1
|
Saha A, Ghosh Roy S, Dwivedi R, Tripathi P, Kumar K, Nambiar SM, Pathak R. Beyond the Pandemic Era: Recent Advances and Efficacy of SARS-CoV-2 Vaccines Against Emerging Variants of Concern. Vaccines (Basel) 2025; 13:424. [PMID: 40333293 PMCID: PMC12031379 DOI: 10.3390/vaccines13040424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccination has been instrumental in curbing the transmission of SARS-CoV-2 and mitigating the severity of clinical manifestations associated with COVID-19. Numerous COVID-19 vaccines have been developed to this effect, including BioNTech-Pfizer and Moderna's mRNA vaccines, as well as adenovirus vector-based vaccines such as Oxford-AstraZeneca. However, the emergence of new variants and subvariants of SARS-CoV-2, characterized by enhanced transmissibility and immune evasion, poses significant challenges to the efficacy of current vaccination strategies. In this review, we aim to comprehensively outline the landscape of emerging SARS-CoV-2 variants of concern (VOCs) and sub-lineages that have recently surfaced in the post-pandemic years. We assess the effectiveness of existing vaccines, including their booster doses, against these emerging variants and subvariants, such as BA.2-derived sub-lineages, XBB sub-lineages, and BA.2.86 (Pirola). Furthermore, we discuss the latest advancements in vaccine technology, including multivalent and pan-coronavirus approaches, along with the development of several next-generation coronavirus vaccines, such as exosome-based, virus-like particle (VLP), mucosal, and nanomaterial-based vaccines. Finally, we highlight the key challenges and critical areas for future research to address the evolving threat of SARS-CoV-2 subvariants and to develop strategies for combating the emergence of new viral threats, thereby improving preparedness for future pandemics.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Sounak Ghosh Roy
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Naval Medical Research Command, Silver Spring, MD 20910, USA;
| | - Richa Dwivedi
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Kamal Kumar
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA;
| | - Shashank Manohar Nambiar
- Division of Hepatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
2
|
Hurst JR, Naghibosadat M, Budowski P, Liu J, Samaan P, Budiman F, Kurtesi A, Qi F, Menon H, Krishnan R, Abioye J, Gingras AC, Ostrowski M, Orozco NM, Kozak RA. Comparison of a SARS-CoV-2 mRNA booster immunization containing additional antigens to a spike-based mRNA vaccine against Omicron BA.5 infection in hACE2 mice. PLoS One 2024; 19:e0314061. [PMID: 39625929 PMCID: PMC11614295 DOI: 10.1371/journal.pone.0314061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
The emergence of SARS-CoV-2 variants presents challenges to vaccine effectiveness, underlining the necessity for next-generation vaccines with multiple antigens beyond the spike protein. Here, we investigated a multiantigenic booster containing spike and a chimeric construct composed of nucleoprotein (N) and membrane (M) proteins, comparing its efficacy to a spike-only booster against Omicron BA.5 in K18-hACE2 mice. Initially, mice were primed and boosted with Beta (B.1.351) spike-only mRNA, showing strong spike-specific T cell responses and neutralizing antibodies, albeit with limited cross-neutralization to Omicron variants. Subsequently, a spike-NM multiantigenic vaccine was then examined as a second booster dose for protection in hACE2-transgenic mice. Mice receiving either homologous spike-only or heterologous spike-NM booster had nearly complete inhibition of infectious virus shedding in oral swabs and reduced viral burdens in both lung and nasal tissues following BA.5 challenge. Examination of lung pathology further revealed that both spike-only and spike-NM boosters provided comparable protection against inflammatory infiltrates and fibrosis. Moreover, the spike-NM booster demonstrated neutralization efficacy in a pseudovirus assay against Wuhan-Hu-1, Beta, and Omicron variants akin to the spike-only booster. These findings indicate that supplementing spike with additional SARS-CoV-2 targets in a booster immunization confers equivalent immunity and protection against Omicron BA.5. This work highlights a promising strategy for individuals previously vaccinated with spike-only vaccines, potentially offering enhanced protection against emerging coronaviruses.
Collapse
Affiliation(s)
- Jacklyn R. Hurst
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Maedeh Naghibosadat
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Patrick Budowski
- Institute of Medical Sciences, University of Toronto, Ontario, Canada
| | - Jun Liu
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Frans Budiman
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alexandra Kurtesi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Fredo Qi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Haritha Menon
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | | | - Jumai Abioye
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mario Ostrowski
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Robert A. Kozak
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
- Division of Microbiology, Sunnybrook Health Sciences Centre, Department of Laboratory Medicine and Molecular Diagnostics, Toronto, ON, Canada
| |
Collapse
|
3
|
Garziano M, Cano Fiestas M, Vanetti C, Strizzi S, Murno ML, Clerici M, Biasin M. SARS-CoV-2 natural infection, but not vaccine-induced immunity, elicits cross-reactive immunity to OC43. Heliyon 2024; 10:e37928. [PMID: 39391514 PMCID: PMC11466580 DOI: 10.1016/j.heliyon.2024.e37928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
Background The recent SARS-CoV-2 pandemic renewed interest toward other non-severe acute respiratory syndrome human coronaviruses. Among these, OC43 is a seasonal human coronavirus widely diffused in the population (90 % seroprevalence in adults) which is responsible for mild respiratory symptoms. As OC43 protective immunity is short lasting, we investigated whether humoral immunity to SARS-CoV-2, induced by vaccination or spontaneous infection, protects against OC43 re-infection at either systemic or mucosal level. Methods A neutralization assay was conducted against "wild type" SARS-CoV-2 lineage B.1 (EU) and OC43 in VeroE6 cell lines using plasma and saliva samples from 49 subjects who were never infected and received three BNT162b2 RNA vaccine doses (SARS-CoV-2-vaccinated: SV) and from 25 SARS-CoV-2-infected and vaccinated subjects (SIV). The assays were performed right before (T0), fifteen days (T1) and three months (T2) after the third dose administration (SV) or post-infection (SIV). Results After the third vaccination dose was administered, SARS-CoV-2-specific neutralizing activity (NA) significantly augmented in SV saliva (p < 0.05) and plasma (p < 0.0001); yet, this NA was not protective against OC43. Conversely, in SIV, at T1, natural infection significantly increased NA against both SARS-CoV-2 (p < 0.01) and OC43 (p < 0.05) at systemic as well as mucosal level; still, this cross-reactivity vanished at T2. Of note, NA against SARS-CoV-2 and OC43 was shown to be higher in SIV compared to SV in plasma and saliva, as well; though, statistically significant differences were evident only in the oral mucosa at T1 (p < 0.05). Conclusions Our findings show that SARS-CoV-2 spontaneous infection triggers a more comprehensive and cross-reactive immunity than vaccine-induced immunity, protecting against OC43 at the systemic and mucosal levels. These results support the development of a pan-coronavirus vaccine able to prompt cross-reactive immunity even against seasonal coronaviruses, which could have enormous economic and health benefits globally.
Collapse
Affiliation(s)
- Micaela Garziano
- Laboratory of Immunobiology, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Mario Cano Fiestas
- Laboratory of Immunobiology, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Claudia Vanetti
- Laboratory of Immunobiology, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Sergio Strizzi
- Laboratory of Immunobiology, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Maria Luisa Murno
- Laboratory of Immunobiology, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Don C. Gnocchi Foundation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation, Milan, Italy
| | - Mara Biasin
- Laboratory of Immunobiology, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Rendon-Marin S, Rincón-Tabares DS, Tabares-Guevara JH, Arbeláez N, Forero-Duarte JE, Díaz FJ, Robledo SM, Hernandez JC, Ruiz-Saenz J. Evaluation of the Safety and Immunogenicity of a Multiple Epitope Polypeptide from Canine Distemper Virus (CDV) in Mice. Vaccines (Basel) 2024; 12:1140. [PMID: 39460307 PMCID: PMC11511104 DOI: 10.3390/vaccines12101140] [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: 08/08/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Morbillivirus canis is the etiological agent of a highly contagious disease that affects diverse domestic and wild animals. Vaccination is considered the most suitable strategy for controlling CDV dissemination, transmission, and distemper disease. However, the emergence of new CDV strains has led to the need to update the current vaccine strategies employed to prevent CDV infection in domestic and wild animals. Currently, there is a lack of effective alternatives for wild animals. Diverse computational tools, especially peptide-based therapies, enable the development of new universal vaccines. OBJECTIVE The aim of this study was to evaluate the safety and humoral and cellular immune response of a new generation of vaccines based on CDV peptides as single-peptide mixtures or multiepitope CDV polypeptides in mice. METHODS Twenty-four BALB/c mice were subjected to a three-dose regimen for 28 days. Seroconversion was evaluated via ELISA, and cellular immune responses were evaluated via flow cytometry through activation-induced markers (AIMs). RESULTS Compared with the placebo, the peptide mixture and multiepitope CDV polypeptide were safe, and seroconversion was statistically significant in the multiepitope CDV polypeptide and commercial vaccine (CV) groups. The numbers of antigen-specific CD4+CD134+ and IFN-γ+ T cells, CD8+ T cells and TNF-α- and IL-6-producing cells were greater in the mice immunized with the multiepitope CDV polypeptide than in the control mice. CONCLUSION This combined approach represents a potential step forward in developing new immunization candidates or enhancing current commercial vaccines to control CDV disease in domestic dogs and wild animals.
Collapse
Affiliation(s)
- Santiago Rendon-Marin
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680001, Colombia;
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050001, Colombia;
| | - Daniel-Santiago Rincón-Tabares
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Jorge H. Tabares-Guevara
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Natalia Arbeláez
- Grupo PECET, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (N.A.); (S.M.R.)
| | - Jorge E. Forero-Duarte
- Grupo de Investigación en Microbiología Ambiental, Escuela de Microbiología, Universidad de Antioquia, Medellín 050001, Colombia;
| | - Francisco J. Díaz
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Sara M. Robledo
- Grupo PECET, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (N.A.); (S.M.R.)
| | - Juan C. Hernandez
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050001, Colombia;
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680001, Colombia;
| |
Collapse
|
5
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
6
|
Notarbartolo S. T-Cell Immune Responses to SARS-CoV-2 Infection and Vaccination. Vaccines (Basel) 2024; 12:1126. [PMID: 39460293 PMCID: PMC11511197 DOI: 10.3390/vaccines12101126] [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: 09/04/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The innate and adaptive immune systems collaborate to detect SARS-CoV-2 infection, minimize the viral spread, and kill infected cells, ultimately leading to the resolution of the infection. The adaptive immune system develops a memory of previous encounters with the virus, providing enhanced responses when rechallenged by the same pathogen. Such immunological memory is the basis of vaccine function. Here, we review the current knowledge on the immune response to SARS-CoV-2 infection and vaccination, focusing on the pivotal role of T cells in establishing protective immunity against the virus. After providing an overview of the immune response to SARS-CoV-2 infection, we describe the main features of SARS-CoV-2-specific CD4+ and CD8+ T cells, including cross-reactive T cells, generated in patients with different degrees of COVID-19 severity, and of Spike-specific CD4+ and CD8+ T cells induced by vaccines. Finally, we discuss T-cell responses to SARS-CoV-2 variants and hybrid immunity and conclude by highlighting possible strategies to improve the efficacy of COVID-19 vaccination.
Collapse
Affiliation(s)
- Samuele Notarbartolo
- Infectious Diseases Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
7
|
Pan D, Choi JS, Martin CA, Nazareth J, Nellums LB, Pareek M. Quantifying the effect of vaccination on transmission in modelling studies. EClinicalMedicine 2024; 73:102669. [PMID: 38911839 PMCID: PMC11192782 DOI: 10.1016/j.eclinm.2024.102669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/29/2024] [Accepted: 05/14/2024] [Indexed: 06/25/2024] Open
Affiliation(s)
- Daniel Pan
- Development Centre for Population Health, University of Leicester, United Kingdom
- Department of Respiratory Sciences, University of Leicester, United Kingdom
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
- NIHR Leicester Biomedical Research Centre, United Kingdom
- Li Ka Shing Centre for Health Information and Discovery, Oxford Big Data Institute, University of Oxford, United Kingdom
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ji Soo Choi
- Development Centre for Population Health, University of Leicester, United Kingdom
- Department of Respiratory Sciences, University of Leicester, United Kingdom
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
- NIHR Leicester Biomedical Research Centre, United Kingdom
| | - Christopher A. Martin
- Development Centre for Population Health, University of Leicester, United Kingdom
- Department of Respiratory Sciences, University of Leicester, United Kingdom
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
- NIHR Leicester Biomedical Research Centre, United Kingdom
| | - Joshua Nazareth
- Development Centre for Population Health, University of Leicester, United Kingdom
- Department of Respiratory Sciences, University of Leicester, United Kingdom
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
- NIHR Leicester Biomedical Research Centre, United Kingdom
| | - Laura B. Nellums
- College of Population Health, University of New Mexico, Albuquerque, USA
| | - Manish Pareek
- Development Centre for Population Health, University of Leicester, United Kingdom
- Department of Respiratory Sciences, University of Leicester, United Kingdom
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
- NIHR Leicester Biomedical Research Centre, United Kingdom
| |
Collapse
|
8
|
Bartsch SM, O'Shea KJ, Strych U, Bottazzi ME, Lee BY. Response to: "Quantifying the effect of vaccination on transmission in modelling studies". EClinicalMedicine 2024; 73:102670. [PMID: 38911838 PMCID: PMC11192783 DOI: 10.1016/j.eclinm.2024.102670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/25/2024] Open
Affiliation(s)
- Sarah M. Bartsch
- Public Health Informatics, Computational, and Operations Research (PHICOR), CUNY Graduate School of Public Health and Health Policy, New York City, NY, USA
- Center for Advanced Technology and Communication in Health (CATCH), CUNY Graduate School of Public Health and Health Policy, New York City, NY, USA
| | - Kelly J. O'Shea
- Public Health Informatics, Computational, and Operations Research (PHICOR), CUNY Graduate School of Public Health and Health Policy, New York City, NY, USA
- Center for Advanced Technology and Communication in Health (CATCH), CUNY Graduate School of Public Health and Health Policy, New York City, NY, USA
| | - Ulrich Strych
- National School of Tropical Medicine, Department of Pediatrics, and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Maria Elena Bottazzi
- National School of Tropical Medicine, Department of Pediatrics, and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Bruce Y. Lee
- Public Health Informatics, Computational, and Operations Research (PHICOR), CUNY Graduate School of Public Health and Health Policy, New York City, NY, USA
- Center for Advanced Technology and Communication in Health (CATCH), CUNY Graduate School of Public Health and Health Policy, New York City, NY, USA
- Pandemic Response Institute, New York City, NY, USA
| |
Collapse
|
9
|
Lee BY, Pavilonis B, John DC, Heneghan J, Bartsch SM, Kavouras I. The Need to Focus More on Climate Change Communication and Incorporate More Systems Approaches. JOURNAL OF HEALTH COMMUNICATION 2024; 29:1-10. [PMID: 38831666 DOI: 10.1080/10810730.2024.2361566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Society is at an inflection point-both in terms of climate change and the amount of data and computational resources currently available. Climate change has been a catastrophe in slow motion with relationships between human activity, climate change, and the resulting effects forming a complex system. However, to date, there has been a general lack of urgent responses from leaders and the general public, despite urgent warnings from the scientific community about the consequences of climate change and what can be done to mitigate it. Further, misinformation and disinformation about climate change abound. A major problem is that there has not been enough focus on communication in the climate change field. Since communication itself involves complex systems (e.g. information users, information itself, communications channels), there is a need for more systems approaches to communication about climate change. Utilizing systems approaches to really understand and anticipate how information may be distributed and received before communication has even occurred and adjust accordingly can lead to more proactive precision climate change communication. The time has come to identify and develop more effective, tailored, and precise communication for climate change.
Collapse
Affiliation(s)
- Bruce Y Lee
- New York City Pandemic Response Institute (PRI), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Center for Advanced Technology and Communication in Health (CATCH), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Public Health Informatics, Computational, and Operations Research (PHICOR), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Artificial Intelligence, Modeling, and Informatics, for Nutrition Guidance and Systems (AIMINGS) Center, CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
| | - Brian Pavilonis
- New York City Pandemic Response Institute (PRI), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
| | - Danielle C John
- New York City Pandemic Response Institute (PRI), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Center for Advanced Technology and Communication in Health (CATCH), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Public Health Informatics, Computational, and Operations Research (PHICOR), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
| | - Jessie Heneghan
- Center for Advanced Technology and Communication in Health (CATCH), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Public Health Informatics, Computational, and Operations Research (PHICOR), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Artificial Intelligence, Modeling, and Informatics, for Nutrition Guidance and Systems (AIMINGS) Center, CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
| | - Sarah M Bartsch
- Center for Advanced Technology and Communication in Health (CATCH), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Public Health Informatics, Computational, and Operations Research (PHICOR), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
- Artificial Intelligence, Modeling, and Informatics, for Nutrition Guidance and Systems (AIMINGS) Center, CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
| | - Ilias Kavouras
- New York City Pandemic Response Institute (PRI), CUNY Graduate School of Public Health and Health Policy, New York City, New York, USA
| |
Collapse
|
10
|
Guan X, Verma AK, Wang G, Roy A, Perlman S, Du L. A Unique mRNA Vaccine Elicits Protective Efficacy against the SARS-CoV-2 Omicron Variant and SARS-CoV. Vaccines (Basel) 2024; 12:605. [PMID: 38932334 PMCID: PMC11209356 DOI: 10.3390/vaccines12060605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
The highly pathogenic coronaviruses SARS-CoV-2 and SARS-CoV have led to the COVID-19 pandemic and SARS outbreak, respectively. The receptor-binding domain (RBD) of the spike (S) protein of SARS-CoV-2, particularly the Omicron variant, has frequent mutations, resulting in the reduced efficiency of current COVID-19 vaccines against new variants. Here, we designed two lipid nanoparticle-encapsulated mRNA vaccines by deleting the mutant RBD of the SARS-CoV-2 Omicron variant (SARS2-S (RBD-del)) or by replacing this mutant RBD with the conserved and potent RBD of SARS-CoV (SARS2-S (SARS-RBD)). Both mRNA vaccines were stable at various temperatures for different time periods. Unlike SARS2-S (RBD-del) mRNA, SARS2-S (SARS-RBD) mRNA elicited effective T-cell responses and potent antibodies specific to both SARS-CoV-2 S and SARS-CoV RBD proteins. It induced strong neutralizing antibodies against pseudotyped SARS-CoV-2 and SARS-CoV infections and protected immunized mice from the challenge of the SARS-CoV-2 Omicron variant and SARS-CoV by significantly reducing the viral titers in the lungs after Omicron challenge and by completely preventing SARS-CoV-induced weight loss and death. SARS2-S (SARS-RBD)-immunized serum antibodies protected naïve mice from the SARS-CoV challenge, with its protective efficacy positively correlating with the neutralizing antibody titers. These findings indicate that this mRNA vaccine has the potential for development as an effective vaccine against current and future SARS-CoV-2 variants and SARS-CoV.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Abhishek K. Verma
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Gang Wang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Abhijeet Roy
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|