, Baric RS, Baylor NW, Gellin BG, Gordon JL, Krammer F, Perlman S, Rees HV, Saville M, Weller CL, Osterholm MT. A research and development (R&D) roadmap for broadly protective coronavirus vaccines: A pandemic preparedness strategy. Vaccine 2023; 41:2101-2112. [PMID: 36870874 PMCID: PMC9941884 DOI: 10.1016/j.vaccine.2023.02.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Broadly protective coronavirus vaccines are an important tool for protecting against future SARS-CoV-2 variants and could play a critical role in mitigating the impact of future outbreaks or pandemics caused by novel coronaviruses. The Coronavirus Vaccines Research and Development (R&D) Roadmap (CVR) is aimed at promoting the development of such vaccines. The CVR, funded by the Bill & Melinda Gates Foundation and The Rockefeller Foundation, was generated through a collaborative and iterative process, which was led by the Center for Infectious Disease Research and Policy (CIDRAP) at the University of Minnesota and involved 50 international subject matter experts and recognized leaders in the field. This report summarizes the major issues and areas of research outlined in the CVR and identifies high-priority milestones. The CVR covers a 6-year timeframe and is organized into five topic areas: virology, immunology, vaccinology, animal and human infection models, and policy and finance. Included in each topic area are key barriers, gaps, strategic goals, milestones, and additional R&D priorities. The roadmap includes 20 goals and 86 R&D milestones, 26 of which are ranked as high priority. By identifying key issues, and milestones for addressing them, the CVR provides a framework to guide funding and research campaigns that promote the development of broadly protective coronavirus vaccines.
Collapse
Affiliation(s)
- Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA; Center for Infectious Disease Research and Policy, C315 Mayo Memorial Building, MMC 263, 420 Delaware Street, SE, Minneapolis, Minnesota 55455, USA.
| | - Tabitha Leighton
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cory J Anderson
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Angela K Ulrich
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eve M Lackritz
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela J Mehr
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ralph S Baric
- University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | - Jennifer L Gordon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Florian Krammer
- Department of Microbiology, Department of Pathology, Molecular and Cell-Based Medicine, and Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | | | - Helen V Rees
- Wits RHI, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Melanie Saville
- Coalition for Epidemic Preparedness Innovations, London, United Kingdom
| | | | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
20
|
Pacheco-García U, Serafín-López J. Indirect Dispersion of SARS-CoV-2 Live-Attenuated Vaccine and Its Contribution to Herd Immunity. Vaccines (Basel) 2023; 11:655. [PMID: 36992239 PMCID: PMC10055900 DOI: 10.3390/vaccines11030655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
It has been 34 months since the beginning of the SARS-CoV-2 coronavirus pandemic, which causes the COVID-19 disease. In several countries, immunization has reached a proportion near what is required to reach herd immunity. Nevertheless, infections and re-infections have been observed even in vaccinated persons. That is because protection conferred by vaccines is not entirely effective against new virus variants. It is unknown how often booster vaccines will be necessary to maintain a good level of protective immunity. Furthermore, many individuals refuse vaccination, and in developing countries, a large proportion of the population has not yet been vaccinated. Some live-attenuated vaccines against SARS-CoV-2 are being developed. Here, we analyze the indirect dispersion of a live-attenuated virus from vaccinated individuals to their contacts and the contribution that this phenomenon could have to reaching Herd Immunity.
Collapse
Affiliation(s)
- Ursino Pacheco-García
- Department of Cardio-Renal Pathophysiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico
| | - Jeanet Serafín-López
- Department of Immunology, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico
| |
Collapse
|
21
|
Dawood AA. The efficacy of Paxlovid against COVID-19 is the result of the tight molecular docking between M pro and antiviral drugs (nirmatrelvir and ritonavir). Adv Med Sci 2023; 68:1-9. [PMID: 36368287 PMCID: PMC9626444 DOI: 10.1016/j.advms.2022.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/05/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022]
Abstract
Purpose Currently, a number of medications for coronavirus disease 2019 (COVID-19) treatment are tested in clinical trials; however, credible clinical studies are becoming increasingly difficult to come by. Paxlovid is a ritonavir-boosted nirmatrelvir drug that the U.S. Food and Drug Administration (FDA) authorized for the treatment of COVID-19. This study aimed to demonstrate the interaction of nirmatrelvir and ritonavir on the active site of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro). Materials and methods To locate the optimal docking between Mpro and antiviral drugs, and to conduct dynamic simulations between atoms in the fusion areas, various bioinformatics and mathematical equations were applied. Results According to the docking data, nirmatrelvir has a stronger interaction with Mpro than ritonavir, which has more multiple bonds. Molecular docking of antiviral drugs such as Paxlovid has a significant impact on the treatment of COVID-19 virus. Conclusions According to this study, Paxlovid may work on new strains, including Omicron, because the Mpro mutation P132H in the Omicron variant has no direct effect on the protein.
Collapse
|
22
|
Alamri SS, Alsaieedi A, Khouqeer Y, Afeef M, Alharbi S, Algaissi A, Alghanmi M, Altorki T, Zawawi A, Alfaleh MA, Hashem AM, Alhabbab R. The importance of combining serological testing with RT-PCR assays for efficient detection of COVID-19 and higher diagnostic accuracy. PeerJ 2023; 11:e15024. [PMID: 37065688 PMCID: PMC10103696 DOI: 10.7717/peerj.15024] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/17/2023] [Indexed: 04/18/2023] Open
Abstract
Misdiagnosing suspected COVID-19 individuals could largely contribute to the viruses transmission, therefore, making an accurate diagnosis of infected subjects vital in minimizing and containing the disease. Although RT-PCR is the standard method in detecting COVID-19, it is associated with some limitations, including possible false negative results. Therefore, serological testing has been suggested as a complement assay to RT-PCR to support the diagnosis of acute infections. In this study, 15 out of 639 unvaccinated healthcare workers (HCWs) were tested negative for COVID-19 by RT-PCR and were found seropositive for SARS-CoV-2 nucleocapsid protein-specific IgM and IgG antibodies. These participants underwent additional confirmatory RT-PCR and SARS-CoV-2 spike-specific ELISA tests. Of the 15 individuals, nine participants were found negative by second RT-PCR but seropositive for anti-spike IgM and IgG antibodies and neutralizing antibodies confirming their acute infection. At the time of collection, these nine individuals were in close contact with COVID-19-confirmed patients, with 77.7% reporting COVID-19-related symptoms. These results indicate that including serological tests in the current testing profile can provide better outcomes and help contain the spread of the virus by increasing diagnostic accuracy to prevent future outbreaks rapidly.
Collapse
Affiliation(s)
- Sawsan S. Alamri
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahdab Alsaieedi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Yousef Khouqeer
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Marwah Afeef
- Study & Research Department, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Samiyah Alharbi
- Intensive Care Unit, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Abdullah Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Medical Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Maimonah Alghanmi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tarfa Altorki
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat Zawawi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed A. Alfaleh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rowa Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
Petrović V, Vuković V, Patić A, Marković M, Ristić M. Immunogenicity of BNT162b2, BBIBP-CorV, Gam-COVID-Vac and ChAdOx1 nCoV-19 Vaccines Six Months after the Second Dose: A Longitudinal Prospective Study. Vaccines (Basel) 2022; 11:56. [PMID: 36679901 PMCID: PMC9865554 DOI: 10.3390/vaccines11010056] [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: 10/31/2022] [Revised: 12/04/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Many available SARS-CoV-2 vaccines demonstrated good humoral response, but studies directly comparing their immunogenicity in the general population are lacking. We evaluated the medium−term kinetics of anti-S SARS-CoV-2 antibodies (Abs) at one and six months after the second dose of BNT162b2, BBIBP-CorV, and Gam-COVID-Vac. Immunogenicity at six months was directly compared between BNT162b2, BBIBP-CorV, Gam-COVID-Vac, and ChAdOx1 nCoV-19. Participants ≥ 20 years old from Novi Sad, Serbia, without prior SARS-CoV-2 infection, were included. Anti S1/S2 IgG antibodies were measured using quantitative LIAISON SARS-CoV-2 assay. A total of 368 participants were included: 231 (62.77%) had sera collected at two time points. Two doses of BNT162b2 were received by 37.50% of participants, followed by BBIBP-CorV (22.01%), Gam-COVID-Vac (21.47%), and ChAdOx1 nCoV-19 (19.02%). Mean Ab levels at the 28th day and 6 months were 216.55 (SD = 105.73) AU/mL and 75.68 (SD = 57.30) for BNT162b2, 194.38 (SD = 140.24) and 90.53 (SD = 111.30) for Gam-COVID-Vac, and 72.74 (SD = 80.04) and 24.43 (SD = 38.43) for BBIBP-CorV group (p < 0.01, between two time points across all three groups), with a significant difference between women and men (p < 0.01, for both sexes). At the sixth month post-vaccination, the highest mean Ab level was detected in Gam-COVID-Vac group (91.28 AU/mL, SD = 95.96), followed by BNT162b2 (85.25 AU/mL, SD = 60.02), ChAdOx1 nCoV-19 (64.22 AU/mL, SD = 65.30), and BBIBP-CorV (25.26 AU/mL, SD = 36.92) (p < 0.01). Anti-spike IgG persistence was demonstrated six months post-vaccination with a significant decline in Ab levels. These results suggest a lower protection against SARS-CoV-2 over time. Our findings support the introduction of additional (booster) doses.
Collapse
Affiliation(s)
- Vladimir Petrović
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
| | - Vladimir Vuković
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
| | - Aleksandra Patić
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
- Department of Microbiology with Parasitology and Immunology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Miloš Marković
- Department of Immunology, Faculty of Medicine, Institute of Microbiology and Immunology, University of Belgrade, 11000 Belgrade, Serbia
| | - Mioljub Ristić
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
| |
Collapse
|
24
|
Sun Y, Zhao B, Wang Y, Chen Z, Zhang H, Qu L, Zhao Y, Song J. Optimization of potential non-covalent inhibitors for the SARS-CoV-2 main protease inspected by a descriptor of the subpocket occupancy. Phys Chem Chem Phys 2022; 24:29940-29951. [PMID: 36468652 DOI: 10.1039/d2cp03681a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The main protease is regarded as an essential drug target for treating Coronavirus Disease 2019. In the present study, 13 marketed drugs were investigated to explore the possible binding mechanism, utilizing molecular docking, molecular dynamics simulation, and MM-PB(GB)SA binding energy calculations. Our results suggest that fusidic acid, polydatin, SEN-1269, AZD6482, and UNC-2327 have high binding affinities of more than 23 kcal mol-1. A descriptor was defined for the energetic occupancy of the subpocket, and it was found that S4 had a low occupancy of less than 10% on average. The molecular optimization of ADZ6482 via reinforcement learning algorithms was carried out to screen out three lead compounds, in which slight structural changes give more considerable binding energies and an occupancy of the S4 subpocket of up to 43%. The energetic occupancy could be a useful descriptor for evaluating the local binding affinity for drug design.
Collapse
Affiliation(s)
- Yujia Sun
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, Henan, 450001, P. R. China.
| | - Bodi Zhao
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, Henan, 450001, P. R. China.
| | - Yuqi Wang
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, Henan, 450001, P. R. China.
| | - Zitong Chen
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, Henan, 450001, P. R. China.
| | - Huaiyu Zhang
- Institute of Computational Quantum Chemistry, College of Chemistry and Materials Science, Hebei Normal University, Shijiazhuang, Hebei, 050024, P. R. China
| | - Lingbo Qu
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, Henan, 450001, P. R. China.
| | - Yuan Zhao
- The Key Laboratory of Natural Medicine and Immuno - Engineering, Henan University, Kaifeng, Henan, 475000, P. R. China
| | - Jinshuai Song
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, Henan, 450001, P. R. China.
| |
Collapse
|
25
|
Abstract
The COVID-19 pandemic has posed a significant health threat globally. Timely and appropriate vaccination is a key step to reduce the morbidity and mortality from COVID-19. The clinical course of COVID-19 infection and the effects of COVID-19 vaccination are influenced by patients' health situations and involve a systemic physiological reaction. Just like an "endocrine phenotype" of COVID-19 infection, endocrine dysfunction after COVID-19 vaccination also acquired clinical concerns. In the present review, we briefly introduce the commonly available vaccines against SARS-CoV-2, summarize the influence of COVID-19 vaccines on the endocrine system, and explore the underlying pathogenic mechanisms.
Collapse
Affiliation(s)
- Ying Zhao
- Geriatric Medicine Center, Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Department of Endocrinology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiaohong Wu
- Geriatric Medicine Center, Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Department of Endocrinology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
26
|
Abstract
The World Health Organisation has reported that the viral disease known as COVID-19, caused by SARS-CoV-2, is the leading cause of death by a single infectious agent. This narrative review examines certain components of the pandemic: its origins, early clinical data, global and UK-focussed epidemiology, vaccination, variants, and long COVID.
Collapse
Affiliation(s)
- A. D. Blann
- School of Applied Sciences, University of Huddersfield Queensgate, Huddersfield, United Kingdom
| | | |
Collapse
|
27
|
Hamill V, Noll L, Lu N, Tsui WNT, Porter EP, Gray M, Sebhatu T, Goerl K, Brown S, Palinski R, Thomason S, Almes K, Retallick J, Bai J. Molecular detection of SARS-CoV-2 strains and differentiation of Delta variant strains. Transbound Emerg Dis 2022; 69:2879-2889. [PMID: 34964565 PMCID: PMC9240106 DOI: 10.1111/tbed.14443] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/03/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022]
Abstract
The Delta variant of SARS-CoV-2 has now become the predominant strain in the global COVID-19 pandemic. Strain coverage of some detection assays developed during the early pandemic stages has declined due to periodic mutations in the viral genome. We have developed a real-time RT-PCR (RT-qPCR) for SARS-CoV-2 detection that provides nearly 100% strain coverage, and differentiation of highly transmissible Delta variant strains. All full or nearly full (≥28 kb) SARS-CoV-2 genomes (n = 403,812), including 6422 Delta and 280 Omicron variant strains, were collected from public databases at the time of analysis and used for assay design. The two amino acid deletions in the spike gene (S-gene, Δ156-157) that is characteristic of the Delta variant were targeted during the assay design. Although strain coverage for the Delta variant was very high (99.7%), detection coverage for non-Delta wild-type strains was 93.9%, mainly due to the confined region of design. To increase strain coverage of the assay, the design for CDC N1 target was added to the assay. In silico analysis of 403,812 genomes indicated a 95.4% strain coverage for the CDC N1 target, however, in combination with our new non-Delta S-gene target, total coverage for non-Delta wild-type strains increased to 99.8%. A human 18S rRNA gene was also analyzed and used as an internal control. The final four-plex RT-qPCR assay generated PCR amplification efficiencies between 95.4% and 102.0% with correlation coefficients (R2 ) of >0.99 for cloned positive controls; Delta and non-Delta human clinical samples generated PCR efficiencies of 93.4%-97.0% and R2 > 0.99. The assay also detects 98.6% of 280 Omicron sequences. Assay primers and probes have no match to other closely related human coronaviruses, and did not produce a signal from samples positive to selected animal coronaviruses. Genotypes of selected clinical samples identified by the RT-qPCR were confirmed by Sanger sequencing.
Collapse
Affiliation(s)
- Vaughn Hamill
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
| | - Lance Noll
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Nanyan Lu
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
- Division of Biology, Kansas State University, Manhattan, KS 66506, United States
| | - Wai Ning Tiffany Tsui
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
| | - Elizabeth Poulsen Porter
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
| | - Mark Gray
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
| | - Tesfaalem Sebhatu
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
| | - Kyle Goerl
- Lafene Health Center, Kansas State University, Manhattan, KS 66506, United States
| | - Susan Brown
- Division of Biology, Kansas State University, Manhattan, KS 66506, United States
| | - Rachel Palinski
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Sasha Thomason
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Kelli Almes
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Jamie Retallick
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Jianfa Bai
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, United States
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| |
Collapse
|
28
|
Replicating RNA platform enables rapid response to the SARS-CoV-2 Omicron variant and elicits enhanced protection in naïve hamsters compared to ancestral vaccine. EBioMedicine 2022; 83:104196. [PMID: 35932641 PMCID: PMC9349033 DOI: 10.1016/j.ebiom.2022.104196] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/01/2022] [Accepted: 07/15/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In late 2021, the SARS-CoV-2 Omicron (B.1.1.529) variant of concern (VoC) was reported with many mutations in the viral spike protein that were predicted to enhance transmissibility and allow viral escape of neutralizing antibodies. Within weeks of the first report of B.1.1.529, this VoC has rapidly spread throughout the world, replacing previously circulating strains of SARS-CoV-2 and leading to a resurgence in COVID-19 cases even in populations with high levels of vaccine- and infection-induced immunity. Studies have shown that B.1.1.529 is less sensitive to protective antibody conferred by previous infections and vaccines developed against earlier lineages of SARS-CoV-2. The ability of B.1.1.529 to spread even among vaccinated populations has led to a global public health demand for updated vaccines that can confer protection against B.1.1.529. METHODS We rapidly developed a replicating RNA vaccine expressing the B.1.1.529 spike and evaluated immunogenicity in mice and hamsters. We also challenged hamsters with B.1.1.529 and evaluated whether vaccination could protect against viral shedding and replication within respiratory tissue. FINDINGS We found that mice previously immunized with A.1-specific vaccines failed to elevate neutralizing antibody titers against B.1.1.529 following B.1.1.529-targeted boosting, suggesting pre-existing immunity may impact the efficacy of B.1.1.529-targeted boosters. Furthermore, we found that our B.1.1.529-targeted vaccine provides superior protection compared to the ancestral A.1-targeted vaccine in hamsters challenged with the B.1.1.529 VoC after a single dose of each vaccine. INTERPRETATION Our data suggest that B.1.1.529-targeted vaccines may provide superior protection against B.1.1.529 but pre-existing immunity and timing of boosting may need to be considered for optimum protection. FUNDING This research was supported in part by the Intramural Research Program, NIAID/NIH, Washington Research Foundation and by grants 27220140006C (JHE), AI100625, AI151698, and AI145296 (MG).
Collapse
|
29
|
Zhang Y, Zheng X, Sheng W, Liang H, Zhao Y, Zhu X, Yang R, Zhang Y, Dong X, Li W, Pei F, Ding L, Chang Z, Deng L, Yuan G, Yang Z, Zhu D, Yang X, Wang H. Alum/CpG Adjuvanted Inactivated COVID-19 Vaccine with Protective Efficacy against SARS-CoV-2 and Variants. Vaccines (Basel) 2022; 10:vaccines10081208. [PMID: 36016098 PMCID: PMC9413105 DOI: 10.3390/vaccines10081208] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Since the beginning of the COVID-19 pandemic, numerous variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged, including five variants of concern (VOC) strains listed by the WHO: Alpha, Beta, Gamma, Delta and Omicron. Extensive studies have shown that most of these VOC strains, especially the currently dominant variant Omicron, can escape the host immune response induced by existing COVID-19 vaccines to different extents, which poses considerable risk to the health of human beings around the world. In the present study, we developed a vaccine based on inactivated SARS-CoV-2 and an adjuvant consisting of aluminum hydroxide (alum) and CpG. The immunogenicity and safety of the vaccine were investigated in rats. The candidate vaccine elicited high titers of SARS-CoV-2-spike-specific IgG antibody and neutralizing antibody in immunized rats, which not only neutralize the original SARS-CoV-2, but also showed great cross-neutralization activity against the Beta, Delta and Omicron variants.
Collapse
Affiliation(s)
- Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
- China National Biotec Group Company Limited, Beijing 100024, China
| | - Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Wang Sheng
- College of Life Sciences and Biotechnology, Beijing University of Technology, Beijing 100021, China;
| | - Hongyang Liang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Yuxiu Zhao
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Xiujuan Zhu
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Rong Yang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Yadan Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Xiaofei Dong
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Weidong Li
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Fei Pei
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Ling Ding
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Zhen Chang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Li Deng
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Guangying Yuan
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Zhaona Yang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Di Zhu
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
| | - Xiaoming Yang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
- China National Biotec Group Company Limited, Beijing 100024, China
- Correspondence: (X.Y.); (H.W.)
| | - Hui Wang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (X.Z.); (H.L.); (Y.Z.); (X.Z.); (R.Y.); (Y.Z.); (X.D.); (W.L.); (F.P.); (L.D.); (Z.C.); (L.D.); (G.Y.); (Z.Y.); (D.Z.)
- Correspondence: (X.Y.); (H.W.)
| |
Collapse
|
30
|
Characterization and Utilization of Disulfide-Bonded SARS-CoV-2 Receptor Binding Domain of Spike Protein Synthesized by Wheat Germ Cell-Free Production System. Viruses 2022; 14:v14071461. [PMID: 35891441 PMCID: PMC9321213 DOI: 10.3390/v14071461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 02/05/2023] Open
Abstract
The spike protein (SP) of SARS-CoV-2 is an important target for COVID-19 therapeutics and vaccines as it binds to the ACE2 receptor and enables viral infection. Rapid production and functional characterization of properly folded SP is of the utmost importance for studying the immunogenicity and receptor-binding activity of this protein considering the emergence of highly infectious viral variants. In this study, we attempted to express the receptor-binding region (RBD) of SARS-CoV-2 SP containing disulfide bonds using the wheat germ cell-free protein synthesis system. By adding protein disulfide isomerase (PDI) and endoplasmic reticulum oxidase (ERO1α) to the translational reaction mixture, we succeeded in synthesizing a functionally intact RBD protein that can interact with ACE2. Using this RBD protein, we have developed a high-throughput AlphaScreen assay to evaluate the RBD–ACE2 interaction, which can be applied for drug screening and mutation analysis. Thus, our method sheds new light on the structural and functional properties of SARS-CoV-2 SP and has the potential to contribute to the development of new COVID-19 therapeutics.
Collapse
|
31
|
Bellamkonda N, Lambe UP, Sawant S, Nandi SS, Chakraborty C, Shukla D. Immune Response to SARS-CoV-2 Vaccines. Biomedicines 2022; 10:1464. [PMID: 35884770 PMCID: PMC9312515 DOI: 10.3390/biomedicines10071464] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 12/21/2022] Open
Abstract
COVID-19 vaccines have been developed to confer immunity against the SARS-CoV-2 infection. Prior to the pandemic of COVID-19 which started in March 2020, there was a well-established understanding about the structure and pathogenesis of previously known Coronaviruses from the SARS and MERS outbreaks. In addition to this, vaccines for various Coronaviruses were available for veterinary use. This knowledge supported the creation of various vaccine platforms for SARS-CoV-2. Before COVID-19 there are no reports of a vaccine being developed in under a year and no vaccine for preventing coronavirus infection in humans had ever been developed. Approximately nine different technologies are being researched and developed at various levels in order to design an effective COVID-19 vaccine. As the spike protein of SARS-CoV-2 is responsible for generating substantial adaptive immune response, mostly all the vaccine candidates have been targeting the whole spike protein or epitopes of spike protein as a vaccine candidate. In this review, we have compiled the immune response to SARS-CoV-2 infection and followed by the mechanism of action of various vaccine platforms such as mRNA vaccines, Adenoviral vectored vaccine, inactivated virus vaccines and subunit vaccines in the market. In the end we have also summarized the various adjuvants used in the COVID-19 vaccine formulation.
Collapse
Affiliation(s)
- Navya Bellamkonda
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | | | - Sonali Sawant
- ICMR-NIV, Mumbai Unit, A. D. Road, Parel, Mumbai 400012, India; (U.P.L.); (S.S.)
| | - Shyam Sundar Nandi
- ICMR-NIV, Mumbai Unit, A. D. Road, Parel, Mumbai 400012, India; (U.P.L.); (S.S.)
| | | | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
32
|
What Do Antibody Studies Tell Us about Viral Infections? Pathogens 2022; 11:pathogens11050560. [PMID: 35631081 PMCID: PMC9143683 DOI: 10.3390/pathogens11050560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
|
33
|
Martínez-Torres I, Tepale-Segura A, Castro-Escamilla O, Cancino-Diaz JC, Rodríguez-Martínez S, Perez-Tapia SM, Bonifaz LC, Cancino-Diaz ME. The Protective Role of pVHL in Imiquimod-Induced Psoriasis-like Skin Inflammation. Int J Mol Sci 2022; 23:ijms23095226. [PMID: 35563616 PMCID: PMC9104378 DOI: 10.3390/ijms23095226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022] Open
Abstract
Psoriasis is a chronic inflammatory disease distinguished by an excessive proliferation and abnormal differentiation of keratinocytes. Immune cells, such as T lymphocytes and neutrophils, and inflammatory cytokines, such as Tumor Necrosis Factor-α (TNF-α) and interleukin 17 (IL-17), are essential for maintaining psoriatic lesions. Additionally, a hypoxic milieu present in the skin promotes the expression of transcriptional factor hypoxia-inducible factor-1 alpha (HIF-1α). This protein regulates the expression of angiogenic and glycolytic factors, such as vascular endothelial grown factor and lactate dehydrogenase (LDH), both relevant in chronic inflammation. The von Hippel–Lindau protein (pVHL) is a negative regulator of HIF-1α. Previously, we found that pVHL was almost absent in the lesions of psoriasis patients; therefore, we investigated the impact of rescue pVHL expression in lesional skin. We used the imiquimod-induced psoriasis-like mouse model as an adenoviral vector that allowed us to express pVHL in the skin. Our data show that, in lesional skin, pVHL expression was reduced, whereas HIF-1α was increased. Remarkably, the retrieval of pVHL prevented psoriatic lesions, diminishing erythema, scale, and epidermal and vascular thickness. Furthermore, pVHL expression was capable of reducing HIF-1α, LDH, TNF-α and immune cell infiltration (mainly IL-17+ neutrophils). In conclusion, our results demonstrate that pVHL has a protective role to play in the pathophysiology of psoriasis.
Collapse
Affiliation(s)
- Isaí Martínez-Torres
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prolongación de Carpio, Col. Santo Tomas, Alcaldía Miguel Hidalgo, Ciudad de México C.P. 11340, Mexico; (I.M.-T.); (A.T.-S.); (S.R.-M.)
| | - Araceli Tepale-Segura
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prolongación de Carpio, Col. Santo Tomas, Alcaldía Miguel Hidalgo, Ciudad de México C.P. 11340, Mexico; (I.M.-T.); (A.T.-S.); (S.R.-M.)
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330 Col. Doctores, Alcaldía Cuauhtémoc, Ciudad de México C.P. 06720, Mexico; (O.C.-E.); or (L.C.B.)
| | - Octavio Castro-Escamilla
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330 Col. Doctores, Alcaldía Cuauhtémoc, Ciudad de México C.P. 06720, Mexico; (O.C.-E.); or (L.C.B.)
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil De México Federico Gómez, Dr. Márquez 162. Col. Doctores, Alcaldía Cuauhtémoc, Ciudad de México C.P. 06720, Mexico
| | - Juan Carlos Cancino-Diaz
- Departamento de Microbiologia, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prolongación de Carpio, Col. Santo Tomas, Alcaldia Miguel Hidalgo, Ciudad de México C.P. 11340, Mexico;
| | - Sandra Rodríguez-Martínez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prolongación de Carpio, Col. Santo Tomas, Alcaldía Miguel Hidalgo, Ciudad de México C.P. 11340, Mexico; (I.M.-T.); (A.T.-S.); (S.R.-M.)
| | - Sonia Mayra Perez-Tapia
- Unidad de Desarrollo e Invstigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México C.P. 11340, Mexico;
| | - Laura C. Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330 Col. Doctores, Alcaldía Cuauhtémoc, Ciudad de México C.P. 06720, Mexico; (O.C.-E.); or (L.C.B.)
- Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330 Col. Doctores, Alcaldía Cuauhtémoc, Ciudad de México C.P. 06720, Mexico
| | - Mario Eugenio Cancino-Diaz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prolongación de Carpio, Col. Santo Tomas, Alcaldía Miguel Hidalgo, Ciudad de México C.P. 11340, Mexico; (I.M.-T.); (A.T.-S.); (S.R.-M.)
- Correspondence: ; Tel.: +52-55-57-29-60-600 (ext. 62355)
| |
Collapse
|
34
|
Gutiérrez-Bautista JF, López-Nevot MÁ, Gómez-Vicente E, Quesada T, Marín EM, Rodríguez A, Rodríguez AI, Rodríguez-Granger J, Cobo F, Sampedro A. Study of humoral and cellular immunity in vaccinated with mRNA-1273. APMIS 2022; 130:261-269. [PMID: 35196403 PMCID: PMC9111507 DOI: 10.1111/apm.13215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/19/2022] [Indexed: 11/29/2022]
Abstract
The new vaccines against SARS‐CoV‐2 have raised a lot of expectations about their ability to induce immunity and the duration of this. This is the case of mRNA vaccines such as Moderna's mRNA‐1273. Therefore, it is necessary to study the humoral and cellular immunity generated by these vaccines. Our objectives are determining what is the normal response of antibody production, and what is the level of protective antibodies and monitoring patients in case of subsequent infection with COVID‐19. We present the first results of a longitudinal study of the humoral response in 601 health workers vaccinated with Moderna. The results show a humoral immunity at 90 days after the second dose of 100%, with a strong decrease between the levels of circulating anti‐S IgG antibodies between days 30 and 90 post‐vaccination. Observing a steeper decline in those who had higher titles at the beginning. In addition, we present a cellular response of 86% at three months after the second dose, which is related to low humoral response.
Collapse
Affiliation(s)
- Juan Francisco Gutiérrez-Bautista
- Departamento de Bioquímica, Biología Molecular III e Inmunología, University of Granada, Granada, Spain.,Programa de doctorado en Biomedicina, University of Granada, Granada, Spain.,Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Miguel Ángel López-Nevot
- Departamento de Bioquímica, Biología Molecular III e Inmunología, University of Granada, Granada, Spain.,Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Esther Gómez-Vicente
- Servicio de Microbiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Trinidad Quesada
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Eva María Marín
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Ana Rodríguez
- Servicio de Microbiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Ana Isabel Rodríguez
- Servicio de Microbiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | - Fernando Cobo
- Servicio de Microbiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Antonio Sampedro
- Servicio de Microbiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| |
Collapse
|
35
|
Ilinykh PA, Periasamy S, Huang K, Kuzmina NA, Ramanathan P, Meyer MN, Mire CE, Kuzmin IV, Bharaj P, Endsley JR, Chikina M, Sealfon SC, Widen SG, Endsley MA, Bukreyev A. A single intranasal dose of human parainfluenza virus type 3-vectored vaccine induces effective antibody and memory T cell response in the lungs and protects hamsters against SARS-CoV-2. NPJ Vaccines 2022; 7:47. [PMID: 35468973 PMCID: PMC9038905 DOI: 10.1038/s41541-022-00471-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/21/2022] [Indexed: 12/31/2022] Open
Abstract
Respiratory tract vaccination has an advantage of needle-free delivery and induction of mucosal immune response in the portal of SARS-CoV-2 entry. We utilized human parainfluenza virus type 3 vector to generate constructs expressing the full spike (S) protein of SARS-CoV-2, its S1 subunit, or the receptor-binding domain, and tested them in hamsters as single-dose intranasal vaccines. The construct bearing full-length S induced high titers of neutralizing antibodies specific to S protein domains critical to the protein functions. Robust memory T cell responses in the lungs were also induced, which represent an additional barrier to infection and should be less sensitive than the antibody responses to mutations present in SARS-CoV-2 variants. Following SARS-CoV-2 challenge, animals were protected from the disease and detectable viral replication. Vaccination prevented induction of gene pathways associated with inflammation. These results indicate advantages of respiratory vaccination against COVID-19 and inform the design of mucosal SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Sivakumar Periasamy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Palaniappan Ramanathan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Michelle N Meyer
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Chad E Mire
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ivan V Kuzmin
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Preeti Bharaj
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Jessica R Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Maria Chikina
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stuart C Sealfon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven G Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mark A Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
- Galveston National Laboratory, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
36
|
Toro-Ascuy D, Cifuentes-Muñoz N, Avaria A, Pereira-Montecinos C, Cruzat G, Peralta-Arancibia K, Zorondo-Rodríguez F, Fuenzalida LF. Factors Influencing the Acceptance of COVID-19 Vaccines in a Country with a High Vaccination Rate. Vaccines (Basel) 2022; 10:vaccines10050681. [PMID: 35632437 PMCID: PMC9145438 DOI: 10.3390/vaccines10050681] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/15/2022] [Accepted: 04/21/2022] [Indexed: 12/13/2022] Open
Abstract
Control of the COVID-19 pandemic largely depends on the effectiveness of the vaccination process. An understanding of the factors that underlie the willingness to accept vaccination contributes pivotal information to controlling the pandemic. We analyzed the association between the willingness to accept the available COVID-19 vaccines and vaccine determinants amidst the Chilean vaccination process. Individual-level survey data was collected from 744 nationally representative respondents and multivariate regression models were used to estimate the association between outcome and explanatory variables. We found that trust in COVID-19 vaccines, scientists, and medical professionals significantly increased the willingness to: accept the vaccines and booster doses, as well as annual vaccinations and the vaccination of children. Our results are critical to understanding the acceptance of COVID-19 vaccines in the context of a country with one of the world’s highest vaccination rates. We provide useful information for decision-making and policy design, in addition to establishing guidelines regarding how to effectively explain vaccination programs to citizens.
Collapse
Affiliation(s)
- Daniela Toro-Ascuy
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile; (D.T.-A.); (N.C.-M.); (C.P.-M.); (G.C.); (K.P.-A.)
| | - Nicolás Cifuentes-Muñoz
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile; (D.T.-A.); (N.C.-M.); (C.P.-M.); (G.C.); (K.P.-A.)
| | - Andrea Avaria
- Facultad de Ciencias Sociales y Humanidades, Universidad Autónoma de Chile, Santiago 8910132, Chile;
| | - Camila Pereira-Montecinos
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile; (D.T.-A.); (N.C.-M.); (C.P.-M.); (G.C.); (K.P.-A.)
| | - Gilena Cruzat
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile; (D.T.-A.); (N.C.-M.); (C.P.-M.); (G.C.); (K.P.-A.)
| | - Katherine Peralta-Arancibia
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile; (D.T.-A.); (N.C.-M.); (C.P.-M.); (G.C.); (K.P.-A.)
| | - Francisco Zorondo-Rodríguez
- Departamento de Gestión Agraria, Facultad Tecnológica, Universidad de Santiago de Chile, Santiago 9170125, Chile
- Correspondence: (F.Z.-R.); (L.F.F.); Tel.: +56-2-27180565 (F.Z.-R.); +56-2-23036662 (L.F.F.)
| | - Loreto F. Fuenzalida
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile; (D.T.-A.); (N.C.-M.); (C.P.-M.); (G.C.); (K.P.-A.)
- Correspondence: (F.Z.-R.); (L.F.F.); Tel.: +56-2-27180565 (F.Z.-R.); +56-2-23036662 (L.F.F.)
| |
Collapse
|
37
|
Brand R, Nosrat S, Späth C, Timme S. Using COVID-19 Pandemic as a Prism: A Systematic Review of Methodological Approaches and the Quality of Empirical Studies on Physical Activity Behavior Change. Front Sports Act Living 2022; 4:864468. [PMID: 35529420 PMCID: PMC9069113 DOI: 10.3389/fspor.2022.864468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/22/2022] [Indexed: 11/28/2022] Open
Abstract
Background The COVID-19 pandemic has highlighted the importance of scientific endeavors. The goal of this systematic review is to evaluate the quality of the research on physical activity (PA) behavior change and its potential to contribute to policy-making processes in the early days of COVID-19 related restrictions. Methods We conducted a systematic review of methodological quality of current research according to PRISMA guidelines using Pubmed and Web of Science, of articles on PA behavior change that were published within 365 days after COVID-19 was declared a pandemic by the World Health Organization (WHO). Items from the JBI checklist and the AXIS tool were used for additional risk of bias assessment. Evidence mapping is used for better visualization of the main results. Conclusions about the significance of published articles are based on hypotheses on PA behavior change in the light of the COVID-19 pandemic. Results Among the 1,903 identified articles, there were 36% opinion pieces, 53% empirical studies, and 9% reviews. Of the 332 studies included in the systematic review, 213 used self-report measures to recollect prepandemic behavior in often small convenience samples. Most focused changes in PA volume, whereas changes in PA types were rarely measured. The majority had methodological reporting flaws. Few had very large samples with objective measures using repeated measure design (pre and during the pandemic). In addition to the expected decline in PA duration, these studies show that many of those who were active prepandemic, continued to be active during the pandemic. Conclusions Research responded quickly at the onset of the pandemic. However, most of the studies lacked robust methodology, and PA behavior change data lacked the accuracy needed to guide policy makers. To improve the field, we propose the implementation of longitudinal cohort studies by larger organizations such as WHO to ease access to data on PA behavior, and suggest those institutions set clear standards for this research. Researchers need to ensure a better fit between the measurement method and the construct being measured, and use both objective and subjective measures where appropriate to complement each other and provide a comprehensive picture of PA behavior.
Collapse
Affiliation(s)
- Ralf Brand
- Sport and Exercise Psychology, University of Potsdam, Potsdam, Germany
- Department of Kinesiology, Iowa State University, Ames, IA, United States
| | - Sanaz Nosrat
- Department of Health Sciences, Lehman College/City University of New York, New York, NY, United States
| | - Constantin Späth
- Sport and Exercise Psychology, University of Potsdam, Potsdam, Germany
| | - Sinika Timme
- Sport and Exercise Psychology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
38
|
Jhuti D, Rawat A, Guo CM, Wilson LA, Mills EJ, Forrest JI. Interferon Treatments for SARS-CoV-2: Challenges and Opportunities. Infect Dis Ther 2022; 11:953-972. [PMID: 35445964 PMCID: PMC9022612 DOI: 10.1007/s40121-022-00633-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/31/2022] [Indexed: 12/24/2022] Open
Abstract
Interferon (IFN) therapies are used to treat a variety of infections and diseases and could be used to treat SARS-CoV-2. However, optimal use and timing of IFN therapy to treat SARS-CoV-2 is not well documented. We aimed to synthesize available evidence to understand whether interferon therapy should be recommended for treatment compared to a placebo or standard of care in adult patients. We reviewed literature comparing outcomes of randomized control trials that used IFN therapy for adults diagnosed with SARS-CoV-2 between 2019 and 2021. Data were extracted from 11 of 669 screened studies. Evidence of IFN effectiveness was mixed. Five studies reported that IFN was a better therapy than the control, four found no or minimal difference between IFN and the control, and two concluded that IFN led to worse patient outcomes than the control. Evidence was difficult to compare because of high variability in outcome measures, intervention types and administration, subtypes of IFNs used and timing of interventions. We recommend standardized indicators and reporting for IFN therapy for SARS-CoV-2 to improve evidence synthesis and generation. While IFN therapy has the potential to be a viable treatment for SARS-CoV-2, especially when combined with antivirals and early administration, the lack of comparable of study outcomes prevents evidence synthesis and uptake.
Collapse
Affiliation(s)
| | - Angeli Rawat
- University of British Columbia, 2329 West Mall, Vancouver, BC, V6T 1Z4, Canada
| | | | - Lindsay A Wilson
- University of British Columbia, 2329 West Mall, Vancouver, BC, V6T 1Z4, Canada.,Platform Life Sciences, Vancouver, Canada
| | - Edward J Mills
- McMaster University, Hamilton, Canada.,Platform Life Sciences, Vancouver, Canada
| | - Jamie I Forrest
- University of British Columbia, 2329 West Mall, Vancouver, BC, V6T 1Z4, Canada. .,Platform Life Sciences, Vancouver, Canada.
| |
Collapse
|
39
|
The E484K Substitution in a SARS-CoV-2 Spike Protein Subunit Vaccine Resulted in Limited Cross-Reactive Neutralizing Antibody Responses in Mice. Viruses 2022; 14:v14050854. [PMID: 35632595 PMCID: PMC9146450 DOI: 10.3390/v14050854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 01/29/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), especially emerging variants, poses an increased threat to global public health. The significant reduction in neutralization activity against the variants such as B.1.351 in the serum of convalescent patients and vaccinated people calls for the design of new potent vaccines targeting the emerging variant. However, since most vaccines approved and in clinical trials are based on the sequence of the original SARS-CoV-2 strain, the immunogenicity and protective efficacy of vaccines based on the B.1.351 variant remain largely unknown. In this study, we evaluated the immunogenicity, induced neutralization activity, and protective efficacy of wild-type spike protein nanoparticle (S-2P) and mutant spike protein nanoparticle (S-4M-2P) carrying characteristic mutations of B.1.351 variant in mice. Although there was no significant difference in the induction of spike-specific IgG responses in S-2P- and S-4M-2P-immunized mice, neutralizing antibodies elicited by S-4M-2P exhibited noteworthy, narrower breadth of reactivity with SARS-CoV-2 variants compared with neutralizing antibodies elicited by S-2P. Furthermore, the decrease of induced neutralizing antibody breadth at least partly resulted from the amino acid substitution at position 484. Moreover, S-4M-2P vaccination conferred insufficient protection against live SARS-CoV-2 virus infection, while S-2P vaccination gave definite protection against SARS-CoV-2 challenge in mice. Together, our study provides direct evidence that the E484K substitution in a SARS-CoV-2 subunit protein vaccine limited the cross-reactive neutralizing antibody breadth in mice and, more importantly, draws attention to the unfavorable impact of this mutation in spike protein of SARS-CoV-2 variants on the induction of potent neutralizing antibody responses.
Collapse
|
40
|
Strategies for fighting pandemic virus infections: Integration of virology and drug delivery. J Control Release 2022; 343:361-378. [PMID: 35122872 PMCID: PMC8810279 DOI: 10.1016/j.jconrel.2022.01.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Respiratory viruses have sometimes resulted in worldwide pandemics, with the influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) being major participants. Long-term efforts have made it possible to control the influenza virus, but seasonal influenza continues to take many lives each year, and a pandemic influenza virus sometimes emerges. Although vaccines for coronavirus disease 2019 (COVID-19) have been developed, we are not yet able to coexist with the SARS-CoV-2. To overcome such viruses, it is necessary to obtain knowledge about international surveillance systems, virology, ecology and to determine that immune responses are effective. The information must then be transferred to drugs. Delivery systems would be expected to contribute to the rational development of drugs. In this review, virologist and drug delivery system (DDS) researchers discuss drug delivery strategies, especially the use of lipid-based nanocarriers, for fighting to respiratory virus infections.
Collapse
|
41
|
Hawman DW, Meade-White K, Archer J, Leventhal SS, Wilson D, Shaia C, Randall S, Khandhar AP, Krieger K, Hsiang TY, Gale M, Berglund P, Fuller DH, Feldmann H, Erasmus JH. SARS-CoV2 variant-specific replicating RNA vaccines protect from disease following challenge with heterologous variants of concern. eLife 2022; 11:e75537. [PMID: 35191378 PMCID: PMC8983041 DOI: 10.7554/elife.75537] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 02/17/2022] [Indexed: 11/14/2022] Open
Abstract
Despite mass public health efforts, the SARS-CoV2 pandemic continues as of late 2021 with resurgent case numbers in many parts of the world. The emergence of SARS-CoV2 variants of concern (VoCs) and evidence that existing vaccines that were designed to protect from the original strains of SARS-CoV-2 may have reduced potency for protection from infection against these VoC is driving continued development of second-generation vaccines that can protect against multiple VoC. In this report, we evaluated an alphavirus-based replicating RNA vaccine expressing Spike proteins from the original SARS-CoV-2 Alpha strain and recent VoCs delivered in vivo via a lipid inorganic nanoparticle. Vaccination of both mice and Syrian Golden hamsters showed that vaccination induced potent neutralizing titers against each homologous VoC but reduced neutralization against heterologous challenges. Vaccinated hamsters challenged with homologous SARS-CoV2 variants exhibited complete protection from infection. In addition, vaccinated hamsters challenged with heterologous SARS-CoV-2 variants exhibited significantly reduced shedding of infectious virus. Our data demonstrate that this vaccine platform can be updated to target emergent VoCs, elicits significant protective immunity against SARS-CoV2 variants and supports continued development of this platform.
Collapse
Affiliation(s)
- David W Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | | | - Shanna S Leventhal
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Drew Wilson
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Samantha Randall
- Department of Microbiology, University of Washington School of MedicineSeattleUnited States
| | | | | | - Tien-Ying Hsiang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of MedicineSeattleUnited States
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of MedicineSeattleUnited States
| | | | | | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Jesse H Erasmus
- HDT BioSeattleUnited States
- Department of Microbiology, University of Washington School of MedicineSeattleUnited States
| |
Collapse
|
42
|
Zhou X, Cheng L, Wang H, Liao X, Wang M, Wei L, Song S, Zhou B, Ma Z, Guo H, Ge X, Ju B, Zhang Z. The SARS-CoV-2 inactivated vaccine enhances the broad neutralization against variants in individuals recovered from COVID-19 up to one year. Emerg Microbes Infect 2022; 11:753-756. [PMID: 35184681 PMCID: PMC8903755 DOI: 10.1080/22221751.2022.2043728] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Xinrong Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Lin Cheng
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Haiyan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Xuejiao Liao
- Follow-up Department of Chronic Diseases, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Miao Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Lanlan Wei
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Shuo Song
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Bing Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Zhenghua Ma
- Follow-up Department of Chronic Diseases, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Huimin Guo
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Xiangyang Ge
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China.,Guangdong Key laboratory for anti-infection Drug Quality Evaluation, Shenzhen 518112, Guangdong Province, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China.,Guangdong Key laboratory for anti-infection Drug Quality Evaluation, Shenzhen 518112, Guangdong Province, China.,Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen 518112, Guangdong Province, China
| |
Collapse
|
43
|
Main protease mutants of SARS-CoV-2 variants remain susceptible to nirmatrelvir. Bioorg Med Chem Lett 2022; 62:128629. [PMID: 35182772 PMCID: PMC8856729 DOI: 10.1016/j.bmcl.2022.128629] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/25/2022]
Abstract
The COVID-19 pandemic continues to be a public health threat. Multiple mutations in the spike protein of emerging variants of SARS-CoV-2 appear to impact on the effectiveness of available vaccines. Specific antiviral agents are keenly anticipated but their efficacy may also be compromised in emerging variants. One of the most attractive coronaviral drug targets is the main protease (Mpro). A promising Mpro inhibitor of clinical relevance is the peptidomimetic nirmatrelvir (PF-07321332). We expressed Mpro of six SARS-CoV-2 lineages (C.37 Lambda, B.1.1.318, B.1.2, B.1.351 Beta, B.1.1.529 Omicron, P.2 Zeta), each of which carries a strongly prevalent missense mutation (G15S, T21I, L89F, K90R, P132H, L205V). Enzyme kinetics reveal that these Mpro variants are catalytically competent to a similar degree as the wildtype. We show that nirmatrelvir has similar potency against the variants as the wildtype. Our in vitro data suggest that the efficacy of the specific Mpro inhibitor nirmatrelvir is not compromised in current COVID-19 variants.
Collapse
|
44
|
Ju B, Zhou B, Song S, Fan Q, Ge X, Wang H, Cheng L, Guo H, Shu D, Liu L, Zhang Z. Potent antibody immunity to SARS-CoV-2 variants elicited by a third dose of inactivated vaccine. Clin Transl Med 2022; 12:e732. [PMID: 35220661 PMCID: PMC8882237 DOI: 10.1002/ctm2.732] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 12/31/2022] Open
Affiliation(s)
- Bin Ju
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
- Guangdong Key Laboratory for Anti‐infection Drug Quality EvaluationShenzhenChina
| | - Bing Zhou
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Shuo Song
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Qing Fan
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Xiangyang Ge
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Haiyan Wang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Lin Cheng
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Huimin Guo
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Dan Shu
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Lei Liu
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
- Department for Infectious DiseasesNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
| | - Zheng Zhang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenChina
- The Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhenChina
- Guangdong Key Laboratory for Anti‐infection Drug Quality EvaluationShenzhenChina
- Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical ScienceShenzhenChina
| |
Collapse
|
45
|
Wu J, Nie J, Zhang L, Song H, An Y, Liang Z, Yang J, Ding R, Liu S, Li Q, Li T, Cui Z, Zhang M, He P, Wang Y, Qu X, Hu Z, Wang Q, Huang W. The antigenicity of SARS-CoV-2 Delta variants aggregated 10 high-frequency mutations in RBD has not changed sufficiently to replace the current vaccine strain. Signal Transduct Target Ther 2022; 7:18. [PMID: 35046385 PMCID: PMC8767530 DOI: 10.1038/s41392-022-00874-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/08/2021] [Accepted: 12/24/2021] [Indexed: 11/08/2022] Open
Abstract
Emerging SARS-CoV-2 variants are the most serious problem for COVID-19 prophylaxis and treatment. To determine whether the SARS-CoV-2 vaccine strain should be updated following variant emergence like seasonal flu vaccine, the changed degree on antigenicity of SARS-CoV-2 variants and H3N2 flu vaccine strains was compared. The neutralization activities of Alpha, Beta and Gamma variants' spike protein-immunized sera were analysed against the eight current epidemic variants and 20 possible variants combining the top 10 prevalent RBD mutations based on the Delta variant, which were constructed using pseudotyped viruses. Meanwhile, the neutralization activities of convalescent sera and current inactivated and recombinant protein vaccine-elicited sera were also examined against all possible Delta variants. Eight HA protein-expressing DNAs elicited-animal sera were also tested against eight pseudotyped viruses of H3N2 flu vaccine strains from 2011-2019. Our results indicate that the antigenicity changes of possible Delta variants were mostly within four folds, whereas the antigenicity changes among different H3N2 vaccine strains were approximately 10-100-fold. Structural analysis of the antigenic characterization of the SARS-CoV-2 and H3N2 mutations supports the neutralization results. This study indicates that the antigenicity changes of the current SARS-CoV-2 may not be sufficient to require replacement of the current vaccine strain.
Collapse
MESH Headings
- Amino Acid Substitution
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/metabolism
- Antibodies, Viral/chemistry
- Antibodies, Viral/genetics
- Antibodies, Viral/metabolism
- Binding Sites
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/chemistry
- COVID-19 Vaccines/metabolism
- Epitopes/chemistry
- Epitopes/genetics
- Epitopes/immunology
- Gene Expression
- Humans
- Immune Sera/chemistry
- Immunogenicity, Vaccine
- Influenza A Virus, H3N2 Subtype/chemistry
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/chemistry
- Influenza Vaccines/metabolism
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Models, Molecular
- Mutation
- Neutralization Tests
- Protein Binding
- Protein Conformation
- Protein Interaction Domains and Motifs
- SARS-CoV-2/chemistry
- SARS-CoV-2/immunology
- SARS-CoV-2/pathogenicity
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Viral Pseudotyping
Collapse
Affiliation(s)
- Jiajing Wu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Li Zhang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Hao Song
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yimeng An
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Ziteng Liang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Jing Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, 100101, Beijing, China
| | - Ruxia Ding
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Shuo Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Qianqian Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Tao Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Zhimin Cui
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Mengyi Zhang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Peng He
- Division of Hepatitis and Enteric Viral Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, 102629, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China
| | - Xiaowang Qu
- Translational Medicine Institute, First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Zhongyu Hu
- Division of Hepatitis and Enteric Viral Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, 102629, Beijing, China.
| | - Qihui Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), 102629, Beijing, China.
| |
Collapse
|
46
|
Wu X, Tang S, Wang Z, Ma X, Zhang L, Zhang F, Xiao L, Zhao S, Li Q, Wang Y, Wang Q, Chen K. Immune Enhancement by the Tetra-Peptide Hydrogel as a Promising Adjuvant for an H7N9 Vaccine against Highly Pathogenic H7N9 Virus. Vaccines (Basel) 2022; 10:130. [PMID: 35062791 PMCID: PMC8778772 DOI: 10.3390/vaccines10010130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Short peptide hydrogel was reported as a possible adjuvant for vaccines. In order to evaluate whether the Tetra-Peptide Hydrogel can be a promising adjuvant for an H7N9 vaccine against the highly pathogenic H7N9 virus, we conducted this study. METHODS Tetra-Peptide Hydrogels (D and L conformations) were prepared by a self-assembly system using a Naproxen acid modified tetra peptide of GFFY (Npx-GFFY). Mice received two immunizations with the D-Tetra-Peptide Hydrogel adjuvant vaccine, the L-Tetra-Peptide Hydrogel adjuvant vaccine, or the split vaccine. Fourteen days following the second dose, the mice were challenged with the highly pathogenic A/Guangdong/GZ8H002/2017(H7N9) virus. The mice were observed for signs of illness, weight loss, pathological alterations of the lung tissues and immune responses in the following 2 weeks. RESULTS The D/L-Tetra-Peptide Hydrogels resembled long bars with hinges on each other, with a diameter of ~10 nm. The H7N9 vaccine was observed to adhere to the hydrogel. All the unvaccinated mice were dead by 8 days post infection with H7N9. The mice immunized by the split H7N9 vaccine were protected against infection with H7N9. Mice immunized by D/L-Tetra-Peptide Hydrogel adjuvant vaccines experienced shorter symptomatic periods and their micro-neutralization titers were higher than in the split H7N9 vaccine at 2 weeks post infection. The hemagglutinating inhibition (HI) titer in the L-Tetra-Peptide Hydrogel adjuvant vaccine group was higher than that in the split H7N9 vaccine 1 week and 2 weeks post infection. The HI titer in the D-Tetra-Peptide Hydrogel adjuvant vaccine group was higher than that in the split H7N9 vaccine at 2 weeks post infection. CONCLUSION The D/L Tetra-Peptide Hydrogels increased the protection of the H7N9 vaccine and could be promising adjuvants for H7N9 vaccines against highly pathogenic H7N9 virus.
Collapse
Affiliation(s)
- Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.W.); (L.Z.); (F.Z.); (L.X.); (S.Z.); (Q.L.)
| | - Songjia Tang
- Plastic and Aesthetic Surgery Department, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China;
| | - Zhehua Wang
- Department of Infectious Disease and Medical Clinical Laboratory, Zhejiang Hospital, 1229 Gudun Road, Xihu, Hangzhou 310012, China; (Z.W.); (X.M.)
| | - Xiaoyun Ma
- Department of Infectious Disease and Medical Clinical Laboratory, Zhejiang Hospital, 1229 Gudun Road, Xihu, Hangzhou 310012, China; (Z.W.); (X.M.)
| | - Lingjian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.W.); (L.Z.); (F.Z.); (L.X.); (S.Z.); (Q.L.)
| | - Fen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.W.); (L.Z.); (F.Z.); (L.X.); (S.Z.); (Q.L.)
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.W.); (L.Z.); (F.Z.); (L.X.); (S.Z.); (Q.L.)
| | - Shuai Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.W.); (L.Z.); (F.Z.); (L.X.); (S.Z.); (Q.L.)
| | - Qian Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.W.); (L.Z.); (F.Z.); (L.X.); (S.Z.); (Q.L.)
| | - Ying Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Qingjing Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Keda Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.W.); (L.Z.); (F.Z.); (L.X.); (S.Z.); (Q.L.)
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| |
Collapse
|
47
|
Li T, Cui Z, Jia Y, Liang Z, Nie J, Zhang L, Wang M, Li Q, Wu J, Xu N, Liu S, Li X, An Y, Han P, Zhang M, Li Y, Qu X, Wang Q, Huang W, Wang Y. Aggregation of high-frequency RBD mutations of SARS-CoV-2 with three VOCs did not cause significant antigenic drift. J Med Virol 2022; 94:2108-2125. [PMID: 35032057 PMCID: PMC9015629 DOI: 10.1002/jmv.27596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 11/19/2022]
Abstract
Variants of SARS‐CoV‐2 continue to emerge, posing great challenges in outbreak prevention and control. It is important to understand in advance the impact of possible variants of concern (VOCs) on infectivity and antigenicity. Here, we constructed one or more of the 15 high‐frequency naturally occurring amino acid changes in the receptor‐binding domain (RBD) of Alpha, Beta, and Gamma variants. A single mutant of A520S, V367F, and S494P in the above three VOCs enhanced infectivity in ACE2‐overexpressing 293T cells of different species, LLC‐MK2 and Vero cells. Aggregation of multiple RBD mutations significantly reduces the infectivity of the possible three VOCs. Regarding neutralization, it is noteworthy that E484K, N501Y, K417N, and N439K predispose to monoclonal antibodies (mAbs) protection failure in the 15 high‐frequency mutations. Most importantly, almost all possible VOCs (single RBD mutation or aggregation of multiple mutations) showed no more than a fourfold decrease in neutralizing activity with convalescent sera, vaccine sera, and immune sera of guinea pigs with different immunogens, and no significant antigenic drift was formed. In conclusion, our pseudovirus results could reduce the concern that the aggregation of multiple high‐frequency mutations in the RBD of the spike protein of the three VOCs would lead to severe antigenic drift, and this would provide value for vaccine development strategies. Infectivity increased by adding three VOCs of V367F, S494P, or A520S. The infectivity of the three VOCs with multiple high‐frequency mutations decreased. Almost all of the possible variants of the three VOCs did not show severe antigenic drift.
Collapse
Affiliation(s)
- Tao Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Zhimin Cui
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China.,National Vaccine & Serum Institute, Beijing, China
| | - Yunfei Jia
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Ziteng Liang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Li Zhang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Meng Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Qianqian Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Jiajing Wu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Nan Xu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Shuo Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Xueli Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Yimeng An
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Pu Han
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Mengyi Zhang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Yuhua Li
- Department of Arboviral Vaccine, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Xiaowang Qu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Qihui Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), No. 31 Huatuo Street, Daxing District, Beijing, China
| |
Collapse
|
48
|
McKimm-Breschkin JL, Hay AJ, Cao B, Cox RJ, Dunning J, Moen AC, Olson D, Pizzorno A, Hayden FG. COVID-19, Influenza and RSV: Surveillance-informed prevention and treatment - Meeting report from an isirv-WHO virtual conference. Antiviral Res 2021; 197:105227. [PMID: 34933044 PMCID: PMC8684224 DOI: 10.1016/j.antiviral.2021.105227] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
The International Society for Influenza and other Respiratory Virus Diseases (isirv) and the WHO held a joint virtual conference from 19th-21st October 2021. While there was a major focus on the global response to the SARS-CoV-2 pandemic, including antivirals, vaccines and surveillance strategies, papers were also presented on treatment and prevention of influenza and respiratory syncytial virus (RSV). Potential therapeutics for SARS-CoV-2 included host-targeted therapies baricitinib, a JAK inhibitor, tocilizumab, an IL-6R inhibitor, verdinexor and direct acting antivirals ensovibep, S-217622, AT-527, and monoclonal antibodies casirivimab and imdevimab, directed against the spike protein. Data from trials of nirsevimab, a monoclonal antibody with a prolonged half-life which binds to the RSV F-protein, and an Ad26.RSV pre-F vaccine were also presented. The expanded role of the WHO Global Influenza Surveillance and Response System to address the SARS-CoV-2 pandemic was also discussed. This report summarizes the oral presentations given at this meeting for the benefit of the broader medical and scientific community involved in surveillance, treatment and prevention of respiratory virus diseases.
Collapse
Affiliation(s)
- Jennifer L McKimm-Breschkin
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia.
| | - Alan J Hay
- The Francis Crick Institute, London, UK.
| | - Bin Cao
- China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China.
| | - Rebecca J Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway.
| | - Jake Dunning
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | - Ann C Moen
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Daniel Olson
- University of Colorado School of Medicine and Colorado School of Public Health, Anschutz Medical Campus, Aurora, CO, USA.
| | - Andrés Pizzorno
- International Center for Research in Infectious Diseases, University of Lyon, Lyon, France.
| | - Frederick G Hayden
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
49
|
Hawman DW, Meade-White K, Archer J, Leventhal S, Wilson D, Shaia C, Randall S, Khandhar AP, Hsiang TY, Gale M, Berglund P, Fuller DH, Feldmann H, Erasmus JH. SARS-CoV2 variant-specific replicating RNA vaccines protect from disease and pathology and reduce viral shedding following challenge with heterologous SARS-CoV2 variants of concern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.12.10.472134. [PMID: 34931189 PMCID: PMC8687464 DOI: 10.1101/2021.12.10.472134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Despite mass public health efforts, the SARS-CoV2 pandemic continues as of late-2021 with resurgent case numbers in many parts of the world. The emergence of SARS-CoV2 variants of concern (VoC) and evidence that existing vaccines that were designed to protect from the original strains of SARS-CoV-2 may have reduced potency for protection from infection against these VoC is driving continued development of second generation vaccines that can protect against multiple VoC. In this report, we evaluated an alphavirus-based replicating RNA vaccine expressing Spike proteins from the original SARS-CoV-2 Alpha strain and recent VoCs delivered in vivo via a lipid inorganic nanoparticle. Vaccination of both mice and Syrian Golden hamsters showed that vaccination induced potent neutralizing titers against each homologous VoC but reduced neutralization against heterologous challenges. Vaccinated hamsters challenged with homologous SARS-CoV2 variants exhibited complete protection from infection. In addition, vaccinated hamsters challenged with heterologous SARS-CoV-2 variants exhibited significantly reduced shedding of infectious virus. Our data demonstrate that this vaccine platform elicits significant protective immunity against SARS-CoV2 variants and supports continued development of this platform.
Collapse
Affiliation(s)
- David W Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | | | - Shanna Leventhal
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Drew Wilson
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Samantha Randall
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | - Tien-Ying Hsiang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | | | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Jesse H Erasmus
- HDT Bio, Seattle, WA 98102, USA
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| |
Collapse
|
50
|
Sanchez S, Palacio N, Dangi T, Ciucci T, Penaloza-MacMaster P. Fractionating a COVID-19 Ad5-vectored vaccine improves virus-specific immunity. Sci Immunol 2021; 6:eabi8635. [PMID: 34648369 DOI: 10.1126/sciimmunol.abi8635] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nicole Palacio
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Thomas Ciucci
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA.,Department of Microbiology and Immunology, Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|