1
|
Chiang JY, Lin TH, Cheng JX, Pan WY. Polyethyleneimine/fucoidan polyplexes as vaccine carriers for enhanced antigen loading and dendritic cell activation. Int J Biol Macromol 2025; 306:141336. [PMID: 39986515 DOI: 10.1016/j.ijbiomac.2025.141336] [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: 09/26/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Vaccination is one of the most effective strategies for preventing infectious diseases. Recently, most research has centered on the development of protein subunit vaccines due to their safety. However, their low immunogenicity remains a challenge. Nanoparticle vaccines offer advantages by protecting proteins from degradation and acting as adjuvants to stimulate the immune system. Herein, a polyplexe (OVA@PEI/Fu) formed by the electrostatic interaction between positively charged polyethyleneimine (PEI) and negatively charged fucoidan was prepared for the encapsulation of a model antigen, ovalbumin (OVA). Experimental results revealed that the incorporation of fucoidan in the polyplexes not only enhanced OVA loading efficiency but also contributed adjuvant effects, significantly boosting dendritic cell activation and maturation in vitro compared to OVA@PEI polyplexes. In vivo experiments showed that the OVA@PEI/Fu can induce strong anti-OVA specific antibody responses, as well as OVA-specific CD4+ and CD8+ T cell responses. The carrier developed in the present study shows promise as a platform for protein-based subunit vaccines.
Collapse
Affiliation(s)
- Jian-Yi Chiang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Tsai-Hsuan Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Jia-Xiang Cheng
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Yu Pan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
2
|
Kim S, Flores RA, Moon SY, Lee SY, Altanzul B, Baek J, Choi EB, Lim H, Jang EY, Lee YK, Ouh IO, Kim WH. Design and Preliminary Immunogenicity Evaluation of Nipah Virus Glycoprotein G Epitope-Based Peptide Vaccine in Mice. Vaccines (Basel) 2025; 13:428. [PMID: 40333318 PMCID: PMC12031491 DOI: 10.3390/vaccines13040428] [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: 02/26/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
Background: The emergence of several paramyxoviruses, including Nipah virus (NiV), makes continued efforts in vaccine development as part of pandemic preparedness efforts necessary. Although NiV is a zoonotic pathogen with high case fatality, there is still no licensed vaccine. Methods: Herein, NiV attachment glycoprotein G (NiV-G), which is crucial to host cell receptor binding, was used to develop Nipah epitope-based peptide vaccines. A total of 39 B- and T-cell epitopes of NiV-G were shortlisted for peptide synthesis and evaluation using in silico analysis. Results: The in vitro antigenicity evaluation of the peptide candidates showed eight synthesized peptides (G7, stalk-domain epitopes) with relatively high binding to NiV-G antibody-positive serum (A450nm: 1.39-3.78). Moreover, nine-mer (9-mer) peptides were found to be less reactive than their longer peptide counterparts (15-30 aa, G7-1, and G7-4), but 9-mer activity was enhanced with cyclization (NPLPFREYK, A450nm: 2.66) and C-terminal amidation modification (NPLPFREYK-NH2, A450nm: 1.39). Subsequently, in vivo validation in immunized mice revealed the immunogenicity potential of the G7-1 peptide vaccine (30 aa, NENVNEKCKFTLPPLKIHECNISCPNPLPF) to elicit a strong antigen-specific antibody response against their homologous peptide antigen (I.V., A450nm: 1.48 ± 0.78; I.M., A450nm: 1.66 ± 0.66). However, antibody binding to recombinant NiV-G protein remained low, suggesting limited recognition to the native antigen. Conclusions: This study focused on the preliminary screening and validation of peptide vaccines using single formulations with minimal modifications in the peptide candidates. Our findings collectively show the immunogenic potential of the NiV-G stalk-based epitope peptide vaccine as a novel therapeutic for NiV and underscores the need for strategic design, delivery, and formulation optimization to enhance its protective efficacy and translational application.
Collapse
Affiliation(s)
- Seungyeon Kim
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Rochelle A. Flores
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| | - Seo Young Moon
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Seung Yun Lee
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| | - Bujinlkham Altanzul
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| | - Jiwon Baek
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Eun Bee Choi
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Heeji Lim
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Eun Young Jang
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Yoo-kyoung Lee
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - In-Ohk Ouh
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Woo H. Kim
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| |
Collapse
|
3
|
Bolu O, Alo OD, Iwara E, Longley AT, Hadley I, Ogar CK, Ezekwe C, Elemuwa U, Adedokun O, Ramadhani HO, Ohakanu S, Ortiz N, Antonza G, Abubakar A, Asekun A, Fraden B, Chen R, Nordenberg D, Adebajo S, Adeyeye MC, Stafford KA. Feasibility of cohort event monitoring and assessment of reactogenicity and adverse events among a cohort of AstraZeneca and Moderna COVID-19 vaccine recipients in Nigeria, 2021. Vaccine 2025; 52:126907. [PMID: 40073670 PMCID: PMC11975468 DOI: 10.1016/j.vaccine.2025.126907] [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: 04/10/2024] [Revised: 01/10/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND To generate COVID-19 vaccine safety data in Nigeria, passive reporting was supplemented with cohort event monitoring (CEM), an active surveillance system. We described reactogenicity within 7 days and adverse events up to 3 months after each AstraZeneca or Moderna COVID-19 vaccine dose while assessing the feasibility of implementing CEM in a low- to middle-income country (LMIC) during a mass vaccination campaign. METHODS Participants were aged ≥18 years with access to mobile phones who received the first dose of an authorized COVID-19 vaccine from participating health facilities in 6 states of Nigeria during September and October 2021. Data collectors interviewed participants via phone on days 0, 3, 7, and thereafter every 7 days for 3 months. The same schedule was restarted if a participant received a second vaccine dose. Proportions of participant-reported adverse events following COVID-19 vaccine receipt were calculated. Investigation and causality assessment were conducted on deaths using the World Health Organization causality guidelines. RESULTS We enrolled 12,317 participants (AstraZeneca 6990; Moderna 5327); 6167/6990 (88.2 %) AstraZeneca and 4879/5327 (91.6 %) Moderna recipients completed a follow-up interview days 0-7 after the first dose; among them, 2685/6167 (43.5 %) AstraZeneca and 3533/4879 (72.4 %) Moderna recipients reported local reactions and 2456/6167 (39.8 %) AstraZeneca and 2087/4879 (42.8 %) Moderna recipients reported systemic reactions. Overall, 3891/6990 (55.7 %) AstraZeneca and 3978/5327 (72.8 %) Moderna recipients received a second dose of COVID-19 vaccine, among whom 897/3891 (23 %) AstraZeneca and 1979/3978 (49.7 %) Moderna recipients reported local reactions and 727/3891 (18.7 %) AstraZeneca and 1680/3978 (42.2 %) Moderna recipients reported systemic reactions. Among all enrolled, 11 died; there was no evidence to suggest any deaths were vaccine-related. CONCLUSIONS No unexpected patterns of adverse events were detected, providing additional data on the safety of these COVID-19 vaccines in Nigerian adults. We demonstrated that implementing CEM was feasible and may be valuable for safety monitoring of vaccines introduced in LMICs.
Collapse
Affiliation(s)
- Omotayo Bolu
- Global Immunization Division, Global Health Center, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Oluwafemi D Alo
- Center for International Health, Education, and Biosecurity, Maryland Global Initiatives Corporation-an affiliate of the University of Maryland Baltimore, Abuja, Federal Capital Territory, Nigeria
| | - Emem Iwara
- Center for International Health, Education, and Biosecurity, Maryland Global Initiatives Corporation-an affiliate of the University of Maryland Baltimore, Abuja, Federal Capital Territory, Nigeria
| | - Ashley T Longley
- Global Immunization Division, Global Health Center, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Ikwe Hadley
- US Centers for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Comfort K Ogar
- The Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Chukwuka Ezekwe
- Center for International Health, Education, and Biosecurity, Maryland Global Initiatives Corporation-an affiliate of the University of Maryland Baltimore, Abuja, Federal Capital Territory, Nigeria
| | - Uchenna Elemuwa
- National Agency for Food and Drug Administration and Control, Abuja, Federal Capital Territory, Nigeria
| | - Oluwasanmi Adedokun
- Center for International Health, Education, and Biosecurity, Maryland Global Initiatives Corporation-an affiliate of the University of Maryland Baltimore, Abuja, Federal Capital Territory, Nigeria
| | - Habib O Ramadhani
- Center for International Health, Education, and Biosecurity, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephen Ohakanu
- Center for International Health, Education, and Biosecurity, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nancy Ortiz
- Global Immunization Division, Global Health Center, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Gladys Antonza
- Center for International Health, Education, and Biosecurity, Maryland Global Initiatives Corporation-an affiliate of the University of Maryland Baltimore, Abuja, Federal Capital Territory, Nigeria
| | - Asmau Abubakar
- National Agency for Food and Drug Administration and Control, Abuja, Federal Capital Territory, Nigeria
| | - Adeyelu Asekun
- US Centers for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Bitrus Fraden
- National Agency for Food and Drug Administration and Control, Abuja, Federal Capital Territory, Nigeria
| | - Robert Chen
- The Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Dale Nordenberg
- The Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Sylvia Adebajo
- Center for International Health, Education, and Biosecurity, Maryland Global Initiatives Corporation-an affiliate of the University of Maryland Baltimore, Abuja, Federal Capital Territory, Nigeria
| | - Moji Christianah Adeyeye
- National Agency for Food and Drug Administration and Control, Abuja, Federal Capital Territory, Nigeria
| | - Kristen A Stafford
- Center for International Health, Education, and Biosecurity, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Purushotham JN, Lutz HL, Parker E, Andersen KG. Immunological drivers of zoonotic virus emergence, evolution, and endemicity. Immunity 2025; 58:784-796. [PMID: 40168990 PMCID: PMC11981831 DOI: 10.1016/j.immuni.2025.03.014] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025]
Abstract
The disruption of natural ecosystems caused by climate change and human activity is amplifying the risk of zoonotic spillover, presenting a growing global health threat. In the past two decades, the emergence of multiple zoonotic viruses has exposed critical gaps in our ability to predict epidemic trajectories and implement effective interventions. RNA viruses, in particular, are challenging to control due to their high mutation rates and ability to adapt and evade immune defenses. To better prepare for future outbreaks, it is vital that we deepen our understanding of the factors driving viral emergence, transmission, and persistence in human populations. Specifically, deciphering the interactions between antibody-mediated immunity and viral evolution will be key. In this perspective, we explore these dynamic relationships and highlight research priorities that may guide the development of more effective strategies to mitigate the impact of emerging infectious diseases.
Collapse
Affiliation(s)
- Jyothi N Purushotham
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - Holly L Lutz
- Denver Museum of Nature and Science, Denver, CO, USA
| | - Edyth Parker
- The Institute of Genomics and Global Health (IGH), Redeemer's University, Ede, Osun, Nigeria
| | - Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA.
| |
Collapse
|
5
|
Chaudhuri A, Bandyopadhyay B, Mondal B, Sarkar A, Ghosh S, Panja AS. Exploration of potential inhibitors against chikungunya envelope: an in-silico clue. In Silico Pharmacol 2025; 13:55. [PMID: 40207105 PMCID: PMC11977042 DOI: 10.1007/s40203-025-00351-3] [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: 02/05/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne virus which causes chikungunya disease. Two biological vectors Aedes aegypti and Aedes albopictus transmit CHIKV to the victim body. According to the report of the European Centre for Disease Prevention and Control, epidemics of chikungunya disease existed in 2024 over America, Africa, Europe and Asia. Although 50% CHIKV infected person show chronic clinical symptoms and several troubles associated with chikungunya, still there are no effective vaccines or medications on market. So availability of another CHIKV inhibiting materials and mechanisms are necessary. For this purpose recently plant-derived bioactive compounds with antiviral properties are used to inhibit chikungunya infection. In this present research work 69 CHIKV inhibiting active compounds were chosen for ADMET analysis. Drug likeness of active compounds was also analyzed based on Lipinski's rule of five. Based on the drug likeness, active compounds (Baicalein, Epicatechin, Genistein, Quercetin, Resveratrol) were finally screened for molecular docking with CHIKV envelope proteins using Auto Dock program. Among the five active compounds, Genistein showed highest binding energy for both E1 (ΔG = - 8.3 kcal/mol) and E2 (ΔG = - 7.1 kcal/mol). Molecular dynamics simulations signify that Genistein forms a stable complex with the CHIKV E1 and E2 proteins over a 50 ns period with a significant number of hydrogen bonds. So this present study concluded that Genistein will act as potent CHIKV E1 and E2 inhibiting active compounds. To evaluate efficiency or inhibiting capacity of finally selected Genistein against CHIKV, in vivo and in vitro validation should be conducted. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-025-00351-3.
Collapse
Affiliation(s)
- Aparna Chaudhuri
- Department of Biotechnology School of Life Science , Swami Vivekananda University , Barrackpore, West Bengal India
| | - Bidyut Bandyopadhyay
- Department of Biotechnology Molecular informatics Laboratory , Oriental Institute of Science and Technology Vidyasagar University , Midnapore, West Bengal India
| | - Buddhadev Mondal
- Department of Zoology , Burdwan Raj College , Purba Bardhaman, West Bengal India
| | - Aniket Sarkar
- Department of Biotechnology Molecular informatics Laboratory , Oriental Institute of Science and Technology Vidyasagar University , Midnapore, West Bengal India
| | - Sabyasachi Ghosh
- Department of Biotechnology School of Life Science , Swami Vivekananda University , Barrackpore, West Bengal India
| | - Anindya Sundar Panja
- Department of Biotechnology Molecular informatics Laboratory , Oriental Institute of Science and Technology Vidyasagar University , Midnapore, West Bengal India
| |
Collapse
|
6
|
Pan Y, Liu Z, Lu J, Quan J, Zhao G, Song G, Wang J, Ren Z. Astragalus polysaccharide inhibits infectious hematopoietic necrosis virus damage to rainbow trout (Oncorhynchus mykiss) spleen by promoting the efficacy of inactivated vaccine. FISH & SHELLFISH IMMUNOLOGY 2025; 159:110180. [PMID: 39923886 DOI: 10.1016/j.fsi.2025.110180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
Pandemic infectious hematopoietic necrosis (IHN) caused by severe acute IHN virus (IHNV) threatens rainbow trout aquaculture in China. Therefore, it is crucial to develop effective vaccines and elucidate their mechanisms of action. Here, we revealed the mechanism of immune response of Astragalus polysaccharide (APS) combined with IHNV inactivated vaccine on rainbow trout spleen by serum enzyme activity assay, histopathological analysis, RNA-seq and viral load measurement. Histopathology and TUNEL showed that the APS combination vaccine significantly inhibited spleen damage and apoptosis by IHNV. Also, APS increased serum SOD, CAT, T-AOC, AKP and ACP activities by enhancing vaccine efficacy. Transcriptome analysis of the spleen showed that immune-related pathways were significantly enriched in the APS + vaccine group. The PPI network identified hub genes including IgM, IRF7, IgT, IgD, TLR7, CD4, CD8, IL-1β, and the APS + vaccine induced expression of these genes in the spleen. Notably, the APS combination vaccine significantly inhibited IHNV replication in the spleen. Overall, APS enhanced IHNV inactivated vaccine efficacy through stronger immune stimulation. These results indicate that the combined use of inactivated vaccine and APS can stimulate strong immunity in rainbow trout spleen, which provides valuable reference data to study the mechanism of aquatic vaccine immunoprophylaxis.
Collapse
Affiliation(s)
- Yucai Pan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhe Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Junhao Lu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jinqiang Quan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Guiyan Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Guolin Song
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jianfu Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhuowei Ren
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| |
Collapse
|
7
|
Nguyen VH, Crépey P, Williams BA, Welch VL, Pivette JM, Jones CH, True JM. Modeling the impact of early vaccination in an influenza pandemic in the United States. NPJ Vaccines 2025; 10:62. [PMID: 40157953 PMCID: PMC11954890 DOI: 10.1038/s41541-025-01081-5] [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: 01/26/2024] [Accepted: 01/30/2025] [Indexed: 04/01/2025] Open
Abstract
We modeled the impact of initiating one-dose influenza vaccination at 3 months vs 6 months after declaration of a pandemic over a 1-year timeframe in the US population. Three vaccine effectiveness (VE) and two pandemic severity levels were considered, using an epidemic curve based on typical seasonal influenza epidemics. Vaccination from 3 months with a high, moderate, or low effectiveness vaccine would prevent ~95%, 84%, or 38% deaths post-vaccination, respectively, compared with 21%, 18%, and 8%, respectively following vaccination at 6 months, irrespective of pandemic severity. While the pandemic curve would not be flattened from vaccination from 6 months, a moderate/high effectiveness vaccine could flatten the curve if administered from 3 months. Overall, speed of initiating a vaccination campaign is more important than VE in reducing the health impacts of an influenza pandemic. Preparedness strategies may be able to minimize future pandemic impacts by prioritizing rapid vaccine roll-out.
Collapse
Affiliation(s)
| | - Pascal Crépey
- EHESP, University of Rennes, CNRS, IEP Rennes, Arènes-UMR 6051, RSMS-Inserm U 1309, Rennes, France
| | | | | | | | | | | |
Collapse
|
8
|
Ceruti A, Kobialka RM, Abd El Wahed A, Truyen U. African Swine Fever: A One Health Perspective and Global Challenges. Animals (Basel) 2025; 15:928. [PMID: 40218322 PMCID: PMC11987808 DOI: 10.3390/ani15070928] [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: 02/20/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
African Swine Fever is non-zoonotic viral disease affecting pigs worldwide, with severe implications for many sectors such as national economies and pig populations. The disease control strategies are variable across the globe and its implementation and outcome depend on many factors. One Health describes the collaborative effort across disciplines to address public health challenges involving human, animal, and environmental aspects. African Swine Fever is not commonly understood under the concept of One Health. However, the disease has a great impact on all One Health interfaces. This paper reviews and discusses how the vast impact of ASF, a non-zoonotic disease, still demands a holistic perspective to address global challenges and opportunities to control the disease.
Collapse
Affiliation(s)
- Arianna Ceruti
- Institute of Animal Hygiene and Veterinary Public Health, Leipzig University, 04103 Leipzig, Germany; (R.M.K.); (A.A.E.W.); (U.T.)
| | | | | | | |
Collapse
|
9
|
Wei Z, Zhang S, Wang X, Xue Y, Dang S, Zhai J. Technological breakthroughs and advancements in the application of mRNA vaccines: a comprehensive exploration and future prospects. Front Immunol 2025; 16:1524317. [PMID: 40103818 PMCID: PMC11913674 DOI: 10.3389/fimmu.2025.1524317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
mRNA vaccines utilize single-stranded linear DNA as a template for in vitro transcription. The mRNA is introduced into the cytoplasm via the corresponding delivery system to express the target protein, which then performs its relevant biological function. mRNA vaccines are beneficial in various fields, including cancer vaccines, infectious disease vaccines, protein replacement therapy, and treatment of rare diseases. They offer advantages such as a simple manufacturing process, a quick development cycle, and ease of industrialization. Additionally, mRNA vaccines afford flexibility in adjusting antigen designs and combining sequences of multiple variants, thereby addressing the issue of frequent mutations in pathogenic microorganisms. This paper aims to provide an extensive review of the global development and current research status of mRNA vaccines, with a focus on immunogenicity, classification, design, delivery vector development, stability, and biomedical application. Moreover, the study highlights current challenges and offers insights into future directions for development.
Collapse
Affiliation(s)
- Zhimeng Wei
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
- Keerqin District First People’s Hospital, Tongliao, China
| | - Shuai Zhang
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
| | - Xingya Wang
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
| | - Ying Xue
- Keerqin District First People’s Hospital, Tongliao, China
| | - Sheng Dang
- Keerqin District First People’s Hospital, Tongliao, China
| | - Jingbo Zhai
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
- Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China
| |
Collapse
|
10
|
Pan Y, Zhao H, Huang W, Liu S, Qi Y, Huang Y. Metal-Protein Hybrid Materials: Unlocking New Frontiers in Biomedical Applications. Adv Healthc Mater 2025; 14:e2404405. [PMID: 39778029 DOI: 10.1002/adhm.202404405] [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: 11/07/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Metal-protein hybrid materials represent a novel class of functional materials that exhibit exceptional physicochemical properties and tunable structures, rendering them remarkable applications in diverse fields, including materials engineering, biocatalysis, biosensing, and biomedicine. The design and development of multifunctional and biocompatible metal-protein hybrid materials have been the subject of extensive research and a key aspiration for practical applications in clinical settings. This review provides a comprehensive analysis of the design strategies, intrinsic properties, and biomedical applications of these hybrid materials, with a specific emphasis on their potential in cancer therapy, drug and vaccine delivery, antibacterial treatments, and tissue regeneration. Through rational design, stable metal-protein hybrid materials can be synthesized using straightforward methods, enabling them with therapeutic, delivery, immunomodulatory, and other desired functionalities. Finally, the review outlines the existing limitations and challenges associated with metal-protein hybrid materials and evaluates their potential for clinical translation, providing insights into their practical implementation within biomedical applications.
Collapse
Affiliation(s)
- Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Han Zhao
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Siyang Liu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| |
Collapse
|
11
|
Vo DK, Trinh KTL. Molecular Farming for Immunization: Current Advances and Future Prospects in Plant-Produced Vaccines. Vaccines (Basel) 2025; 13:191. [PMID: 40006737 PMCID: PMC11860421 DOI: 10.3390/vaccines13020191] [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: 01/10/2025] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Using plants as bioreactors, molecular farming has emerged as a versatile and sustainable platform for producing recombinant vaccines, therapeutic proteins, industrial enzymes, and nutraceuticals. This innovative approach leverages the unique advantages of plants, including scalability, cost-effectiveness, and reduced risk of contamination with human pathogens. Recent advancements in gene editing, transient expression systems, and nanoparticle-based delivery technologies have significantly enhanced the efficiency and versatility of plant-based systems. Particularly in vaccine development, molecular farming has demonstrated its potential with notable successes such as Medicago's Covifenz for COVID-19, illustrating the capacity of plant-based platforms to address global health emergencies rapidly. Furthermore, edible vaccines have opened new avenues in the delivery of vaccines, mainly in settings with low resources where the cold chain used for conventional logistics is a challenge. However, optimization of protein yield and stability, the complexity of purification processes, and regulatory hurdles are some of the challenges that still remain. This review discusses the current status of vaccine development using plant-based expression systems, operational mechanisms for plant expression platforms, major applications in the prevention of infectious diseases, and new developments, such as nanoparticle-mediated delivery and cancer vaccines. The discussion will also touch on ethical considerations, the regulatory framework, and future trends with respect to the transformative capacity of plant-derived vaccines in ensuring greater global accessibility and cost-effectiveness of the vaccination. This field holds great promise for the infectious disease area and, indeed, for applications in personalized medicine and biopharmaceuticals in the near future.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kieu The Loan Trinh
- Bionano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
12
|
Alqahtani SM. Discovering broad-spectrum inhibitors for SARS-CoV-2 variants: a cheminformatics and biophysical approach targeting the main protease. Front Pharmacol 2025; 16:1459581. [PMID: 39974733 PMCID: PMC11835822 DOI: 10.3389/fphar.2025.1459581] [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: 07/04/2024] [Accepted: 01/10/2025] [Indexed: 02/21/2025] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 still lacks effective antiviral drugs. Therefore, a thorough receptor-based virtual screening study was conducted to screen different natural and synthetic drug libraries, such as the Asinex Antiviral, Seaweed Metabolite Database, Medicinal Fungi Secondary Metabolite and Therapeutics Library, and Comprehensive Marine Natural Products Database comprising 6,827, 1,191, 1,830, and 45,000 compounds, respectively, against the main protease enzyme of SARS-CoV-2. Accordingly, three drug molecules (BBB-26580140, BDE-32007849, and LAS-51378804) are highlighted as the best binding molecules to the main protease S1 pocket. The docking binding energy scores of BBB-26580140, BDE-32007849, and LAS-51378804 were -13.02, -13.0, and -12.56 kcal/mol, respectively. Compared to the control Z1741970824 molecule with a binding energy score of -11.59 kcal/mol, the lead structures identified herein showed robust hydrophilic and van der Waals interactions with the enzyme active site residues, such as His41 and Cys145, and achieved highly stable binding modes. The simulations showed a stable structure of the main protease enzyme with the shortlisted leads in the pocket, and the network of binding interactions remained intact during the simulations. The overall molecular mechanics with generalized Born and surface area solvation binding energies of the BBB-26580140, BDE-32007849, LAS-51378804, and control molecules are -53.02, -56.85, -55.44, and -48.91 kcal/mol, respectively. Similarly, the net molecular mechanics Poisson-Boltzmann surface area binding energies of BBB-26580140, BDE-32007849, LAS-51378804, and control are -53.6, -57.61, -54.41, and -50.09 kcal/mol, respectively. The binding entropy energies of these systems showed lower free energies, indicating their stable nature. Furthermore, the binding energies were revalidated using the water swap method that considers the role of the water molecules in bridging the ligands to the enzyme active site residues. The compounds also revealed good ADMET properties and followed all major rules of drug-likeness. Thus, these compounds are predicted as promising leads and can be subjected to further experimental studies for evaluation of their biological activities.
Collapse
Affiliation(s)
- Safar M. Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| |
Collapse
|
13
|
Wang Z, Lin Z, Mei X, Cai L, Lin KC, Rodríguez JF, Ye Z, Parraguez XS, Guajardo EM, García Luna PC, Zhang JYJ, Zhang YS. Engineered Living Systems Based on Gelatin: Design, Manufacturing, and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416260. [PMID: 39910847 DOI: 10.1002/adma.202416260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/26/2024] [Indexed: 02/07/2025]
Abstract
Engineered living systems (ELSs) represent purpose-driven assemblies of living components, encompassing cells, biomaterials, and active agents, intricately designed to fulfill diverse biomedical applications. Gelatin and its derivatives have been used extensively in ELSs owing to their mature translational pathways, favorable biological properties, and adjustable physicochemical characteristics. This review explores the intersection of gelatin and its derivatives with fabrication techniques, offering a comprehensive examination of their synergistic potential in creating ELSs for various applications in biomedicine. It offers a deep dive into gelatin, including its structures and production, sources, processing, and properties. Additionally, the review explores various fabrication techniques employing gelatin and its derivatives, including generic fabrication techniques, microfluidics, and various 3D printing methods. Furthermore, it discusses the applications of ELSs based on gelatin in regenerative engineering as well as in cell therapies, bioadhesives, biorobots, and biosensors. Future directions and challenges in gelatin fabrication are also examined, highlighting emerging trends and potential areas for improvements and innovations. In summary, this comprehensive review underscores the significance of gelatin-based ELSs in advancing biomedical engineering and lays the groundwork for guiding future research and developments within the field.
Collapse
Affiliation(s)
- Zhenwu Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Zeng Lin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ling Cai
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ko-Chih Lin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Jimena Flores Rodríguez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Zixin Ye
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ximena Salazar Parraguez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Emilio Mireles Guajardo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Pedro Cortés García Luna
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Jun Yi Joey Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| |
Collapse
|
14
|
Zúñiga E, Contreras-Trigo B, Buchert J, Sáez-Ahumada F, Hernández L, Fica-León V, Nova-Lamperti E, Kobe B, Guzmán F, Diaz-García V, Guzmán-Gutiérrez E, Oyarzún P. Gold Nanoparticles as a Platform for Delivery of Immunogenic Peptides to THP-1 Derived Macrophages: Insights into Nanotoxicity. Vaccines (Basel) 2025; 13:119. [PMID: 40006666 PMCID: PMC11860437 DOI: 10.3390/vaccines13020119] [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: 12/14/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Peptide-based nanovaccines have emerged as a promising strategy for combating infectious diseases, as they overcome the low immunogenicity that is inherent to short epitope-containing synthetic peptides. Gold nanoparticles (AuNPs) present several advantages as peptide nanocarriers, but a deeper understanding of the design criteria is paramount to accelerate the development of peptide-AuNPs nanoconjugates (p-AuNPs). METHODS Herein, we synthesized and characterized p-AuNPs of 23 nm (p-Au23) and 68 nm (p-Au68) with varying levels of peptide surface coverage and different peptide designs, investigating their effect on the cell viability (cell death and mitochondrial activity), cellular uptake, and cathepsin B activity in THP-1 macrophages. RESULTS p-Au23 proved no negative effect in the cell viability and high levels of nanoconjugate uptake, but p-Au68 induced strong toxicity to the cell line. The peptide sequences were successfully designed with spacer regions and a cell-penetrating peptide (pTAT) that enhanced cellular uptake and cathepsin B activity for p-Au23, while pTAT induced severe effects in the THP-1 viability (~40-60% cell death). CONCLUSIONS These findings provide valuable insight into the design criteria of AuNPs and immunogenic peptides, along with nanotoxicity effects associated with AuNP size and surface charge in human monocyte-derived macrophages.
Collapse
Affiliation(s)
- Eduardo Zúñiga
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Braulio Contreras-Trigo
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Jorge Buchert
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Fabián Sáez-Ahumada
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Leonardo Hernández
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Víctor Fica-León
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Estefania Nova-Lamperti
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Fanny Guzmán
- Núcleo Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Valparaíso 2373223, Chile;
| | - Víctor Diaz-García
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Enrique Guzmán-Gutiérrez
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Patricio Oyarzún
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| |
Collapse
|
15
|
Jeon J, Kim E. Exploring Future Pandemic Preparedness Through the Development of Preventive Vaccine Platforms and the Key Roles of International Organizations in a Global Health Crisis. Vaccines (Basel) 2025; 13:56. [PMID: 39852835 PMCID: PMC11768803 DOI: 10.3390/vaccines13010056] [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: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Background: The emergence of more than 40 new infectious diseases since the 1980s has emerged as a serious global health concern, many of which are zoonotic. In response, many international organizations, including the US Centers for Disease Control and Prevention (CDC), the World Health Organization (WHO), and the European Center for Disease Prevention and Control (ECDC), have developed strategies to combat these health threats. The need for rapid vaccine development has been highlighted by Coronavirus disease 2019 (COVID-19), and mRNA technology has shown promise as a platform. While the acceleration of vaccine development has been successful, concerns have been raised about the technical limits, safety, supply, and distribution of vaccines. Objective: This study analyzes the status of vaccine platform development in global pandemics and explores ways to respond to future pandemic crises through an overview of the roles of international organizations and their support programs. It examines the key roles and partnerships of international organizations such as the World Health Organization (WHO), vaccine research and development expertise of the Coalition for Epidemic Preparedness Innovations (CEPI), control of the vaccine supply chain and distribution by the Global Alliance for Vaccines and Immunization (GAVI), and technology transfer capabilities of the International Vaccine Institute (IVI) in supporting the development, production, and supply of vaccine platform technologies for pandemic priority diseases announced by WHO and CEPI and analyzes their vaccine support programs and policies to identify effective ways to rapidly respond to future pandemics caused by emerging infectious diseases. Methods: This study focused on vaccine platform technology and the key roles of international organizations in the pandemic crisis. Literature data on vaccine platform development was collected, compared, and analyzed through national and international literature data search sites, referring to articles, journals, research reports, publications, books, guidelines, clinical trial data, and related reports. In addition, the websites of international vaccine support organizations, such as WHO, CEPI, GAVI, and IVI, were used to examine vaccine support projects, initiatives, and collaborations through literature reviews and case study methods. Results: The COVID-19 pandemic brought focus on the necessity for developing innovative vaccine platforms. Despite initial concerns, the swift integration of cutting-edge development technologies, mass production capabilities, and global collaboration have made messenger RNA (mRNA) vaccines a game-changing technology. As a result of the successful application of novel vaccine platforms, it is important to address the remaining challenges, including technical limits, safety concerns, and equitable global distribution. To achieve this, it is essential to review the regulatory, policy, and support initiatives that have been implemented in response to the COVID-19 pandemic, with particular emphasis on the key stages of vaccine development, production, and distribution, to prepare for future pandemics. An analysis of the status of vaccine development for priority pandemic diseases implies the need for balanced vaccine platform development. Also, international organizations such as WHO, CEPI, GAVI, and IVI play key roles in pandemic preparedness and the development and distribution of preventive vaccines. These organizations collaborated to improve accessibility to vaccines, strengthen the global response to infectious diseases, and address global health issues. The COVID-19 pandemic response demonstrates how the synergistic collaboration of WHO's standardized guidelines, CEPI's vaccine research and development expertise, GAVI's control of the vaccine supply chain and distribution, and IVI's technology transfer capabilities can be united to create a successful process for vaccine development and distribution. Conclusions: In preparation for future pandemics, a balanced vaccine platform development is essential. It should include a balanced investment in both novel technologies such as mRNA and viral vector-based vaccines and traditional platforms. The goal is to develop vaccine platform technologies that can be applied to emerging infectious diseases efficiently and increase manufacturing and distribution capabilities for future pandemics. Moreover, international vaccine support organizations should play key roles in setting the direction of global networking and preparing for international vaccine support programs to address the limitations of previous pandemic responses. As a result, by transforming future pandemic threats from unpredictable crises to surmountable challenges, it is expected to strengthen global health systems and reduce the social and economic burden of emerging infectious diseases in the long term.
Collapse
Affiliation(s)
- Jihee Jeon
- Pharmaceutical Regulatory Affairs, Department of Pharmaceutical Industry, Graduate School, Chung-Ang University, Seoul 06974, Republic of Korea;
| | - Eunyoung Kim
- Pharmaceutical Regulatory Affairs, Department of Pharmaceutical Industry, Graduate School, Chung-Ang University, Seoul 06974, Republic of Korea;
- Central Research Center of Epigenome Based Platform and Its Application for Drug Development, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
- Data Science, Evidence-Based and Clinical Research Laboratory, Department of Health, Social, and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
- Regulatory Science Policy, Pharmaceutical Regulatory Sciences, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
16
|
Bello MB, Alsaadi A, Naeem A, Almahboub SA, Bosaeed M, Aljedani SS. Development of nucleic acid-based vaccines against dengue and other mosquito-borne flaviviruses: the past, present, and future. Front Immunol 2025; 15:1475886. [PMID: 39840044 PMCID: PMC11747009 DOI: 10.3389/fimmu.2024.1475886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/06/2024] [Indexed: 01/23/2025] Open
Abstract
Due to their widespread geographic distribution and frequent outbreaks, mosquito-borne flaviviruses, such as DENV (DENV), Zika virus (ZIKV), Japanese encephalitis virus (JEV), yellow fever virus (YFV), and West Nile virus (WNV), are considered significant global public health threats and contribute to dramatic socioeconomic imbalances worldwide. The global prevalence of these viruses is largely driven by extensive international travels and ecological disruptions that create favorable conditions for the breeding of Aedes and Culex species, the mosquito vectors responsible for the spread of these pathogens. Currently, vaccines are available for only DENV, YFV, and JEV, but these face several challenges, including safety concerns, lengthy production processes, and logistical difficulties in distribution, especially in resource-limited regions, highlighting the urgent need for innovative vaccine approaches. Nucleic acid-based platforms, including DNA and mRNA vaccines, have emerged as promising alternatives due to their ability to elicit strong immune responses, facilitate rapid development, and support scalable manufacturing. This review provides a comprehensive update on the progress of DNA and mRNA vaccine development against mosquito-borne flaviviruses, detailing early efforts and current strategies that have produced candidates with remarkable protective efficacy and strong immunogenicity in preclinical models. Furthermore, we explore future directions for advancing nucleic acid vaccine candidates, which hold transformative potential for enhancing global public health.
Collapse
Affiliation(s)
- Muhammad Bashir Bello
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Ahlam Alsaadi
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Asif Naeem
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Sarah A. Almahboub
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammad Bosaeed
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Safia S. Aljedani
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Shen Z, Li C, Song W, Liu L, Kong Y, Huang A, Bao Q, Ying T, Wu Y. Enhancing vaccine half-life as a novel strategy for improving immune response durability of subunit vaccines. PLoS Pathog 2025; 21:e1012845. [PMID: 39778011 PMCID: PMC11750101 DOI: 10.1371/journal.ppat.1012845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/21/2025] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Vaccines are widely regarded as one of the most effective strategies for combating infectious diseases. However, significant challenges remain, such as insufficient antibody levels, limited protection against rapidly evolving variants, and poor immune durability, particularly in subunit vaccines, likely due to their short in vivo exposure. Recent advances in extending the half-life of protein therapeutics have shown promise in improving drug efficacy, yet whether increasing in vivo persistence can enhance the efficacy of subunit vaccines remains underexplored. In this study, we developed two trimeric SARS-CoV-2 subunit vaccines with distinct pharmacokinetic profiles to evaluate the impact of vaccine persistence on immune efficacy. A self-assembling trimeric subunit vaccine (RBD-HR/trimer) was designed, followed by an extended-persistence variant (RBD-sFc-HR/trimer) incorporating a soluble monomeric IgG1 fragment crystallizable. We demonstrated that RBD-sFc-HR/trimer elicited more robust and higher levels of neutralizing antibodies, with potent and broad neutralization activity against multiple SARS-CoV-2 variants. Notably, RBD-sFc-HR/trimer induced a durable immune response, significantly increasing the number of memory B cells and T cells. This study provides critical insights for designing vaccines that achieve potent and long-lasting immune responses against infectious diseases.
Collapse
Affiliation(s)
- Zhaoling Shen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Cheng Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenping Song
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Litong Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yu Kong
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ailing Huang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Qingui Bao
- Fosun Diagnostics (Shanghai) Co., Ltd, Shanghai, China
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, China
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, China
| |
Collapse
|
18
|
Pinyon JL, von Jonquieres G, Mow SL, Abed AA, Lai K, Manoharan M, Crawford EN, Xue SH, Smith‐Moore S, Caproni LJ, Milsom S, Klugmann M, Lovell NH, Housley GD. Vector-Free Deep Tissue Targeting of DNA/RNA Therapeutics via Single Capacitive Discharge Conductivity-Clamped Gene Electrotransfer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406545. [PMID: 39601152 PMCID: PMC11744645 DOI: 10.1002/advs.202406545] [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: 06/13/2024] [Revised: 09/10/2024] [Indexed: 11/29/2024]
Abstract
Viral vector and lipid nanoparticle based gene delivery have limitations around spatiotemporal control, transgene packaging size, and vector immune reactivity, compromising translation of nucleic acid (NA) therapeutics. In the emerging field of DNA and particularly RNA-based gene therapies, vector-free delivery platforms are identified as a key unmet need. Here, this work addresses these challenges through gene electrotransfer (GET) of "naked" polyanionic DNA/mRNA using a single needle form-factor which supports "electro-lens" based compression of the local electric field, and local control of tissue conductivity, enabling single capacitive discharge minimal charge gene delivery. Proof-of-concept studies for "single capacitive discharge conductivity-clamped gene electrotransfer" (SCD-CC-GET) deep tissue delivery of naked DNA and mRNA in the mouse hindlimb skeletal muscle achieve stable (>18 month) expression of luciferase reporter synthetic DNA, and mRNA encoding the reporter yield rapid onset (<3 h) high transient expression for several weeks. Delivery of DNAs encoding secreted alkaline phosphatase and Cal/09 influenza virus hemagglutinin antigen generate high systemic circulating recombinant protein levels and antibody titres. The findings support adoption of SCD-CC-GET for vaccines and immunotherapies, and extend the utility of this technology to meet the demand for efficient vector-free, precision, deep tissue delivery of NA therapeutics.
Collapse
Affiliation(s)
- Jeremy L. Pinyon
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
- Charles Perkins CentreSchool of Medical SciencesFaculty of Medicine and HealthUniversity of SydneyCamperdownNSW2006Australia
| | - Georg von Jonquieres
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Stephen L. Mow
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Amr Al Abed
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Keng‐Yin Lai
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Mathumathi Manoharan
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Edward N. Crawford
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Stanley H. Xue
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | | | | | - Sarah Milsom
- Touchlight Genetics LtdLower Sunbury RoadHamptonUKTW12 2ER
| | - Matthias Klugmann
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Nigel H. Lovell
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Gary D. Housley
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| |
Collapse
|
19
|
Im SH, Chung Y, Duskunovic N, Choi H, Park SH, Chung HJ. Oligonucleotide-Linked Lipid Nanoparticles as a Versatile mRNA Nanovaccine Platform. Adv Healthc Mater 2024; 13:e2401868. [PMID: 39363681 DOI: 10.1002/adhm.202401868] [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: 06/21/2024] [Revised: 09/07/2024] [Indexed: 10/05/2024]
Abstract
An effective delivery platform is crucial for the development of mRNA vaccines and therapeutics. Here, a versatile platform utilizing cholesterol-modified oligonucleotides (L-oligo) that bind to the mRNA within lipid nanoparticles (LNP), and enables the effective delivery of the mRNA into target cells is introduced. mRNA incorporated into LNPs via linkage with L-oligo, termed oligonucleotide-linked LNP (lnLNP), is superior in cellular uptake and transfection efficiency in target cells in vitro and in vivo, compared to the conventional LNP formulations. It is further applied lnLNP as an mRNA vaccine platform for SARS-CoV-2, demonstrating robust induction of neutralizing activity as well as polyfunctional SARS-CoV-2-specific T-cell response in vivo. The current strategy can be versatilely applied to different LNP platforms, for vaccine and therapeutic applications against various diseases, such as infections and cancers.
Collapse
Affiliation(s)
- San Hae Im
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Youseung Chung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Nevena Duskunovic
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Heewon Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Hyun Jung Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| |
Collapse
|
20
|
Liu CH, Ling J, Liu C, Schrader K, Ammigan R, Mclntire E. Vaccination rates among international students: Insights from a university health vaccination initiative. JOURNAL OF AMERICAN COLLEGE HEALTH : J OF ACH 2024; 72:3322-3329. [PMID: 36595642 DOI: 10.1080/07448481.2022.2155470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/03/2022] [Accepted: 10/23/2022] [Indexed: 06/17/2023]
Abstract
Objective: To examine the effects of a university's health vaccination initiative in increasing vaccination rates among international students/scholars in the United States. Methods: The vaccination initiative included: increasing vaccination opportunities by holding a pre-registration event, providing vaccine recommendations from healthcare professionals including a bilingual health interpreter, implementing campus-based marketing strategies, sending reminders using social media, and offering free and affordable vaccines. Results: Total 575 international students/scholars attended from 2016 to 2019 (N = 118, 163, 193, and 101, respectively), showing an increase compared to 2015. The most common vaccines administered were for influenza, human papillomavirus (HPV), tetanus, diphtheria, and acellular pertussis (Tdap), and Hepatitis A. Slightly less than one-quarter of participants received three or more vaccines. More women than men received HPV vaccine. Participants shared they would not have been vaccinated without this initiative and wished for more vaccination events. Conclusions: Future efforts are needed to implement this initiative across universities to further evaluate its effectiveness.
Collapse
Affiliation(s)
| | - Jiying Ling
- Michigan State University College of Nursing, East Lansing, Michigan, USA
| | - Charles Liu
- Michigan State University Neighborhood Students Success Collaborative, East Lansing, Michigan, USA
| | - Kara Schrader
- Michigan State University College of Nursing, East Lansing, Michigan, USA
| | | | - Emily Mclntire
- Michigan State University College of Nursing, East Lansing, Michigan, USA
| |
Collapse
|
21
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
22
|
Wei Y, Qiu T, Ai Y, Zhang Y, Xie J, Zhang D, Luo X, Sun X, Wang X, Qiu J. Advances of computational methods enhance the development of multi-epitope vaccines. Brief Bioinform 2024; 26:bbaf055. [PMID: 39951549 PMCID: PMC11827616 DOI: 10.1093/bib/bbaf055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/28/2024] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Vaccine development is one of the most promising fields, and multi-epitope vaccine, which does not need laborious culture processes, is an attractive alternative to classical vaccines with the advantage of safety, and efficiency. The rapid development of algorithms and the accumulation of immune data have facilitated the advancement of computer-aided vaccine design. Here we systemically reviewed the in silico data and algorithms resource, for different steps of computational vaccine design, including immunogen selection, epitope prediction, vaccine construction, optimization, and evaluation. The performance of different available tools on epitope prediction and immunogenicity evaluation was tested and compared on benchmark datasets. Finally, we discuss the future research direction for the construction of a multiepitope vaccine.
Collapse
Affiliation(s)
- Yiwen Wei
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Tianyi Qiu
- Institute of Clinical Science, Zhongshan Hospital; Intelligent Medicine Institute; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Medical College, Fudan University, No. 180, Fenglin Road, Xuhui Destrict, Shanghai 200032, China
| | - Yisi Ai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Yuxi Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Junting Xie
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Dong Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Xiaochuan Luo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Xiulan Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Foods, Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214122, China
| | - Xin Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Jingxuan Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| |
Collapse
|
23
|
Pan Y, Liu Z, Quan J, Gu W, Wang J, Zhao G, Lu J, Wang J. Purified Astragalus Polysaccharide Combined with Inactivated Vaccine Markedly Prevents Infectious Haematopoietic Necrosis Virus Infection in Rainbow Trout ( Oncorhynchus mykiss). ACS Biomater Sci Eng 2024; 10:6938-6953. [PMID: 39375226 DOI: 10.1021/acsbiomaterials.4c01478] [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] [Indexed: 10/09/2024]
Abstract
Rainbow trout (Oncorhynchus mykiss) is experiencing a catastrophic pandemic. In recent years, infectious hematopoietic necrosis virus (IHNV) has spread nationwide, resulting in significant mortality. Currently, there are no available treatments or vaccines for IHNV in China. Here, the Astragalus extract was purified and characterized. Then, we developed an inactivated IHNV vaccine with purified Astragalus polysaccharide (P-APS) as an adjuvant. Safety assays showed that IHNV was successfully inactivated. After a serious IHNV challenge, the cumulative mortality rates were 76.0, 38.0, and 22.1% in control, vaccine, and P-APS + vaccine groups, respectively. P-APS + vaccine was effective at reducing head kidney damage and apoptosis after IHNV challenge by histopathological and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) analyses. The P-APS + vaccine group showed better results in enhancing specific antibodies (IgM) and immune enzyme activities (C3, LZM, GOT, and GPT). RNA-seq revealed that many immune-related pathways were significantly enriched. TLR2, TLR7, C3, IFN-γ, IgM, MHC1, MHC2, MX1, and VIG1 were identified as core genes based on RNA-seq and PPI networks. Mechanistic investigations showed that P-APS + vaccine activates the immune pathway by upregulating the expression of these genes. P-ASP+vaccine induced effective innate and adaptive immune responses that were stronger than single vaccines after vaccination and IHNV challenged. Our findings will provide a promising vaccine candidate against IHNV.
Collapse
Affiliation(s)
- Yucai Pan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Zhe Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jinqiang Quan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Wei Gu
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Junwei Wang
- Shandong Wanzefeng Ocean Development Group Co., Ltd, Rizhao 276800, China
| | - Guiyan Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Junhao Lu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jianfu Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
24
|
Huang R, Wang K, Flamm MH, Vazquez J, Gercke C, Ton C, Whitmer T, Mathis PK, Ploeger KJM, Rameez S. Development and qualification of 3 L scale-down model for large scale vaccine process on Vero cell culture using microcarriers. Biotechnol Bioeng 2024; 121:3402-3414. [PMID: 38993032 DOI: 10.1002/bit.28785] [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: 03/01/2024] [Revised: 05/22/2024] [Accepted: 06/15/2024] [Indexed: 07/13/2024]
Abstract
Scale-down models (SDM) are pivotal tools for process understanding and improvement to accelerate the development of vaccines from laboratory research to global commercialization. In this study, a 3 L SDM representing a 50 L scale Vero cell culture process of a live-attenuated virus vaccine using microcarriers was developed and qualified based on the constant impeller power per volume principle. Both multivariate data analysis (MVDA) and the traditional univariate data analysis showed comparable and equivalent cell growth, metabolic activity, and product quality results across scales. Computational fluid dynamics simulation further confirmed similar hydrodynamic stress between the two scales.
Collapse
Affiliation(s)
- Renjing Huang
- Bioprocess Drug Substance Commercialization, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Kai Wang
- Bioprocess Drug Substance Commercialization, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Matthew H Flamm
- Applied Mathematics and Modeling, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Jorge Vazquez
- Center of Mathematical Sciences, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Chris Gercke
- Bioprocess Drug Substance Commercialization, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Christopher Ton
- Vaccine Process Development, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Travis Whitmer
- Bioprocess Drug Substance Commercialization, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Pamela K Mathis
- Global Quality Large Molecule Analytical Sciences, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Kristin J M Ploeger
- Bioprocess Drug Substance Commercialization, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Shahid Rameez
- Bioprocess Drug Substance Commercialization, Merck & Co., Inc., West Point, Pennsylvania, USA
| |
Collapse
|
25
|
Joseph J, Baby HM, Quintero JR, Kenney D, Mebratu YA, Bhatia E, Shah P, Swain K, Lee D, Kaur S, Li XL, Mwangi J, Snapper O, Nair R, Agus E, Ranganathan S, Kage J, Gao J, Luo JN, Yu A, Park D, Douam F, Tesfaigzi Y, Karp JM, Joshi N. Toward a Radically Simple Multi-Modal Nasal Spray for Preventing Respiratory Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406348. [PMID: 39318086 DOI: 10.1002/adma.202406348] [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: 05/03/2024] [Revised: 08/18/2024] [Indexed: 09/26/2024]
Abstract
Nasal sprays for pre-exposure prophylaxis against respiratory infections show limited protection (20-70%), largely due to their single mechanism of action-either neutralizing pathogens or blocking their entry at the nasal lining, and a failure to maximize the capture of respiratory droplets, allowing them to potentially rebound and reach deeper airways. This report introduces the Pathogen Capture and Neutralizing Spray (PCANS), which utilizes a multi-modal approach to enhance efficacy. PCANS coats the nasal cavity, capturing large respiratory droplets from the air, and serving as a physical barrier against a broad spectrum of viruses and bacteria, while rapidly neutralizing them with over 99.99% effectiveness. The formulation consists of excipients identified from the FDA's Inactive Ingredient Database and Generally Recognized as Safe list to maximize efficacy for each step in the multi-modal approach. PCANS demonstrates nasal retention for up to 8 hours in mice. In a severe Influenza A mouse model, a single pre-exposure dose of PCANS leads to a >99.99% reduction in lung viral titer and ensures 100% survival, compared to 0% in the control group. PCANS suppresses pathological manifestations and offers protection for at least 4 hours. This data suggest PCANS as a promising daily-use prophylactic against respiratory infections.
Collapse
Affiliation(s)
- John Joseph
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Helna Mary Baby
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Joselyn Rojas Quintero
- Harvard Medical School, Boston, MA, 02115, USA
- Division of Pulmonology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Devin Kenney
- National Emerging Infectious Diseases Laboratories, Department of Microbiology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Yohannes A Mebratu
- Harvard Medical School, Boston, MA, 02115, USA
- Division of Pulmonology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Eshant Bhatia
- Indian Institute of Technology, Mumbai, 400076, India
| | - Purna Shah
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Kabir Swain
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Dongtak Lee
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Shahdeep Kaur
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Xiang-Ling Li
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - John Mwangi
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Olivia Snapper
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Remya Nair
- Harvard Medical School, Boston, MA, 02115, USA
| | - Eli Agus
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Sruthi Ranganathan
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Julian Kage
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Jingjing Gao
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - James N Luo
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Anthony Yu
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Dongsung Park
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Florian Douam
- National Emerging Infectious Diseases Laboratories, Department of Microbiology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Yohannes Tesfaigzi
- Harvard Medical School, Boston, MA, 02115, USA
- Division of Pulmonology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Jeffrey M Karp
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Nitin Joshi
- Center for Accelerated Medical Innovation, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
26
|
Troncoso-Bravo T, Ramírez MA, Loaiza RA, Román-Cárdenas C, Papazisis G, Garrido D, González PA, Bueno SM, Kalergis AM. Advancement in the development of mRNA-based vaccines for respiratory viruses. Immunology 2024; 173:481-496. [PMID: 39161170 DOI: 10.1111/imm.13844] [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: 03/25/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Acute respiratory infections are the leading cause of death and illness in children under 5 years old and represent a significant burden in older adults. Primarily caused by viruses infecting the lower respiratory tract, symptoms include cough, congestion, and low-grade fever, potentially leading to bronchiolitis and pneumonia. Messenger ribonucleic acid (mRNA)-based vaccines are biopharmaceutical formulations that employ mRNA molecules to induce specific immune responses, facilitating the expression of viral or bacterial antigens and promoting immunization against infectious diseases. Notably, this technology had significant relevance during the COVID-19 pandemic, as these formulations helped to limit SARS-CoV-2 virus infections, hospitalizations, and deaths. Importantly, mRNA vaccines promise to be implemented as new alternatives for fighting other respiratory viruses, such as influenza, human respiratory syncytial virus, and human metapneumovirus. This review article analyzes mRNA-based vaccines' main contributions, perspectives, challenges, and implications against respiratory viruses.
Collapse
Affiliation(s)
- Tays Troncoso-Bravo
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ingeniería Química y Bioprocesos, Facultad de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mario A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ricardo A Loaiza
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Román-Cárdenas
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Georgios Papazisis
- Laboratory of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Clinical Research Unit, Special Unit for Biomedical Research and Education (SUBRE), School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Daniel Garrido
- Departamento de Ingeniería Química y Bioprocesos, Facultad de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
27
|
Agampodi S, Tadesse BT, Sahastrabuddhe S, Excler JL, Kim JH. Biases in COVID-19 vaccine effectiveness studies using cohort design. Front Med (Lausanne) 2024; 11:1474045. [PMID: 39540039 PMCID: PMC11557388 DOI: 10.3389/fmed.2024.1474045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Observational studies on COVID-19 vaccine effectiveness (VE) have provided critical real-world data, informing public health policy globally. These studies, primarily using pre-existing data sources, have been indispensable in assessing VE across diverse populations and developing sustainable vaccination strategies. Cohort design is frequently employed in VE research. The rapid implementation of vaccination campaigns during the COVID-19 pandemic introduced differential vaccination influenced by sociodemographic disparities, public policies, perceived risks, health-promoting behaviors, and health status, potentially resulting in biases such as healthy user bias, healthy vaccinee effect, frailty bias, differential depletion of susceptibility bias, and confounding by indication. The overwhelming burden on healthcare systems has escalated the risk of data inaccuracies, leading to outcome misclassifications. Additionally, the extensive array of diagnostic tests used during the pandemic has also contributed to misclassification biases. The urgency to publish quickly may have further influenced these biases or led to their oversight, affecting the validity of the findings. These biases in studies vary considerably depending on the setting, data sources, and analytical methods and are likely more pronounced in low- and middle-income country (LMIC) settings due to inadequate data infrastructure. Addressing and mitigating these biases is essential for accurate VE estimates, guiding public health strategies, and sustaining public trust in vaccination programs. Transparent communication about these biases and rigorous improvement in the design of future observational studies are essential.
Collapse
Affiliation(s)
- Suneth Agampodi
- Innovation, Initiatives and Enterprise Development Unit, International Vaccine Institute, Seoul, Republic of Korea
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Birkneh Tilahun Tadesse
- Epidemiology, Public Health, and Impact Unit, International Vaccine Institute, Seoul, Republic of Korea
| | - Sushant Sahastrabuddhe
- Innovation, Initiatives and Enterprise Development Unit, International Vaccine Institute, Seoul, Republic of Korea
| | | | - Jerome Han Kim
- International Vaccine Institute, Seoul, Republic of Korea
- College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Liu X, Min Q, Li Y, Chen S. Enhanced Cellular Immunity for Hepatitis B Virus Vaccine: A Novel Polyinosinic-Polycytidylic Acid-Incorporated Adjuvant Leveraging Cytoplasmic Retinoic Acid-Inducible Gene-Like Receptor Activation and Increased Antigen Uptake. Biomater Res 2024; 28:0096. [PMID: 39469105 PMCID: PMC11513446 DOI: 10.34133/bmr.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Conventional aluminum adjuvants exhibit limited cellular immunity. Polyinosinic-polycytidylic acid (poly I:C) activates cytoplasmic retinoic acid-inducible gene-like receptor (RLR), triggering strong T cell activation and cellular responses. However, when applied as an adjuvant, its limited endocytosis and restricted cytoplasmic delivery diminish its effectiveness and increase its toxicity. Hybrid polymer-lipid nanoparticle (PLNP) possesses numerous benefits such as good stability, reduced drug leakage, simple fabrication, easy property modulation, and excellent reproducibility compared to the lipid nanoparticle or the polymeric vector. Here, we developed a novel cationic polymer-lipid hybrid adjuvant capable of incorporating poly I:C to enhance cellular immunity. The hepatitis B surface antigen (HBsAg) was immobilized onto poly I:C-incorprated PLNP (PPLNP) via electrostatic interactions, forming the HBsAg/PPLNP vaccine formulation. The PPLNP adjuvant largely enhanced the cellular endocytosis and cytoplasmic transport of poly I:C, activating RLR followed by promoting type I interferon (IFN) secretion. Meanwhile, PPLNP obviously enhanced the antigen uptake, prolonged antigen retention at the site of administration, and facilitated enhanced transport of antigens to lymph nodes. The HBsAg/PPLNP nanovaccine led to amplified concentrations of antigen-specific immunoglobulin G (IgG), IFN-γ, granzyme B, and an enhanced IgG2a/IgG1 ratio, alongside the FasL+/CD8+ T cell activation, favoring a T helper 1 (TH1)-driven immune response. PPLNP, distinguished by its biocompatibility, ease of fabrication, and effectiveness in augmenting cellular immunity, holds significant promise as a new adjuvant.
Collapse
Affiliation(s)
- Xuhan Liu
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Institute for Inheritance-Based Innovation of Chinese Medicine, Marshall Laboratory of Biomedical Engineering, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Qiuxia Min
- Department of Pharmacy, First People’s Hospital of Yunnan Province,
Kunming University of Science and Technology, Kunming, 650034 Yunnan, China
| | - Yihui Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Siyuan Chen
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816 China
| |
Collapse
|
29
|
Muthukutty P, MacDonald J, Yoo SY. Combating Emerging Respiratory Viruses: Lessons and Future Antiviral Strategies. Vaccines (Basel) 2024; 12:1220. [PMID: 39591123 PMCID: PMC11598775 DOI: 10.3390/vaccines12111220] [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: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging viral diseases, including seasonal illnesses and pandemics, pose significant global public health risks. Respiratory viruses, particularly coronaviruses and influenza viruses, are associated with high morbidity and mortality, imposing substantial socioeconomic burdens. This review focuses on the current landscape of respiratory viruses, particularly influenza and SARS-CoV-2, and their antiviral treatments. It also discusses the potential for pandemics and the development of new antiviral vaccines and therapies, drawing lessons from past outbreaks to inform future strategies for managing viral threats.
Collapse
Affiliation(s)
| | | | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea; (P.M.); (J.M.)
| |
Collapse
|
30
|
Chen Z, Gou Q, Yuan Y, Zhang X, Zhao Z, Liao J, Zeng X, Jing H, Jiang S, Zhang W, Zeng H, Huang W, Zou Q, Zhang J. Vaccination with a trivalent Klebsiella pneumoniae vaccine confers protection in a murine model of pneumonia. Vaccine 2024; 42:126217. [PMID: 39163713 DOI: 10.1016/j.vaccine.2024.126217] [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: 02/16/2024] [Revised: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024]
Abstract
Klebsiella pneumoniae (K. pneumoniae) is an opportunistic pathogen and the major cause of healthcare-associated infections, which are increasingly complicated by the prevalence of highly invasive and hyper-virulent K. pneumoniae strains, necessitating the development of alternative strategies for combatting infections caused by this bacterium. In this study, we successfully constructed a fusion antigen called KP-Ag1, comprising three antigens (GlnH, FimA, and KPN_00466) that were previously identified through reverse vaccinology. Immunization with KP-Ag1 formulated with Al(OH)3 adjuvant elicited robust humoral and cellular immune response in mice, and conferred protective immunity in a murine model of K. pneumoniae lung infection. Further analysis of serum IgG subtypes from mice immunized with KP-Ag1 revealed a predominant IgG1 response, indicating that KP-Ag1 predominantly induces a Th2-biased immune response. Additionally, opsonophagocytic killing assay suggested that humoral immune responses play a pivotal role in mediating protection conferred by KP-Ag1. Moreover, KP-Ag1 was found to promote the activation and maturation of BMDCs in vitro, which is essential for subsequent efficient antigen presentation. More importantly, vaccination with KP-Ag1 demonstrated cross-protective efficacy against clinical isolates of K. pneumoniae varying in serotypes, antibiotic resistance, and virulence profiles. Therefore, KP-Ag1 holds promise as a candidate for K. pneumoniae vaccine development.
Collapse
MESH Headings
- Animals
- Klebsiella pneumoniae/immunology
- Klebsiella Infections/prevention & control
- Klebsiella Infections/immunology
- Mice
- Disease Models, Animal
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Adjuvants, Immunologic/administration & dosage
- Female
- Immunity, Humoral
- Vaccination/methods
- Antigens, Bacterial/immunology
- Pneumonia, Bacterial/prevention & control
- Pneumonia, Bacterial/immunology
- Mice, Inbred BALB C
- Immunity, Cellular
- Cross Protection/immunology
Collapse
Affiliation(s)
- Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Qiang Gou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Yue Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xiaoli Zhang
- Department of Clinical Hematology, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Jingwen Liao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xi Zeng
- Department of Pharmacy, General Hospital of Northern Theatre Command, Shenyang 110016, PR China
| | - Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Shichun Jiang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Wei Huang
- Department of Medical Laboratory, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, PR China.
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| |
Collapse
|
31
|
Lu RM, Hsu HE, Perez SJLP, Kumari M, Chen GH, Hong MH, Lin YS, Liu CH, Ko SH, Concio CAP, Su YJ, Chang YH, Li WS, Wu HC. Current landscape of mRNA technologies and delivery systems for new modality therapeutics. J Biomed Sci 2024; 31:89. [PMID: 39256822 PMCID: PMC11389359 DOI: 10.1186/s12929-024-01080-z] [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: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Realizing the immense clinical potential of mRNA-based drugs will require continued development of methods to safely deliver the bioactive agents with high efficiency and without triggering side effects. In this regard, lipid nanoparticles have been successfully utilized to improve mRNA delivery and protect the cargo from extracellular degradation. Encapsulation in lipid nanoparticles was an essential factor in the successful clinical application of mRNA vaccines, which conclusively demonstrated the technology's potential to yield approved medicines. In this review, we begin by describing current advances in mRNA modifications, design of novel lipids and development of lipid nanoparticle components for mRNA-based drugs. Then, we summarize key points pertaining to preclinical and clinical development of mRNA therapeutics. Finally, we cover topics related to targeted delivery systems, including endosomal escape and targeting of immune cells, tumors and organs for use with mRNA vaccines and new treatment modalities for human diseases.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Hsiang-En Hsu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Monika Kumari
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Guan-Hong Chen
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ming-Hsiang Hong
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Yin-Shiou Lin
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ching-Hang Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Yi-Jen Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Yi-Han Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Wen-Shan Li
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Chemistry, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| | - Han-Chung Wu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| |
Collapse
|
32
|
Tang B, Ma K, Liu Y, Wang X, Tang S, Xiao Y, Cheke RA. Managing spatio-temporal heterogeneity of susceptibles by embedding it into an homogeneous model: A mechanistic and deep learning study. PLoS Comput Biol 2024; 20:e1012497. [PMID: 39348420 PMCID: PMC11476686 DOI: 10.1371/journal.pcbi.1012497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/10/2024] [Accepted: 09/17/2024] [Indexed: 10/02/2024] Open
Abstract
Accurate prediction of epidemics is pivotal for making well-informed decisions for the control of infectious diseases, but addressing heterogeneity in the system poses a challenge. In this study, we propose a novel modelling framework integrating the spatio-temporal heterogeneity of susceptible individuals into homogeneous models, by introducing a continuous recruitment process for the susceptibles. A neural network approximates the recruitment rate to develop a Universal Differential Equations (UDE) model. Simultaneously, we pre-set a specific form for the recruitment rate and develop a mechanistic model. Data from a COVID Omicron variant outbreak in Shanghai are used to train the UDE model using deep learning methods and to calibrate the mechanistic model using MCMC methods. Subsequently, we project the attack rate and peak of new infections for the first Omicron wave in China after the adjustment of the dynamic zero-COVID policy. Our projections indicate an attack rate and a peak of new infections of 80.06% and 3.17% of the population, respectively, compared with the homogeneous model's projections of 99.97% and 32.78%, thus providing an 18.6% improvement in the prediction accuracy based on the actual data. Our simulations demonstrate that heterogeneity in the susceptibles decreases herd immunity for ~37.36% of the population and prolongs the outbreak period from ~30 days to ~70 days, also aligning with the real case. We consider that this study lays the groundwork for the development of a new class of models and new insights for modelling heterogeneity.
Collapse
Affiliation(s)
- Biao Tang
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi’an, People’s Republic of China
- The Interdisciplinary Research Center for Mathematics and Life Sciences, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Kexin Ma
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yan Liu
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xia Wang
- School of Mathematics and Statistics, Shaanxi Normal University, Xi’an, People’s Republic of China
| | - Sanyi Tang
- School of Mathematics and Statistics, Shaanxi Normal University, Xi’an, People’s Republic of China
| | - Yanni Xiao
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi’an, People’s Republic of China
- The Interdisciplinary Research Center for Mathematics and Life Sciences, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Robert A. Cheke
- Natural Resources Institute, University of Greenwich at Medway, Central Avenue, Chatham Maritime, Kent, United Kingdom
- Department of Infectious Disease Epidemiology, Imperial College London, School of Public Health, White City Campus, London, United Kingdom
| |
Collapse
|
33
|
Shams N, Jaydari A, Najafi H, Hataminejad M, Khanizadeh S, Pouladi I. An Overview of the Types of Adjuvants Used in the Vaccination Industry And Their Mechanisms of Action. Viral Immunol 2024; 37:324-336. [PMID: 39172659 DOI: 10.1089/vim.2024.0032] [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] [Indexed: 08/24/2024] Open
Abstract
The widespread use of efficient vaccines against infectious diseases is regarded as one of the most significant advancements in public health and techniques for preventing and protecting against infectious diseases and cancer. Because the purpose of vaccination is to elicit an appropriate, powerful, and long-lasting immune response against the pathogen, compounds such as adjuvants must be used to enhance these responses. Adjuvants have been widely used since their discovery to boost immune responses, prevent diseases, and activate protective immunity. Today, several types of adjuvants with varying properties are available for specific applications. Adjuvants are supramolecular substances or complexes that strengthen and prolong the immune response to antigens. These compounds have long-term immunological effects and are low in toxicity. They also lower the amount of antigen or the number of immunogenic reactions needed to improve vaccine efficacy and are used in specific populations. This article provides an overview of the adjuvants commonly used in the vaccination industry, their respective mechanisms of action, and discusses how they function to stimulate the immune system. Understanding the mechanisms of action of adjuvants is crucial for the development of effective and safe vaccines.
Collapse
Affiliation(s)
- Nemat Shams
- Department of Microbiology and Food Hygiene, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Amin Jaydari
- Department of Microbiology and Food Hygiene, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Hamideh Najafi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Hataminejad
- Department of Parasitology and Mycology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Sayyad Khanizadeh
- Hepatitis Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Iman Pouladi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
34
|
Ku KB, Chae J, Park WH, La J, Lee SS, Lee HK. Assessment of immunopathological responses of a novel non-chemical biocide in C57BL/6 for safe disinfection usage. Lab Anim Res 2024; 40:28. [PMID: 39135094 PMCID: PMC11320990 DOI: 10.1186/s42826-024-00214-6] [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: 04/01/2024] [Revised: 07/15/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Water electrospray technology has been developed and extensively studied for its physical properties and potential application as a non-chemical biocide against airborne pathogens. However, there are still concerns regarding the safety and potential toxicity of inhaling water electrospray (WE) particles. To address these potential hazards and offer insights into the impact of WE on humans, we analyzed the immunopathological response to WE by employing an intranasal challenge C57BL/6 mouse model. This analysis aimed to compare the effects of WE with those of sodium hypochlorite (SH), a well-known biocidal agent. RESULTS The study findings suggest that the WE did not trigger any pathological immune reactions in the intranasal-challenged C57BL/6 mouse model. Mice challenged with WE did not experience body weight loss, and there was no increase in inflammatory cytokine production compared to SH-treated mice. Histopathological analysis revealed that WE did not cause any damage to the lung tissue. In contrast, mice treated with SH exhibited significant lung tissue damage, characterized by the infiltration of neutrophils and eosinophils. Transcriptomic analysis of lung tissue further confirmed the absence of a pathological immune response in mice treated with WE compared to those treated with SH. Upon intranasal challenge with WE, the C57BL/6 mouse model did not show any evidence of immunopathological damage. CONCLUSIONS The results of this study suggest that WE is a safe technology for disinfecting airborne pathogens. It demonstrated little to no effect on immune system activation and pathological outcomes in the intranasal challenge C57BL/6 mouse model. These findings not only support the potential use of WE as an effective and safe method for air disinfection but also highlight the value of the intranasal challenge of the C57BL/6 mouse model in providing significant immunopathological insights for assessing the inhalation of novel materials for potential use.
Collapse
Affiliation(s)
- Keun Bon Ku
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Center for Infectious Disease Vaccine and Diagnosis Innovation, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Jihwan Chae
- Department of Mechanical Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Won Hyung Park
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Jeongwoo La
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Seung S Lee
- Department of Mechanical Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
35
|
Gong X, Peng L, Wang F, Liu J, Tang Y, Peng Y, Niu S, Yin J, Guo L, Lu H, Liu Y, Yang Y. Repeated Omicron infection dampens immune imprinting from previous vaccination and induces broad neutralizing antibodies against Omicron sub-variants. J Infect 2024; 89:106208. [PMID: 38908522 DOI: 10.1016/j.jinf.2024.106208] [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: 04/26/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE Similar with influenza virus, antigenic drift is highly relevant to SARS-CoV-2 evolution, and immune imprinting has been found to limit the performance of updated vaccines based on the emerging variants of SARS-CoV-2. We aimed to investigate whether repeated exposure to Omicron variant could reduce the immune imprinting from previous vaccination. METHODS A total of 194 participants with different status of vaccination (unvaccinated, regular vaccination and booster vaccination) confirmed for first infection and re-infection with BA.5, BF.7 and XBB variants were enrolled, and the neutralizing profiles against wild type (WT) SARS-CoV-2 and Omicron sub-variants were analyzed. RESULTS Neutralizing potency against the corresponding infected variant is significantly hampered along with the doses of vaccination during first infection. However, for the participants with first infection of BA.5/BF.7 variants and re-infection of XBB variant, immune imprinting was obviously alleviated, indicated as significantly increased ratio of the corresponding infected variant/WT ID50 titers and higher percentage of samples with high neutralizing activities (ID50 > 500) against BA.5, BF.7 and XBB variants. Moreover, repeated Omicron infection could induce strong neutralizing potency with broad neutralizing profiles against a series of other Omicron sub-variants, both in the vaccine naive and vaccine experienced individuals. CONCLUSIONS Our results demonstrate that repeated Omicron infection dampens immune imprinting from vaccination with WT SARS-CoV-2 and induces broad neutralizing profiles against Omicron sub-variants.
Collapse
Affiliation(s)
- Xiaohua Gong
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China
| | - Ling Peng
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China
| | - Fuxiang Wang
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China
| | - Jiexiang Liu
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Shenzhen Clinical School of Medicine, Guangzhou University of Chinese Medicine, China
| | - Yimin Tang
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China
| | - Yun Peng
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China
| | - Shiyu Niu
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China
| | - Juzhen Yin
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China
| | - Liping Guo
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China
| | - Hongzhou Lu
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China.
| | - Yingxia Liu
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China.
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China; Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, China; National Clinical Research Center for Infectious Disease, Shenzhen, China.
| |
Collapse
|
36
|
Papaneophytou C. Breaking the Chain: Protease Inhibitors as Game Changers in Respiratory Viruses Management. Int J Mol Sci 2024; 25:8105. [PMID: 39125676 PMCID: PMC11311956 DOI: 10.3390/ijms25158105] [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: 06/01/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Respiratory viral infections (VRTIs) rank among the leading causes of global morbidity and mortality, affecting millions of individuals each year across all age groups. These infections are caused by various pathogens, including rhinoviruses (RVs), adenoviruses (AdVs), and coronaviruses (CoVs), which are particularly prevalent during colder seasons. Although many VRTIs are self-limiting, their frequent recurrence and potential for severe health complications highlight the critical need for effective therapeutic strategies. Viral proteases are crucial for the maturation and replication of viruses, making them promising therapeutic targets. This review explores the pivotal role of viral proteases in the lifecycle of respiratory viruses and the development of protease inhibitors as a strategic response to these infections. Recent advances in antiviral therapy have highlighted the effectiveness of protease inhibitors in curtailing the spread and severity of viral diseases, especially during the ongoing COVID-19 pandemic. It also assesses the current efforts aimed at identifying and developing inhibitors targeting key proteases from major respiratory viruses, including human RVs, AdVs, and (severe acute respiratory syndrome coronavirus-2) SARS-CoV-2. Despite the recent identification of SARS-CoV-2, within the last five years, the scientific community has devoted considerable time and resources to investigate existing drugs and develop new inhibitors targeting the virus's main protease. However, research efforts in identifying inhibitors of the proteases of RVs and AdVs are limited. Therefore, herein, it is proposed to utilize this knowledge to develop new inhibitors for the proteases of other viruses affecting the respiratory tract or to develop dual inhibitors. Finally, by detailing the mechanisms of action and therapeutic potentials of these inhibitors, this review aims to demonstrate their significant role in transforming the management of respiratory viral diseases and to offer insights into future research directions.
Collapse
Affiliation(s)
- Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| |
Collapse
|
37
|
Zheng J, Li X, Masci AM, Kahn H, Huffman A, Asfaw E, Pan Y, Guo J, He V, Song J, Seleznev AI, Lin AY, He Y. Empowering standardization of cancer vaccines through ontology: enhanced modeling and data analysis. J Biomed Semantics 2024; 15:12. [PMID: 38890666 PMCID: PMC11186274 DOI: 10.1186/s13326-024-00312-3] [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: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The exploration of cancer vaccines has yielded a multitude of studies, resulting in a diverse collection of information. The heterogeneity of cancer vaccine data significantly impedes effective integration and analysis. While CanVaxKB serves as a pioneering database for over 670 manually annotated cancer vaccines, it is important to distinguish that a database, on its own, does not offer the structured relationships and standardized definitions found in an ontology. Recognizing this, we expanded the Vaccine Ontology (VO) to include those cancer vaccines present in CanVaxKB that were not initially covered, enhancing VO's capacity to systematically define and interrelate cancer vaccines. RESULTS An ontology design pattern (ODP) was first developed and applied to semantically represent various cancer vaccines, capturing their associated entities and relations. By applying the ODP, we generated a cancer vaccine template in a tabular format and converted it into the RDF/OWL format for generation of cancer vaccine terms in the VO. '12MP vaccine' was used as an example of cancer vaccines to demonstrate the application of the ODP. VO also reuses reference ontology terms to represent entities such as cancer diseases and vaccine hosts. Description Logic (DL) and SPARQL query scripts were developed and used to query for cancer vaccines based on different vaccine's features and to demonstrate the versatility of the VO representation. Additionally, ontological modeling was applied to illustrate cancer vaccine related concepts and studies for in-depth cancer vaccine analysis. A cancer vaccine-specific VO view, referred to as "CVO," was generated, and it contains 928 classes including 704 cancer vaccines. The CVO OWL file is publicly available on: http://purl.obolibrary.org/obo/vo/cvo.owl , for sharing and applications. CONCLUSION To facilitate the standardization, integration, and analysis of cancer vaccine data, we expanded the Vaccine Ontology (VO) to systematically model and represent cancer vaccines. We also developed a pipeline to automate the inclusion of cancer vaccines and associated terms in the VO. This not only enriches the data's standardization and integration, but also leverages ontological modeling to deepen the analysis of cancer vaccine information, maximizing benefits for researchers and clinicians. AVAILABILITY The VO-cancer GitHub website is: https://github.com/vaccineontology/VO/tree/master/CVO .
Collapse
Affiliation(s)
- Jie Zheng
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xingxian Li
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Anna Maria Masci
- Data Impact and Governance, Technology Data and Innovation, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hayleigh Kahn
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Anthony Huffman
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Eliyas Asfaw
- University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yuanyi Pan
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jinjing Guo
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Virginia He
- The College of Brown University, Brown University, Providence, RI, 02912, USA
| | - Justin Song
- College of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrey I Seleznev
- Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Asiyah Yu Lin
- Axle Research and Technology, Rockville, MD, 20852, USA
| | - Yongqun He
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
38
|
El-Fakharany EM, El-Gendi H, Saleh AK, El-Sayed MH, Alalawy AI, Jame R, Abdelaziz MA, Alshareef SA, El-Maradny YA. The use of proteins and peptides-based therapy in managing and preventing pathogenic viruses. Int J Biol Macromol 2024; 270:132254. [PMID: 38729501 DOI: 10.1016/j.ijbiomac.2024.132254] [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: 04/20/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Therapeutic proteins have been employed for centuries and reached approximately 50 % of all drugs investigated. By 2023, they represented one of the top 10 largest-selling pharma products ($387.03 billion) and are anticipated to reach around $653.35 billion by 2030. Growth hormones, insulin, and interferon (IFN α, γ, and β) are among the leading applied therapeutic proteins with a higher market share. Protein-based therapies have opened new opportunities to control various diseases, including metabolic disorders, tumors, and viral outbreaks. Advanced recombinant DNA biotechnology has offered the production of therapeutic proteins and peptides for vaccination, drugs, and diagnostic tools. Prokaryotic and eukaryotic expression host systems, including bacterial, fungal, animal, mammalian, and plant cells usually applied for recombinant therapeutic proteins large-scale production. However, several limitations face therapeutic protein production and applications at the commercial level, including immunogenicity, integrity concerns, protein stability, and protein degradation under different circumstances. In this regard, protein-engineering strategies such as PEGylation, glycol-engineering, Fc-fusion, albumin conjugation, and fusion, assist in increasing targeting, product purity, production yield, functionality, and the half-life of therapeutic protein circulation. Therefore, a comprehensive insight into therapeutic protein research and findings pave the way for their successful implementation, which will be discussed in the current review.
Collapse
Affiliation(s)
- Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute GEBRI, City of Scientific Research and Technological Applications (SRTA city), New Borg El-Arab, Alexandria 21934, Egypt; Pharmaceutical and Fermentation Industries Development Centre (PFIDC), City of Scientific Research and Technological Applications (SRTA-City), New Borg Al-Arab, Alexandria, Egypt.
| | - Hamada El-Gendi
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA city), New Borg El-Arab, Alexandria 21934, Egypt
| | - Ahmed K Saleh
- Cellulose and Paper Department, National Research Centre, El-Tahrir St., Dokki 12622, Giza, Egypt
| | - Mohamed H El-Sayed
- Department of Biology, College of Sciences and Arts-Rafha, Northern Border University, Arar, Saudi Arabia
| | - Adel I Alalawy
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rasha Jame
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mahmoud A Abdelaziz
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | | | - Yousra A El-Maradny
- Pharmaceutical and Fermentation Industries Development Centre (PFIDC), City of Scientific Research and Technological Applications (SRTA-City), New Borg Al-Arab, Alexandria, Egypt; Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Alamein 51718, Egypt
| |
Collapse
|
39
|
Ramos-Duarte VA, Orlowski A, Jaquenod de Giusti C, Corigliano MG, Legarralde A, Mendoza-Morales LF, Atela A, Sánchez MA, Sander VA, Angel SO, Clemente M. Safe plant Hsp90 adjuvants elicit an effective immune response against SARS-CoV2-derived RBD antigen. Vaccine 2024; 42:3355-3364. [PMID: 38631949 DOI: 10.1016/j.vaccine.2024.04.036] [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: 01/08/2024] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
To better understand the role of pHsp90 adjuvant in immune response modulation, we proposed the use of the Receptor Binding Domain (RBD) of the Spike protein of SARS-CoV2, the principal candidate in the design of subunit vaccines. We evaluated the humoral and cellular immune responses against RBD through the strategy "protein mixture" (Adjuvant + Antigen). The rRBD adjuvanted with rAtHsp81.2 group showed a higher increase of the anti-rRBD IgG1, while the rRBD adjuvanted with rNbHsp90.3 group showed a significant increase in anti-rRBD IgG2b/2a. These results were consistent with the cellular immune response analysis. Spleen cell cultures from rRBD + rNbHsp90.3-immunized mice showed significantly increased IFN-γ production. In contrast, spleen cell cultures from rRBD + rAtHsp81.2-immunized mice showed significantly increased IL-4 levels. Finally, vaccines adjuvanted with rNbHsp90.3 induced higher neutralizing antibody responses compared to those adjuvanted with rAtHsp81.2. To know whether both chaperones must form complexes to generate an effective immune response, we performed co-immunoprecipitation (co-IP) assays. The results indicated that the greater neutralizing capacity observed in the rRBD adjuvanted with rNbHsp90.3 group would be given by the rRBD-rNbHsp90.3 interaction rather than by the quality of the immune response triggered by the adjuvants. These results, together with our previous results, provide a comparative benchmark of these two novel and safe vaccine adjuvants for their capacity to stimulate immunity to a subunit vaccine, demonstrating the capacity of adjuvanted SARS-CoV2 subunit vaccines. Furthermore, these results revealed differences in the ability to modulate the immune response between these two pHsp90s, highlighting the importance of adjuvant selection for future rational vaccine and adjuvant design.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Vaccine
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19 Vaccines/immunology
- HSP90 Heat-Shock Proteins/immunology
- Immunity, Cellular
- Immunity, Humoral
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Mice, Inbred BALB C
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
Collapse
Affiliation(s)
- Victor A Ramos-Duarte
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Alejandro Orlowski
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" (CONICET), Universidad Nacional de La Plata, Facultad de Ciencias Médicas, La Plata, Argentina
| | - Carolina Jaquenod de Giusti
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" (CONICET), Universidad Nacional de La Plata, Facultad de Ciencias Médicas, La Plata, Argentina
| | - Mariana G Corigliano
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Ariel Legarralde
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Luisa F Mendoza-Morales
- Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina; Laboratorio de Biotecnologías en Bovinos y Ovinos, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Provincia de Buenos Aires, Argentina
| | - Agustín Atela
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Manuel A Sánchez
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Valeria A Sander
- Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina; Laboratorio de Biotecnologías en Bovinos y Ovinos, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Provincia de Buenos Aires, Argentina
| | - Sergio O Angel
- Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina; Laboratorio de Parasitología Molecular-UB2, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires, Argentina
| | - Marina Clemente
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina.
| |
Collapse
|
40
|
Sharif-Nia H, She L, Allen KA, Marôco J, Kaur H, Arslan G, Gorgulu O, Osborne JW, Rahmatpour P, Khoshnavay Fomani F. Parental hesitancy toward children vaccination: a multi-country psychometric and predictive study. BMC Public Health 2024; 24:1348. [PMID: 38762744 PMCID: PMC11102617 DOI: 10.1186/s12889-024-18806-1] [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: 10/23/2023] [Accepted: 05/09/2024] [Indexed: 05/20/2024] Open
Abstract
AIM Understanding vaccine hesitancy, as a critical concern for public health, cannot occur without the use of validated measures applicable and relevant to the samples they are assessing. The current study aimed to validate the Vaccine Hesitancy Scale (VHS) and to investigate the predictors of children's vaccine hesitancy among parents from Australia, China, Iran, and Turkey. To ensure the high quality of the present observational study the STROBE checklist was utilized. DESIGN A cross-sectional study. METHOD In total, 6,073 parent participants completed the web-based survey between 8 August 2021 and 1 October 2021. The content and construct validity of the Vaccine Hesitancy Scale was assessed. Cronbach's alpha and McDonald's omega were used to assess the scale's internal consistency, composite reliability (C.R.) and maximal reliability (MaxR) were used to assess the construct reliability. Multiple linear regression was used to predict parental vaccine hesitancy from gender, social media activity, and perceived financial well-being. RESULTS The results found that the VHS had a two-factor structure (i.e., lack of confidence and risk) and a total of 9 items. The measure showed metric invariance across four very different countries/cultures, showed evidence of good reliability, and showed evidence of validity. As expected, analyses indicated that parental vaccine hesitancy was higher in people who identify as female, more affluent, and more active on social media. CONCLUSIONS The present research marks one of the first studies to evaluate vaccine hesitancy in multiple countries that demonstrated VHS validity and reliability. Findings from this study have implications for future research examining vaccine hesitancy and vaccine-preventable diseases and community health nurses.
Collapse
Affiliation(s)
- Hamid Sharif-Nia
- Psychosomatic Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Nursing, Amol School of Nursing and Midwifery, Mazandaran University of Medical Sciences, Sari, Iran
| | - Long She
- Sunway Business School, Sunway University, Sunway City, Malaysia
| | - Kelly-Ann Allen
- School of Educational Psychology and Counselling, Faculty of Education, Monash University, Clayton, Australia
- Centre for Wellbeing Science, Faculty of Education, University of Melbourne, Parkville, Australia
| | - João Marôco
- William James Centre for Research ISPA - Instituto Universitário, Lisboa, Portugal
| | - Harpaljit Kaur
- Business School, Taylor's University Lakeside Campus, Subang Jaya, Malaysia
| | - Gökmen Arslan
- Department of Psychological Counseling, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ozkan Gorgulu
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Kırşehir Ahi Evran University, Kırşehir, Turkey
| | | | - Pardis Rahmatpour
- School of Nursing, Alborz University of Medical Sciences, Karaj, Iran
| | - Fatemeh Khoshnavay Fomani
- Nursing and Midwifery Care Research Center, School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Khan Z, Ahmed A, Ilyas U. The crucial role of pharmacovigilance in managing infectious diseases in lower and middle-income countries. Ther Adv Drug Saf 2024; 15:20420986241251593. [PMID: 38745585 PMCID: PMC11092529 DOI: 10.1177/20420986241251593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 04/04/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Zakir Khan
- Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University, Islamabad, Pakistan
| | - Ali Ahmed
- Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University, Islamabad, Pakistan
| | - Umair Ilyas
- Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University, Islamabad, Pakistan
| |
Collapse
|
42
|
Kumar A, Misra G, Mohandas S, Yadav PD. Multi-epitope vaccine design using in silico analysis of glycoprotein and nucleocapsid of NIPAH virus. PLoS One 2024; 19:e0300507. [PMID: 38728300 PMCID: PMC11086869 DOI: 10.1371/journal.pone.0300507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/29/2024] [Indexed: 05/12/2024] Open
Abstract
According to the 2018 WHO R&D Blueprint, Nipah virus (NiV) is a priority disease, and the development of a vaccine against NiV is strongly encouraged. According to criteria used to categorize zoonotic diseases, NiV is a stage III disease that can spread to people and cause unpredictable outbreaks. Since 2001, the NiV virus has caused annual outbreaks in Bangladesh, while in India it has caused occasional outbreaks. According to estimates, the mortality rate for infected individuals ranges from 70 to 91%. Using immunoinformatic approaches to anticipate the epitopes of the MHC-I, MHC-II, and B-cells, they were predicted using the NiV glycoprotein and nucleocapsid protein. The selected epitopes were used to develop a multi-epitope vaccine construct connected with linkers and adjuvants in order to improve immune responses to the vaccine construct. The 3D structure of the engineered vaccine was anticipated, optimized, and confirmed using a variety of computer simulation techniques so that its stability could be assessed. According to the immunological simulation tests, it was found that the vaccination elicits a targeted immune response against the NiV. Docking with TLR-3, 7, and 8 revealed that vaccine candidates had high binding affinities and low binding energies. Finally, molecular dynamic analysis confirms the stability of the new vaccine. Codon optimization and in silico cloning showed that the proposed vaccine was expressed to a high degree in Escherichia coli. The study will help in identifying a potential epitope for a vaccine candidate against NiV. The developed multi-epitope vaccine construct has a lot of potential, but they still need to be verified by in vitro & in vivo studies.
Collapse
Affiliation(s)
- Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida, Uttar Pradesh, India
| | - Gauri Misra
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida, Uttar Pradesh, India
| | - Sreelekshmy Mohandas
- Maximum Containment Laboratory, ICMR-National Institute of Virology, Microbial Containment Complex, Pashan, Pune, India
| | - Pragya D. Yadav
- Maximum Containment Laboratory, ICMR-National Institute of Virology, Microbial Containment Complex, Pashan, Pune, India
| |
Collapse
|
43
|
Saini I, Joshi J, Kaur S. Leishmania vaccine development: A comprehensive review. Cell Immunol 2024; 399-400:104826. [PMID: 38669897 DOI: 10.1016/j.cellimm.2024.104826] [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: 01/15/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Abstract
Infectious diseases like leishmaniasis, malaria, HIV, tuberculosis, leprosy and filariasis are responsible for an immense burden on public health systems. Among these, leishmaniasis is under the category I diseases as it is selected by WHO (World Health Organization) on the ground of diversity and complexity. High cost, resistance and toxic effects of Leishmania traditional drugs entail identification and development of therapeutic alternative. Since the natural infection elicits robust immunity, consistence efforts are going on to develop a successful vaccine. Clinical trials have been conducted on vaccines like Leish-F1, F2, and F3 formulated using specific Leishmania antigen epitopes. Current strategies utilize individual or combined antigens from the parasite or its insect vector's salivary gland extract, with or without adjuvant formulation for enhanced efficacy. Promising animal data supports multiple vaccine candidates (Lmcen-/-, LmexCen-/-), with some already in or heading for clinical trials. The crucial challenge in Leishmania vaccine development is to translate the research knowledge into affordable and accessible control tools that refines the outcome for those who are susceptible to infection. This review focuses on recent findings in Leishmania vaccines and highlights difficulties facing vaccine development and implementation.
Collapse
Affiliation(s)
- Isha Saini
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India
| | - Jyoti Joshi
- Goswami Ganesh Dutta Sanatan Dharma College, Sector-32C, Chandigarh, India
| | - Sukhbir Kaur
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India.
| |
Collapse
|
44
|
Tang X, Huo M, Chen Y, Huang H, Qin S, Luo J, Qin Z, Jiang X, Liu Y, Duan X, Wang R, Chen L, Li H, Fan N, He Z, He X, Shen B, Li SC, Song X. A novel deep generative model for mRNA vaccine development: Designing 5' UTRs with N1-methyl-pseudouridine modification. Acta Pharm Sin B 2024; 14:1814-1826. [PMID: 38572113 PMCID: PMC10985129 DOI: 10.1016/j.apsb.2023.11.003] [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: 08/24/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 04/05/2024] Open
Abstract
Efficient translation mediated by the 5' untranslated region (5' UTR) is essential for the robust efficacy of mRNA vaccines. However, the N1-methyl-pseudouridine (m1Ψ) modification of mRNA can impact the translation efficiency of the 5' UTR. We discovered that the optimal 5' UTR for m1Ψ-modified mRNA (m1Ψ-5' UTR) differs significantly from its unmodified counterpart, highlighting the need for a specialized tool for designing m1Ψ-5' UTRs rather than directly utilizing high-expression endogenous gene 5' UTRs. In response, we developed a novel machine learning-based tool, Smart5UTR, which employs a deep generative model to identify superior m1Ψ-5' UTRs in silico. The tailored loss function and network architecture enable Smart5UTR to overcome limitations inherent in existing models. As a result, Smart5UTR can successfully design superior 5' UTRs, greatly benefiting mRNA vaccine development. Notably, Smart5UTR-designed superior 5' UTRs significantly enhanced antibody titers induced by COVID-19 mRNA vaccines against the Delta and Omicron variants of SARS-CoV-2, surpassing the performance of vaccines using high-expression endogenous gene 5' UTRs.
Collapse
Affiliation(s)
- Xiaoshan Tang
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Miaozhe Huo
- Department of Computer Science, City University of Hong Kong, Hong Kong 99907, China
| | - Yuting Chen
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Hai Huang
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Shugang Qin
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Jiaqi Luo
- Department of Computer Science, City University of Hong Kong, Hong Kong 99907, China
| | - Zeyi Qin
- Department of Biology, Brandeis University, Boston, MA 02453, USA
| | - Xin Jiang
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yongmei Liu
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Xing Duan
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ruohan Wang
- Department of Computer Science, City University of Hong Kong, Hong Kong 99907, China
| | - Lingxi Chen
- Department of Computer Science, City University of Hong Kong, Hong Kong 99907, China
| | - Hao Li
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Na Fan
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Zhongshan He
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Xi He
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Bairong Shen
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, Hong Kong 99907, China
| | - Xiangrong Song
- Institute of Systems Genetics, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| |
Collapse
|
45
|
Auzenbergs M, Maure C, Kang H, Clark A, Brady O, Sahastrabuddhe S, Abbas K. Programmatic considerations and evidence gaps for chikungunya vaccine introduction in countries at risk of chikungunya outbreaks: Stakeholder analysis. PLoS Negl Trop Dis 2024; 18:e0012075. [PMID: 38574163 PMCID: PMC11020901 DOI: 10.1371/journal.pntd.0012075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 04/16/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024] Open
Abstract
Chikungunya can have longstanding effects on health and quality of life. Alongside the recent approval of the world's first chikungunya vaccine by the US Food and Drug Administration in November 2023 and with new chikungunya vaccines in the pipeline, it is important to understand the perspectives of stakeholders before vaccine rollout. Our study aim is to identify key programmatic considerations and gaps in Evidence-to-Recommendation criteria for chikungunya vaccine introduction. We used purposive and snowball sampling to identify global, national, and subnational stakeholders from outbreak prone areas, including Latin America, Asia, and Africa. Semi-structured in-depth interviews were conducted and analysed using qualitative descriptive methods. We found that perspectives varied between tiers of stakeholders and geographies. Unknown disease burden, diagnostics, non-specific disease surveillance, undefined target populations for vaccination, and low disease prioritisation were critical challenges identified by stakeholders that need to be addressed to facilitate rolling out a chikungunya vaccine. Future investments should address these challenges to generate useful evidence for decision-making on new chikungunya vaccine introduction.
Collapse
Affiliation(s)
- Megan Auzenbergs
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Clara Maure
- International Vaccine Institute, Seoul, South Korea
| | - Hyolim Kang
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Andrew Clark
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Oliver Brady
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Kaja Abbas
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
46
|
Choi SI, Kim S, Jin Y, Valentini C, Badham M, Colleoni E, Romenti S. Effects of Individuals' Cultural Orientations and Trust in Government Health Communication Sources on Behavioral Intentions During a Pandemic: A Cross-Country Study. HEALTH COMMUNICATION 2024; 39:107-121. [PMID: 36576168 DOI: 10.1080/10410236.2022.2159975] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Public health messages disseminated by trusted government authorities are likely to have more influence over individuals' intentions and behaviors. However, individuals worldwide have different levels of trust in government authorities, which leads to varying levels of compliance intentions. Additionally, these trust levels may vary during major public crises, such as pandemics. Based on a COVID-19 pandemic communication survey (N = 3,065) disseminated throughout six countries (Australia, Finland, Italy, South Korea, Sweden, and the United States), this study examined the association among trust in distinct government sources, cultural orientations, and health behavioral intentions. Findings indicated that trust in official health communication sources at four governmental levels (i.e. national government, the head of the national government, the national health authority, and the chief representative of the national health authority) was related to vaccination intentions and other behavioral compliance intentions (i.e. willingness to prevent COVID-19 infection in other ways). Meanwhile, these direct associations were mediated by the cultural orientations of power distance and uncertainty avoidance. Findings also revealed that the direct association of trust in government sources and the indirect relationship through the above cultural orientations varied by country. This study offers insight into the important role of credible sources and individuals' cultural orientations in the domain of health communication aimed at influencing behavioral intentions.
Collapse
Affiliation(s)
- Sung In Choi
- Grady College of Journalism and Mass Communication, University of Georgia
| | - Sungsu Kim
- School of Communication, Kookmin University
| | - Yan Jin
- Grady College of Journalism and Mass Communication, University of Georgia
| | | | - Mark Badham
- Leeds Business School, Leeds Beckett University
| | - Elanor Colleoni
- Department of Business, Law, Economics and Consumer Behavior, IULM University
| | - Stefania Romenti
- Department of Business, Law, Economics and Consumer Behavior, IULM University
| |
Collapse
|
47
|
Jitsuk NC, Chadsuthi S, Modchang C. Vaccination strategies impact the probability of outbreak extinction: A case study of COVID-19 transmission. Heliyon 2024; 10:e28042. [PMID: 38524580 PMCID: PMC10958689 DOI: 10.1016/j.heliyon.2024.e28042] [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: 05/31/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024] Open
Abstract
Mass vaccination has proven to be an effective control measure for mitigating the transmission of infectious diseases. Throughout history, various vaccination strategies have been employed to control infections and terminate outbreaks. In this study, we utilized the transmission of COVID-19 as a case study and constructed a stochastic age-structured compartmental model to investigate the effectiveness of different vaccination strategies. Our analysis focused on estimating the outbreak extinction probability under different vaccination scenarios in both homogeneous and heterogeneous populations. Notably, we found that population heterogeneity can enhance the likelihood of outbreak extinction at varying levels of vaccine coverage. Prioritizing vaccinations for individuals with higher infection risk was found to maximize outbreak extinction probability and reduce overall infections, while allocating vaccines to those with higher mortality risk has been proven more effective in reducing deaths. Moreover, our study highlighted the significance of booster doses as the vaccine effectiveness wanes over time, showing that they can significantly enhance the extinction probability and mitigate disease transmission.
Collapse
Affiliation(s)
- Natcha C. Jitsuk
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Center for Disease Modeling, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sudarat Chadsuthi
- Center for Disease Modeling, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Physics, Research Center for Academic Excellence in Applied Physics, Faculty of Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Charin Modchang
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Center for Disease Modeling, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Centre of Excellence in Mathematics, MHESI, Bangkok, 10400, Thailand
- Thailand Center of Excellence in Physics, Ministry of Higher Education, Science, Research and Innovation, 328 Si Ayutthaya Road, Bangkok, 10400, Thailand
| |
Collapse
|
48
|
Chen H, Wang L, Zhao X, Jiang H, Wu M, Ding Y, Jia X, Zhang Y, Li T, Zhang Y, Zhou W, Zheng P, Yang Y, Du J. A Polymer-Based Antigen Carrier Activates Two Innate Immune Pathways for Adjuvant-Free Subunit Vaccines. ACS NANO 2024; 18:9160-9175. [PMID: 38478910 DOI: 10.1021/acsnano.4c00925] [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: 03/27/2024]
Abstract
The activation of multiple Pattern Recognition Receptors (PRRs) has been demonstrated to trigger inflammatory responses and coordinate the host's adaptive immunity during pathogen infections. The use of PRR agonists as vaccine adjuvants has been reported to synergistically induce specific humoral and cellular immune responses. However, incorporating multiple PRR agonists as adjuvants increases the complexity of vaccine design and manufacturing. In this study, we discovered a polymer that can activate both the Toll-like receptor (TLR) pathway and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. The polymer was then conjugated to protein antigens, creating an antigen delivery system for subunit vaccines. Without additional adjuvants, the antigen-polymer conjugates elicited strong antigen-specific humoral and cellular immune responses. Furthermore, the antigen-polymer conjugates, containing the Receptor Binding Domain (RBD) of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Protein or the Monkeypox Antigen M1R as the antigens, were found to induce potent antigen-specific antibodies, neutralizing antibodies, and cytotoxic T cells. Immunization with M1R-polymer also resulted in effective protection in a lethal challenge model. In conclusion, this vaccine delivery platform offers an effective, safe, and simple strategy for inducing antigen-specific immunity against infectious diseases.
Collapse
Affiliation(s)
- Hang Chen
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Luyao Wang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Xiaofan Zhao
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Haolin Jiang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
- Academy for Advanced Interdisciplinary Studies (AAIS), Peking University-Tsinghua University-National Institute Biological Sciences (PTN) Joint Graduate Program, Peking University, Beijing 100871, China
| | - Mengling Wu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yanchao Ding
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Xiangqian Jia
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yaning Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Tiantian Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yue Zhang
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Wen Zhou
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Peiyuan Zheng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yilong Yang
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Juanjuan Du
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
49
|
Liu M, Feng Y, Lu Y, Huang R, Zhang Y, Zhao Y, Mo R. Lymph-targeted high-density lipoprotein-mimetic nanovaccine for multi-antigenic personalized cancer immunotherapy. SCIENCE ADVANCES 2024; 10:eadk2444. [PMID: 38478602 PMCID: PMC10936870 DOI: 10.1126/sciadv.adk2444] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/07/2024] [Indexed: 11/02/2024]
Abstract
Cancer vaccines show huge potential for cancer prevention and treatment. However, their efficacy remains limited due to weak immunogenicity regarding inefficient stimulation of cytotoxic T lymphocyte (CTL) responses. Inspired by the unique characteristic and biological function of high-density lipoprotein (HDL), we here develop an HDL-mimicking nanovaccine with the commendable lymph-targeted capacity to potently elicit antitumor immunity using lipid nanoparticle that is co-loaded with specific cancer cytomembrane harboring a collection of tumor-associated antigens and an immune adjuvant. The nanoparticulate impact is explored on the efficiency of lymphatic targeting and dendritic cell uptake. The optimized nanovaccine promotes the co-delivery of antigens and adjuvants to lymph nodes and maintains antigen presentation of dendritic cells, resulting in long-term immune surveillance as the elevated frequency of CTLs within lymphoid organs and tumor tissue. Immunization of nanovaccine suppresses tumor formation and growth and augments the therapeutic efficacy of checkpoint inhibitors notably on the high-stemness melanoma in the mouse models.
Collapse
Affiliation(s)
| | | | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Renqi Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yanan Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | | |
Collapse
|
50
|
Pérez-Rubio P, Lavado-García J, Bosch-Molist L, Romero EL, Cervera L, Gòdia F. Extracellular vesicle depletion and UGCG overexpression mitigate the cell density effect in HEK293 cell culture transfection. Mol Ther Methods Clin Dev 2024; 32:101190. [PMID: 38327808 PMCID: PMC10847930 DOI: 10.1016/j.omtm.2024.101190] [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: 09/01/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
The hitherto unexplained reduction of cell-specific productivity in transient gene expression (TGE) at high cell density (HCD) is known as the cell density effect (CDE). It currently represents a major challenge in TGE-based bioprocess intensification. This phenomenon has been largely reported, but the molecular principles governing it are still unclear. The CDE is currently understood to be caused by the combination of an unknown inhibitory compound in the extracellular medium and an uncharacterized cellular change at HCD. This study investigates the role of extracellular vesicles (EVs) as extracellular inhibitors for transfection through the production of HIV-1 Gag virus-like particles (VLPs) via transient transfection in HEK293 cells. EV depletion from the extracellular medium restored transfection efficiency in conditions that suffer from the CDE, also enhancing VLP budding and improving production by 60%. Moreover, an alteration in endosomal formation was observed at HCD, sequestering polyplexes and preventing transfection. Overexpression of UDP-glucose ceramide glucosyltransferase (UGCG) enzyme removed intracellular polyplex sequestration, improving transfection efficiency. Combining EV depletion and UGCG overexpression improved transfection efficiency by ∼45% at 12 × 106 cells/mL. These results suggest that the interaction between polyplexes and extracellular and intracellular vesicles plays a crucial role in the CDE, providing insights for the development of strategies to mitigate its impact.
Collapse
Affiliation(s)
- Pol Pérez-Rubio
- Grup d’Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Jesús Lavado-García
- Grup d’Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Laia Bosch-Molist
- Grup d’Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Elianet Lorenzo Romero
- Grup d’Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Laura Cervera
- Grup d’Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Francesc Gòdia
- Grup d’Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| |
Collapse
|