1
|
Zheng X, Liu B, Ni P, Cai L, Shi X, Ke Z, Zhang S, Hu B, Yang B, Xu Y, Long W, Fang Z, Wang Y, Zhang W, Xu Y, Wang Z, Pan K, Zhou K, Wang H, Geng H, Hu H, Liu B. Development and application of an uncapped mRNA platform. Ann Med 2025; 57:2437046. [PMID: 39648715 PMCID: PMC11632943 DOI: 10.1080/07853890.2024.2437046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/28/2023] [Accepted: 06/01/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND A novel uncapped mRNA platform was developed. METHODS Five lipid nanoparticle (LNP)-encapsulated mRNA constructs were made to evaluate several aspects of our platform, including transfection efficiency and durability in vitro and in vivo and the activation of humoral and cellular immunity in several animal models. The constructs were eGFP-mRNA-LNP (for enhanced green fluorescence mRNA), Fluc-mRNA-LNP (for firefly luciferase mRNA), SδT-mRNA-LNP (for Delta strain SARS-CoV-2 spike protein trimer mRNA), gDED-mRNA-LNP (for truncated glycoprotein D mRNA coding ectodomain from herpes simplex virus type 2 (HSV2)) and gDFR-mRNA-LNP (for truncated HSV2 glycoprotein D mRNA coding amino acids 1-400). RESULTS Quantifiable target protein expression was achieved in vitro and in vivo with eGFP- and Fluc-mRNA-LNP. SδT-mRNA-LNP, gDED-mRNA-LNP and gDFR-mRNA-LNP induced both humoral and cellular immune responses comparable to those obtained by previously reported capped mRNA-LNP constructs. Notably, SδT-mRNA-LNP elicited neutralizing antibodies in hamsters against the Omicron and Delta strains. Additionally, gDED-mRNA-LNP and gDFR-mRNA-LNP induced potent neutralizing antibodies in rabbits and mice. The mRNA constructs with uridine triphosphate (UTP) outperformed those with N1-methylpseudouridine triphosphate (N1mψTP) in the induction of antibodies via SδT-mRNA-LNP. CONCLUSIONS Our uncapped, process-simplified and economical mRNA platform may have broad utility in vaccines and protein replacement drugs.KEY MESSAGESThe mRNA platform described in our paper uses internal ribosome entry site (IRES) (Rapid, Amplified, Capless and Economical, RACE; Register as BH-RACE platform) instead of caps and uridine triphosphate (UTP) instead of N1-methylpseudouridine triphosphate (N1mψTP) to synthesize mRNA.Through the self-developed packaging instrument and lipid nanoparticle (LNP) delivery system, mRNA can be expressed in cells more efficiently, quickly and economically.Particularly exciting is that potent neutralizing antibodies against Delta and Omicron real viruses were induced with the new coronavirus S protein mRNA vaccine from the BH-RACE platform.
Collapse
Affiliation(s)
- Xiaodi Zheng
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Biao Liu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Peng Ni
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Linkang Cai
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Xiaotai Shi
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zonghuang Ke
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Siqi Zhang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Bing Hu
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Binfeng Yang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Yiyan Xu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Wei Long
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zhizheng Fang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Yang Wang
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Xu
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zhong Wang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Kai Pan
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Kangping Zhou
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Hanming Wang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Hui Geng
- School of Life Science, Huazhong Normal University, Wuhan, China
| | - Han Hu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Binlei Liu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| |
Collapse
|
2
|
Azeem M, Cancemi P, Mukhtar F, Marino S, Peri E, Di Prima G, De Caro V. Efficacy and limitations of SARS-CoV-2 vaccines - A systematic review. Life Sci 2025; 371:123610. [PMID: 40189198 DOI: 10.1016/j.lfs.2025.123610] [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: 01/03/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
The emergence of the SARS-CoV-2 virus worldwide led to the call for the development of effective and safe vaccines to contain the spread and effects of COVID-19. Using information from 40 publications, including clinical trials and observational studies from 2019 to 2024, this review assesses the effectiveness, safety, and limitations of four major vaccines: Sinopharm (BBIBP-CorV), Moderna (mRNA-1273), Pfizer-BioNTech (BNT162b2), and CoronaVac. Pfizer-BioNTech and Moderna's mRNA vaccines proved to be more effective than others; Moderna's vaccines showed an efficacy of 94.1 % against symptomatic infection, while Pfizer-BioNTech's vaccines showed an efficacy of up to 95 %, against severe diseases and hospitalization. These vaccinations, which included protection against Omicron and Delta variants, offered notable protection against serious illness, hospitalization, and mortality. Severe adverse events were rare while most adverse events were mild to moderate, such as headaches, fatigue, and localized reactions. In contrast, inactivated virus vaccines such as Sinopharm and CoronaVac with efficacies ranging from 50 to 79 % against symptomatic infection showed lower levels of effectiveness. In Phase 3 trial, Sinopharm showed 72.8 % efficacy, whereas CoronaVac demonstrated roughly 67 % efficacy in population against hospitalization and severe disease. Booster doses were required for adequate immunological response, especially against novel strains, as these vaccinations proved to be less effective in older populations. They showed considerable safety profiles, with mild side effects, but their low immunogenicity is concerning. This review emphasizes the importance of continuously evaluating vaccines in response to the evolving virus, essential for improving international immunization programs.
Collapse
Affiliation(s)
- Muhammad Azeem
- Dipartimento di Medicina di Precisione in Area Medica, Chirurgica e Critica (Me.Pre.C.C.), Università degli Studi di Palermo, Via Liborio Giuffre, 590127 Palermo, Italy
| | - Patrizia Cancemi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Farwa Mukhtar
- Dipartimento di Medicina e Scienze della Salute "V. Tiberio", Università degli Studi del Molise, Campobasso, Italy
| | - Sefora Marino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Emanuela Peri
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Giulia Di Prima
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Viviana De Caro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
3
|
Khalil Z, Fouly M, Helito P. Unprecedented case of rapid-onset autoimmune hepatitis triggered by COVID-19 mRNA vaccination, complicated by concurrent severe hemophagocytic lymphohistiocytosis and acute liver failure in a previously healthy adult: a multidisciplinary approach and review of therapeutic interventions. Clin J Gastroenterol 2025; 18:408-416. [PMID: 40198479 DOI: 10.1007/s12328-025-02109-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/22/2025] [Indexed: 04/10/2025]
Abstract
A previously healthy 35-year-old male developed acute autoimmune hepatitis 5 weeks after receiving his second dose of an mRNA vaccine, presenting with jaundice, elevated liver enzymes, and abdominal pain. The condition rapidly progressed to severe hemophagocytic lymphohistiocytosis (HLH) and acute liver failure, confirmed by liver biopsy and bone marrow aspirate. Despite aggressive multidisciplinary treatment, including corticosteroids, immunoglobulin, and IL-1 antagonists, the patient deteriorated, developing multi-organ failure. Emergency liver transplantation was considered but was not viable due to his unstable condition. The case highlights a potential rare vaccine-associated immune response which we believe has not been reported in the literature, requiring prompt recognition and multidisciplinary management. Further research is needed to understand the underlying immunogenic triggers and optimize treatment.
Collapse
Affiliation(s)
- Zeyad Khalil
- College of Medicine, October 6 University, October City, Egypt.
| | - Mohamed Fouly
- College of Medicine, October 6 University, October City, Egypt
| | - Paulo Helito
- Research Department, October 6 University, October City, Egypt
| |
Collapse
|
4
|
Wu X, Qiao J, Xiao F, Guo L. Enhancing GLP-1 expression via IVT mRNA and fusion protein technology for diabetes therapy. J Pharm Sci 2025; 114:103829. [PMID: 40393145 DOI: 10.1016/j.xphs.2025.103829] [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: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Diabetes is a chronic metabolic disorder with high incidence and prevalence worldwide. This study explores a novel glucagon-like peptide-1-Fc (GLP-1-Fc) mRNA designed to improve diabetes management by inducing stable and persistent production of GLP-1-Fc protein. METHODS The GLP-1-Fc mRNA was generated using in vitro transcription and fusion protein technology. Protein expression was assessed via western blot and enzyme-linked immunosorbent assay (ELISA) in Human Embryonic Kidney 293T (HEK293T) cells. GLP-1-Fc mRNA and GLP-1-Fc protein (dulaglutide) were administered to C57BL/6J and db/db mice to evaluate protein levels, GLP-1 receptor activity, hypoglycemic effects, and safety using ELISA, lance ultra cAMP assay, blood glucose levels detection, immunofluorescence, and hematoxylin and eosin staining. RESULTS The designed mRNA fused with the Fc region successfully encoded GLP-1-Fc, showing optimal stability and translation efficiency. The GLP-1-Fc protein levels were significantly higher in the GLP-1-Fc mRNA treatment group than those in the control mice. The GLP-1-Fc mRNA effectively reduced blood glucose levels and increased GLP-1 receptor expression in db/db mice after both single and repeated administrations. Moreover, the GLP-1-Fc mRNA provided prolonged glucose reduction with similar efficacy to GLP-1 protein drug, dulaglutide. Besides, intraperitoneal delivery of GLP-1-Fc mRNA does not induce tissue damage. CONCLUSIONS Compared to conventional peptide-based therapies, GLP-1-Fc mRNA represents a promising strategy for diabetes treatment by enabling sustained in vivo protein expression, achieving effective glycemic control, and offering a streamlined manufacturing process with reduced production complexity.
Collapse
Affiliation(s)
- Xiaoying Wu
- Peking University Fifth School of Clinical Medicine, Beijing, China; Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Beijing, China
| | - Jingtao Qiao
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Beijing, China
| | - Fei Xiao
- Peking University Fifth School of Clinical Medicine, Beijing, China; Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| | - Lixin Guo
- Peking University Fifth School of Clinical Medicine, Beijing, China; Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Beijing, China.
| |
Collapse
|
5
|
McCormack MJ, Scott S, Logan N, Raveendran S, Newman J, Santos IA, Bailey D, Murcia PR, Thomson EC, Hosie MJ, Willett BJ. Estimating population immunity to SARS-CoV-2 by random sampling from primary and secondary healthcare in Scotland, May 2024. EBioMedicine 2025; 116:105760. [PMID: 40381379 DOI: 10.1016/j.ebiom.2025.105760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND As the COVID-19 pandemic has ended, the global focus has shifted from "pandemic response" to "long-term management". With no ongoing nationwide serosurveillance studies, our understanding of the level of immunity in the general population has diminished. In this study, we screened random samples from a biorepository serving the largest health board in Scotland for antibodies against SARS-CoV-2 to define the current immunological landscape, informing vaccine strategies going forward. METHODS 997 pseudonymized serum samples were obtained from NHS Greater Glasgow and Clyde (NHS GGC) biorepository in May 2024, along with associated data for age, sex, and COVID-19 vaccine history. Samples spanned ages from 19 to 98 years, with 59.0% female and 41.0% male, and 39.1% from primary healthcare (GP practices) and 61.0% from secondary healthcare (hospitals). Anti-SARS-CoV-2 receptor binding domain (RBD)-specific antibodies were measured by enzyme-linked immunosorbent assay (ELISA), while neutralising antibodies were quantified using HIV(SARS-CoV-2) pseudotype-based virus neutralisation assay (PVNA). ELISAs measured both total IgG and IgG4-mediated responses. Pseudotypes were prepared bearing spike proteins from vaccine antigens B.1 and XBB.1.5, contemporaneous circulating variants KP.3.1.1 and LB.1, and the emerging variant XEC. Samples were grouped by number of COVID-19 vaccine doses received (from no vaccination to ≥8 doses) and 12 samples from each group were screened by ELISA and PVNA. FINDINGS The random selection of 1000 samples provided a broad cross-section of the population derived from patients with a range of individual vaccine histories, from those having received no COVID-19 vaccines to those having received 8 or more doses. The number of doses received increased with age, from a mean age of ∼40 for those having received one dose to a mean age of 77-78 for those having received 7 or 8 doses. While total IgG responses were similar across each of the groups, irrespective of vaccine history, repeated exposure to mRNA-based vaccines elicited an increase in SARS-CoV-2-specific IgG4. Neutralising antibody titres against the vaccine antigens B.1 and XBB.1.5 increased with age, reaching maximum geometric mean titres of 5610 (95% CI, 2773-11,349) for B.1 and 4577 (1832-11,440) for XBB.1.5 in those receiving 8 doses. In all groups, titres measured against the KP.3.1.1, LB.1 and XEC were significantly lower, consistent with the emergence of immune evasive variants over time. Cross-neutralisation of KP.3.1.1 was limited to maxima of 145 (62.2-336) and 187 (83.8-418) in the 7 and 8 dose groups, while titres against XEC were 105 (47-233) and 90.9 (48.1-172) respectively. INTERPRETATION In the absence of systematic COVID-19 serosurveillance, random sampling of sera from biorepositories associated with major health boards can generate valuable data about the level of immunity in the general population, informing estimates of vaccine effectiveness and antigen selection. FUNDING United Kingdom Medical Research Council and Genotype-to-Phenotype National Virology Consortium.
Collapse
Affiliation(s)
- Mhairi J McCormack
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| | - Sam Scott
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| | - Nicola Logan
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| | - Savitha Raveendran
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| | - Joseph Newman
- The Pirbright Institute, Guildford, Surrey, United Kingdom.
| | - Igor A Santos
- The Pirbright Institute, Guildford, Surrey, United Kingdom.
| | - Dalan Bailey
- The Pirbright Institute, Guildford, Surrey, United Kingdom.
| | - Pablo R Murcia
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| | - Emma C Thomson
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| | - Margaret J Hosie
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| | - Brian J Willett
- MRC University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow, United Kingdom.
| |
Collapse
|
6
|
Elliott L, Foster T, Castillo P, Mendez-Gomez H, Sayour EJ. Therapeutic mRNA vaccine applications in oncology. Mol Ther 2025:S1525-0016(25)00362-4. [PMID: 40336197 DOI: 10.1016/j.ymthe.2025.04.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
The emergence of mRNA vaccines for infectious diseases has heralded development for a slew of other indications, including cancer. Lipid particle delivery vehicles can protect RNA from degradation and promote delivery to intended targets in vivo. In this mini-review, we discuss mRNA vaccine mechanisms and capacity for enhancement followed by progress to date in the clinical development of mRNA cancer vaccines and implications of mRNA localization patterns and biologic response correlates. These advances position emerging mRNA biotechnologies for success against personalized diseases such as cancer.
Collapse
Affiliation(s)
- Leighton Elliott
- Department of Medicine, Division of Hematology & Oncology, University of Florida, Gainesville, FL 32608, USA; Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Florida, Gainesville, FL 32610, USA
| | - Timothy Foster
- Department of Pediatrics, Division of Endocrinology, University of Florida, Gainesville, FL 32610, USA
| | - Paul Castillo
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Florida, Gainesville, FL 32610, USA
| | - Hector Mendez-Gomez
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| | - Elias J Sayour
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Florida, Gainesville, FL 32610, USA; Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
7
|
Damase TR, Cooke JP. RNA therapeutics in cardiovascular medicine. Curr Opin Cardiol 2025; 40:139-149. [PMID: 39998478 PMCID: PMC12055242 DOI: 10.1097/hco.0000000000001210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
PURPOSE OF REVIEW RNA therapeutics came to global attention when mRNA-based vaccines provided an answer to the SARS-CoV-2 pandemic. The immense significance of this development notwithstanding, it is important to note that almost a decade prior to the pandemic, RNA drugs had made important inroads toward the amelioration of disease. The first class of RNA therapies to be introduced into clinical use were the antisense oligomers and siRNA drugs which generally induce a therapeutic effect by acting to brake or to modulate mRNA expression. RNA therapeutics is quickly becoming the fourth pillar of pharmacotherapy, and will have broad applications, including for the treatment of cardiovascular disease. RECENT FINDINGS The United States (US) Food and Drug Administration (FDA) has approved several antisense oligomers (ASOs) and siRNA-based drugs to treat disorders associated with cardiovascular disease. In addition, multiple RNA-based drugs are in clinical trials to assess their safety and efficacy in patients with cardiovascular disorders, such as Zodasiran, a siRNA therapy that targets angiopoietin-like protein 3 (ANGPTL3) to reduce LDL cholesterol. SUMMARY Because of limitless sequence choice; speed of design; and relative ease of synthesis, RNA drugs will be rapidly developed, will have broad applications, and will be generated at lower cost than other drug types. This review aims to highlight RNA therapies for cardiovascular diseases that are approved, and those that are under clinical evaluation.
Collapse
Affiliation(s)
- Tulsi Ram Damase
- Center for RNA Therapeutics, Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, Texas, USA
| | | |
Collapse
|
8
|
Sethi SK, Bradley CE, Bialkowski L, Pang YY, Thompson CD, Schiller JT, Çuburu N. Repurposing anti-viral subunit and mRNA vaccines T cell immunity for intratumoral immunotherapy against solid tumors. NPJ Vaccines 2025; 10:84. [PMID: 40280970 PMCID: PMC12032097 DOI: 10.1038/s41541-025-01131-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Intratumoral (IT) immunotherapy can stimulate the tumor microenvironment and enhance anti-tumor immunity. We investigated IT delivery of three licensed viral vaccines-Shingrix (VZV shingles), Gardasil-9 (HPV), and Spikevax (SARS-CoV-2)-in prevaccinated mice using the murine tumor model TC-1, which expresses HPV16 oncogenes E6 and E7. Shingrix IT injection often induced tumor regression and resistance to secondary challenge. Injecting a VZV glycoprotein E (gE)-derived MHC-II-restricted peptide with polyI:C also led to durable remission, highlighting the role of gE-specific CD4+ T cells. While Gardasil-9 IT injection alone was ineffective, combining a HPV L1-derived MHC-I-restricted peptide with polyI:C or Shingrix enhanced tumor regression. Both approaches elicited CD8+ T cells against the E7 tumor viral oncoprotein. Tumor microenvironment analysis revealed remodeling of the myeloid compartment, significant induction of IFN-γ, TNF-α, and CXCL9 and broad gene expression reprograming. In a dual-flank model, IT injection of Shingrix with an MHC-I-restricted E7 tumor-specific peptide eliminated primary and non-injected tumors. Finally, Spikevax IT injection showed modest tumor growth delay, while improved control was observed with a SARS-CoV-2 spike-derived MHC-I-restricted peptide and polyI:C. These results demonstrate the potential of licensed vaccines as promising platforms for IT immunotherapy, either alone or combined with vaccine- or tumor-derived MHC-I-restricted peptide epitopes.
Collapse
Affiliation(s)
- Shiv K Sethi
- National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Lukas Bialkowski
- National Cancer Institute, NIH, Bethesda, MD, USA
- Beckman Coulter, Bethesda, USA
| | | | | | | | | |
Collapse
|
9
|
Fierro C, Sanchez-Crespo N, Makrinos D, Zhang W, Sun Y, Rohilla P, Girard B, Adeniji A, DiPiazza A, Paris R. Shared clinical and immunologic features of mRNA vaccines: preliminary results from a comparative clinical study. Front Immunol 2025; 16:1501275. [PMID: 40276503 PMCID: PMC12018429 DOI: 10.3389/fimmu.2025.1501275] [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: 09/24/2024] [Accepted: 02/20/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Clinical trials do not typically assess underlying molecular mechanisms of vaccine immunogenicity or reactogenicity. We evaluated the reactogenicity and immunogenicity of 4 mRNA vaccines and potential contributing mechanisms and identified shared and unique clinical and immunologic features. Methods This ongoing, open-label, phase 1 trial randomized healthy adults (18-75 years) to receive a single dose of mRNA-1273.222 (bivalent COVID-19), mRNA-1345 (RSV), mRNA-1010 (influenza), and FLUAD (active influenza comparator) or 2 or 3 doses of mRNA-1647 (CMV). The primary objective was to assess the safety and reactogenicity of each study vaccine, with humoral immunogenicity (neutralizing antibody [nAb] responses) as the secondary objective. This interim analysis reports safety and reactogenicity in all study vaccines and humoral immunogenicity in single-dose vaccines (mRNA-1273.222, mRNA-1345, mRNA-1010, and FLUAD). Exploratory objectives included antigen-specific T-cell responses after single-dose mRNA-1345 or mRNA-1273.222, and soluble mediators of inflammation and innate immunity following vaccination in single-dose vaccine groups and two doses of mRNA-1647. Results At the interim analysis data cutoff (February 1, 2023), 302 participants received 1 dose of the study vaccines. Reactogenicity exhibited a consistent trend across vaccine groups; most solicited local and systemic adverse reactions within 7 days were mild or moderate in severity. There were no deaths or serious, severe, or treatment-related adverse events leading to study discontinuation. At Day 29, nAb titers against vaccine-specific antigens increased 2- to 8-fold versus baseline for all single-dose vaccine groups. In an exploratory analysis, mRNA-1273.222 and mRNA-1345 induced antigen-specific Th1-biased CD4+ and CD8+ T-cell responses at Day 29. The cytokine response analysis showed increased levels of IFN-γ, IL-6, IL-2Ra, CXCL9, IP-10, MCP-2, and MIP-1β on Day 2 following vaccination, with generally greater increases observed with mRNA vaccines versus FLUAD. Regardless of age and across mRNA vaccine groups, peak serum levels of IL-1Ra and MCP-1/MCP-2 on Day 2 weakly correlated with systemic reactogenicity scores (correlation coefficient range: 0.15-0.27). Conclusions The 4 mRNA vaccines had acceptable reactogenicity, demonstrated changes in serum biomarkers of innate immune activation, and were immunogenic. This suggests that the observed reactogenicity of mRNA vaccines may be related to shared features of the mRNA platform (LNP platform). Clinical trial registration ClinicalTrials.gov, identifier NCT05397223.
Collapse
Affiliation(s)
- Carlos Fierro
- Johnson County Clin-Trials, Clinical Research, Lenexa, KS, United States
| | | | | | | | - Yanbo Sun
- Moderna, Inc., Cambridge, MA, United States
| | | | | | | | | | | |
Collapse
|
10
|
Kunath P, Pflumm D, Moehrle B, Sakk V, Seidel A, Münch J, Geiger H, Schirmbeck R. Old hematopoietic stem cells retain competence to reconstitute a youthful B cell system that is highly responsive to protein-based vaccination. Immun Ageing 2025; 22:14. [PMID: 40188072 PMCID: PMC11971919 DOI: 10.1186/s12979-025-00507-x] [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: 01/20/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Ageing-associated remodeling of the murine B cell system is accompanied with a reduction of CD19+ B cells such as follicular B cells (FOB) and an accumulation of age-associated B cells (ABC) or activated B cell subsets. This remodeling is thought to confer an attenuated antibody response, such as to SARS-CoV-2 spike (S) vaccines in both aged mice and humans. To gain insight into the de novo development and function of an old B cell system, we reconstituted young and old immune systems by transferring hematopoietic stem cells (HSCs) from immune-competent young (2-3 months) CD45.1+ donors (DY-HSC) or old (20-24 months) donors (DO-HSC) into T and B cell-deficient young recipient CD45.2+ RAG1-/- mice, followed by protein-based vaccination. RESULTS In the same environment of young RAG1-/- mice, transplanted DO-HSCs compared to DY-HSCs reconstituted lower numbers of CD19+ B cells and CD45.1+ cells, though the engraftment of donor-derived HSCs in the young bone marrow (BM) was very similar. Furthermore, indicative for youthful and unchallenged B cell systems, and in contrast to aged mice, very low levels of antigen-experienced memory B cells or age-associated B cells (ABC) developed in both DY-HSC and DO-HSC hosts. The commercially available recombinant SARS-CoV-2 S vaccine (NVX-CoV2373) induced lower IgG+ S-antibody titers and pseudovirus neutralization activity in old compared to young mice. In contrast, very similar high IgG+ S-antibody titers were induced in DO-HSC and DY-HSC hosts, and pseudovirus neutralization activity was even enhanced in DO-HSC compared with DY-HSC hosts. CONCLUSIONS Both DO-HSCs and DY-HSCs established in the young recipient BM to a similar extend, suggesting that the concomitant reduction in the de novo reconstitution of CD19+ B cells in DO-HSC vs. DY-HSC transplanted animals is specifically related to old HSCs. DO-HSCs and DY-HSCs reconstitute very similar unchallenged B cell systems that efficiently elicit antigen-specific IgG antibodies by protein-based vaccination. Old HSCs thus retain competence to reconstitute a youthful and functional B cell system, at least in the young environment of transplanted RAG1-/- mice. This suggests that it is primarily age-related factors, and not HSCs per se, that influence the composition and functionality of the old B cell system.
Collapse
Affiliation(s)
- Paul Kunath
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany
| | - Dominik Pflumm
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany
| | - Bettina Moehrle
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Vadim Sakk
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Reinhold Schirmbeck
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany.
| |
Collapse
|
11
|
Foster T, Lim P, Wagle SR, Ionescu CM, Kovacevic B, McLenachan S, Carvalho L, Brunet A, Mooranian A, Al-Salami H. Nanoparticle-Based gene therapy strategies in retinal delivery. J Drug Target 2025; 33:508-527. [PMID: 39749456 DOI: 10.1080/1061186x.2024.2433563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/31/2024] [Accepted: 11/17/2024] [Indexed: 01/04/2025]
Abstract
Vision loss and blindness are significant issues in both developed and developing countries. There are a wide variety of aetiologies that can cause vision loss, which are outlined in this review. Although treatment has significantly improved over time for some conditions, nearly half of all people with vision impairment are left untreated. Gene delivery is an emerging field that may assist with the treatment of some of these difficult to manage forms of vision loss. Here we review how a component of nanotechnology-based, non-viral gene delivery systems are being applied to help resolve vision impairment. This review focuses on the use of lipid and polymer nanoparticles, and quantum dots as gene delivery vectors to the eye. Finally, we also highlight some emerging technologies that may be useful in this discipline.
Collapse
Affiliation(s)
- Thomas Foster
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Department of Clinical Biochemistry, Pathwest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Patrick Lim
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Samuel McLenachan
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), The University of Western Australia, Crawley, Western Australia, Australia
| | - Livia Carvalho
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), The University of Western Australia, Crawley, Western Australia, Australia
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Alicia Brunet
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), The University of Western Australia, Crawley, Western Australia, Australia
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- School of Pharmacy, University of Otago, Dunedin, Otago, New Zealand
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Medical School, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
12
|
Farnsworth CW, Roemmich B, Prostko J, Davis G, Murtagh G, Jackson L, Jacobson C, Jeanblanc N, Griffiths T, Frias E, Daghfal DJ. Systemic inflammation is associated with worse outcomes from SARS-CoV-2 infection but not neutralizing antibody. Microbiol Spectr 2025; 13:e0245924. [PMID: 39969178 PMCID: PMC11960045 DOI: 10.1128/spectrum.02459-24] [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: 09/30/2024] [Accepted: 12/16/2024] [Indexed: 02/20/2025] Open
Abstract
Systemic inflammation is associated with COVID-19 mortality rates, but the impact of inflammation on neutralizing antibodies to severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) and on outcomes is poorly understood. This study aimed to determine the association between neutralizing antibody responses, inflammation, and clinical outcomes in hospitalized patients with COVID-19. Two hundred and eight patients presenting to the ED with symptomatic SARS-CoV-2 were included. Neutralization was assessed using the architect angiotensin-converting enzyme-2 (ACE2) binding inhibition assay, and inflammation was assessed using C reactive protein (CRP) and interleukin 6 (IL-6). Medical records were examined for 30-day mortality and 10-day intubation. Correlation between biomarkers was assessed and Kaplan-Meier curves and Cox proportional hazards models were constructed for outcomes. Thirty-seven (18%) patients died and 59 (28%) required intubation. There was a correlation between IL-6 and CRP (r = 0.34) but not ACE-2 (r < 0.06). Patients that died had higher CRP (14 mg/dl, 8-21) than those that survived (5 mg/dl, 2-11) and IL-6 (died = 344 pg/ml, 138-870 vs. survived = 65 pg/ml, 28-140). ACE-2 inhibition trended higher in those who survived (18%, 0%-65%) than those who died (3%, 0%-48%). Patients with elevated IL-6, elevated CRP, or low ACE2 inhibition had higher mortality. Only IL-6 (hazard ratio: 1.28, 95% CI 1.08-1.52) and age (1.04, 1.01-1.08) were associated with mortality in multivariate models. Elevated IL-6 was associated with 30-day mortality from SARS-CoV-2 infection. Lower ACE-2 inhibition was not independently associated with mortality or correlated with inflammatory markers, implying the importance of other aspects of the immune response for reducing SARS-CoV-2 mortality risk.IMPORTANCEWhile systemic inflammation associated with worse outcomes from SARS-CoV-2 infection, it is not associated with neutralizing antibody concentrations, implying the importance of other aspects of the immune response for reducing SARS-CoV-2 mortality risk.
Collapse
Affiliation(s)
- Christopher W. Farnsworth
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Brittany Roemmich
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | - Edwin Frias
- Abbott Diagnostics, Abbott Park, Illinois, USA
| | | |
Collapse
|
13
|
Seitz I, Saarinen S, Wierzchowiecka J, Kumpula EP, Shen B, Cornelissen JJLM, Linko V, Huiskonen JT, Kostiainen MA. Folding of mRNA-DNA Origami for Controlled Translation and Viral Vector Packaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417642. [PMID: 40012449 DOI: 10.1002/adma.202417642] [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: 11/14/2024] [Revised: 01/28/2025] [Indexed: 02/28/2025]
Abstract
mRNA is an important molecule in vaccine development and treatment of genetic disorders. Its capability to hybridize with DNA oligonucleotides in a programmable manner facilitates the formation of RNA-DNA origami structures, which can possess a well-defined morphology and serve as rigid supports for mRNA delivery. However, to date, comprehensive studies on the requirements for efficient folding of mRNA into distinct mRNA-DNA structures while preserving its translation functionality remain elusive. Here, the impact of design parameters on the folding of protein-encoding mRNA into mRNA-DNA origami structures is systematically investigated and the importance of the availability of ribosome-binding sequences on the translation efficiency is demonstrated. Furthermore, these hybrid structures are encapsulated inside virus capsids resulting in protecting them against nuclease degradation and also in enhancement of their cellular uptake. This multicomponent system therefore showcases a modular and versatile nanocarrier. The work provides valuable insight into the design of mRNA-DNA origami structures contributing to the development of mRNA-based gene delivery platforms.
Collapse
Affiliation(s)
- Iris Seitz
- Department of Bioproducts and Biosystems, Aalto University, 00076, Aalto, Finland
| | - Sharon Saarinen
- Department of Bioproducts and Biosystems, Aalto University, 00076, Aalto, Finland
| | - Julia Wierzchowiecka
- Department of Bioproducts and Biosystems, Aalto University, 00076, Aalto, Finland
| | - Esa-Pekka Kumpula
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Boxuan Shen
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Jeroen J L M Cornelissen
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology, University of Twente, 7522, Enschede, The Netherlands
| | - Veikko Linko
- Department of Bioproducts and Biosystems, Aalto University, 00076, Aalto, Finland
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia
| | - Juha T Huiskonen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Mauri A Kostiainen
- Department of Bioproducts and Biosystems, Aalto University, 00076, Aalto, Finland
- LIBER Center of Excellence, Aalto University, 00076, Aalto, Finland
| |
Collapse
|
14
|
Mathur S, Chaturvedi A, Ranjan R. Advances in RNAi-based nanoformulations: revolutionizing crop protection and stress tolerance in agriculture. NANOSCALE ADVANCES 2025; 7:1768-1783. [PMID: 40046252 PMCID: PMC11877354 DOI: 10.1039/d5na00044k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/08/2025] [Indexed: 03/27/2025]
Abstract
Nucleic acid-based therapeutics have the ability to tackle a wide range of diseases and stress tolerance that present significant obstacles for conventional approaches in agriculture. RNA-based medicines have become a promising approach, using nanoformulation treatments to specifically target certain diseases. Nanoformulations offer numerous benefits in comparison to alternative treatment methods, such as precise administration, minimal toxicity, and medication loading compatibility due to their bioactivity. There are a variety of nanoformulations available today, such as liposomes, polymeric nanoparticles (NPs), magnetic NPs, nanogels, and solid lipid nanoparticles (SLNs). RNA-based therapy employs intracellular gene nanoparticles containing messenger RNA (mRNA), which play an important role in stress management and pest as well as disease control. The adoption of mRNA-based technology paves the way for future technological progress. This review focuses on elucidating the process underlying the development of RNA interference (RNAi) and the diverse array of nanocarriers employed for the transportation of RNAi. Currently, this technique is being employed in the field of crop protection to combat diseases, pests, and environmental stress. The article highlights the benefits of RNAi mediated nanoformulations and discusses the significant obstacles that must be overcome to improve the viability of this technology for future applications.
Collapse
Affiliation(s)
- Shivangi Mathur
- Plant Molecular Biology Lab, Department of Botany, Dayalbagh Educational Institute Dayalbagh Agra 282005 India
| | - Ambika Chaturvedi
- Plant Molecular Biology Lab, Department of Botany, Dayalbagh Educational Institute Dayalbagh Agra 282005 India
| | - Rajiv Ranjan
- Plant Molecular Biology Lab, Department of Botany, Dayalbagh Educational Institute Dayalbagh Agra 282005 India
| |
Collapse
|
15
|
Tang S, Huang L, Ge J, Li J, Qiu M, Zhang Y, Long M, Wu G, Zhang R, Ma X, Xia Q, Wan P, Yang T. Influence of salt solution on the physicochemical properties and in vitro/ in vivo expression of mRNA/LNP. J Nanobiotechnology 2025; 23:223. [PMID: 40108620 PMCID: PMC11921543 DOI: 10.1186/s12951-025-03318-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Lipid nanoparticles (LNPs) have revolutionized nucleic acid delivery, enabling significant advances in mRNA-based therapeutics. While extensive research has focused on lipid composition, the impact of preparation solutions on LNP performance remains underexplored. This study systematically investigated the effects of pH, salt type, and concentration across key preparation solutions-mRNA aqueous, dilution, exchange, and storage solutions-on the physicochemical properties, stability, and expression efficiency of SM102-based mRNA/LNPs. Findings revealed that the pH of the mRNA aqueous solution was critical, with a pH of 4 optimizing encapsulation efficiency (EE) and cellular expression. The exchange solution's pH significantly influenced biodistribution, particularly liver-specific expression following intravenous and intramuscular administration. Sucrose was identified as essential for freeze-thaw stability, with a 300 mM concentration minimizing aggregation and mRNA leakage. Furthermore, preparation solutions were shown to influence the structural integrity of LNPs, impacting their in vivo and in vitro performance. These insights highlight the importance of preparation conditions in optimizing LNP formulations for clinical applications, offering a foundation for enhanced therapeutic design and delivery.
Collapse
Affiliation(s)
- Siyuan Tang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lei Huang
- Micro & Nano Inc, Shanghai, 200127, China
| | - Jiahao Ge
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jie Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mingxia Qiu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yiqing Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mei Long
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Gang Wu
- Micro & Nano Inc, Shanghai, 200127, China
| | - Rui Zhang
- XGen Bio Inc, Woburn, MA, 01801, USA
| | - Xueyun Ma
- Laboratory Animal Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Shanghai Institute of Organ Transplantation, Shanghai, 200127, China.
- Shanghai Organ Transplantation and Immune Engineering Technology Research Center, Shanghai, 200127, China.
| | - Ping Wan
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Taihua Yang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
16
|
Fatima M, An T, Hong KJ. Revolutionizing mRNA Vaccines Through Innovative Formulation and Delivery Strategies. Biomolecules 2025; 15:359. [PMID: 40149895 PMCID: PMC11940278 DOI: 10.3390/biom15030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Modernization of existing methods for the delivery of mRNA is vital in advanced therapeutics. Traditionally, mRNA has faced obstacles of poor stability due to enzymatic degradation. This work examines cutting-edge formulation and emerging techniques for safer delivery of mRNA vaccines. Inspired by the success of lipid nanoparticles (LNP) in delivering mRNA vaccines for COVID-19, a variety of other formulations have been developed to deliver mRNA vaccines for diverse infections. The meritorious features of nanoparticle-based mRNA delivery strategies, including LNP, polymeric, dendrimers, polysaccharide-based, peptide-derived, carbon and metal-based, DNA nanostructures, hybrid, and extracellular vesicles, have been examined. The impact of these delivery platforms on mRNA vaccine delivery efficacy, protection from enzymatic degradation, cellular uptake, controlled release, and immunogenicity has been discussed in detail. Even with significant developments, there are certain limitations to overcome, including toxicity concerns, limited information about immune pathways, the need to maintain a cold chain, and the necessity of optimizing administration methods. Continuous innovation is essential for improving delivery systems for mRNA vaccines. Future research directions have been proposed to address the existing challenges in mRNA delivery and to expand their potential prophylactic and therapeutic application.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Timothy An
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
17
|
Amaral C, Lourenço EC, Morais I, Ascenso O, Aguiar SI, Grilo D, Ventura MR, Pimentel C. Synthetic Osmolytes for Enhanced RNA Thermotolerance and Extended Plasmid Storage at Room Temperature. Biotechnol J 2025; 20:e202400608. [PMID: 40059573 DOI: 10.1002/biot.202400608] [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: 10/11/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 05/13/2025]
Abstract
Stable formulations for RNA and plasmid DNA are a matter of paramount significance in several fields, ranging from medicine to biotechnology. We have investigated the potential of 15 compounds derived from natural osmolytes to enhance the thermostability and protection of both RNA and plasmid DNA. Our findings demonstrated that several compounds exhibit remarkable effects, enhancing the long-term storage of plasmid DNA at room temperature and the resilience of RNA to high-temperature stress, surpassing the performance of commercial osmolytes. Importantly, we found that one of the compounds enhanced the detection efficacy of a cost-effective RT-PCR test for COVID-19 that we had previously developed. This work offers new possibilities for expanding the capabilities of molecular diagnostic assays and nucleic acid storage methods.
Collapse
Affiliation(s)
- Catarina Amaral
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | - Inês Morais
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | - Sandra I Aguiar
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, Lisboa, Portugal
| | - Diana Grilo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - M Rita Ventura
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Pimentel
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
18
|
Zhao T, Wang Z, Tong M, Fei Y. The development of therapeutics and vaccines against COVID-19. Diagn Microbiol Infect Dis 2025; 111:116643. [PMID: 39637679 DOI: 10.1016/j.diagmicrobio.2024.116643] [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: 11/02/2024] [Revised: 11/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Since the COVID-19 pandemic, it has caused a great threat to the global economy and public health, initiatives have been launched to control the spread of the virus. To explore the efficacy of drugs, a large number of clinical trials have been carried out, with the purpose of providing guidelines based on high-quality evidence for clinicians. We mainly discuss therapeutic agents for COVID-19 and explain the mechanism, including antiviral agents, tocilizumab, Janus kinase (JAK) inhibitors, neutralizing antibody therapies and corticosteroids. In addition, the COVID-19 vaccine has been proven to be efficacious in preventing SARS-CoV-2 infection. We systematically analyzed four mainstream vaccine platforms: messenger RNA (mRNA) vaccines, viral vector vaccines, inactivated vaccines and protein subunit vaccines. We evaluated the therapeutic effects of drugs and vaccines through enumerating the most typical clinical trials. However, the emergence of novel variants has further complicated the interpretation of the available clinical data, especially vaccines and antibody therapies. In the post-epidemic era, therapeutic agents are still the first choice for controlling the progression of disease, whereas the protective effect of vaccines against different strains should be assessed comprehensively.
Collapse
Affiliation(s)
- Tianyu Zhao
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China
| | - Zhiwei Wang
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China
| | - Mingjiong Tong
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China
| | - Yingming Fei
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China.
| |
Collapse
|
19
|
Solchenberger H, Odendahl M, Schriefer D, Proschmann U, Rahbani GKA, Ziemssen T, Akgün K. Extensive T-Cell Profiling Following SARS-CoV-2 mRNA Vaccination in Multiple Sclerosis Patients Treated with DMTs. Pathogens 2025; 14:235. [PMID: 40137720 PMCID: PMC11944680 DOI: 10.3390/pathogens14030235] [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: 02/07/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Disease-modifying therapies (DMTs) are known to impact cellular and humoral immune response in persons with multiple sclerosis (pwMS). In this study, we performed in-depth SARS-CoV-2-specific T-cell profiling using flow cytometry. T-cell immunity in pwMS with or without DMTs was evaluated before a first SARS-CoV-2 messenger ribonucleic acid (mRNA) vaccination and at one-, two- and six-month follow-up. T-cell stimulation without SARS-CoV-2-specific antigens was used as a control. T-cell response was compared to B-cell response by evaluating SARS-CoV-2-specific antibodies. We observed an upregulation of specific subpopulations of SARS-CoV-2 spike-specific CD4+ T cells. Thus, our results demonstrate the induction of a broad and distinct CD4+ T-cell response in pwMS even on anti-CD20 treatment and sphingosine-1-phosphate receptor modulation after SARS-CoV-2 mRNA vaccination. This was particularly seen in CD4+high and CD4+CD154+ T cells. Our results do not support the induction of a CD8+ T-cell immune response. While humoral immune response was impaired in pwMS during ocrelizumab and fingolimod treatment, there was evidence of a compensatory upregulation of subpopulations of SARS-CoV-2-specific CD4+ T cells at low levels of seroconversion in pwMS. In conclusion, our results provide important insights into the mechanisms of the adaptive immune response in pwMS following SARS-CoV-2 mRNA vaccination.
Collapse
Affiliation(s)
- Hannah Solchenberger
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (H.S.); (D.S.); (U.P.); (G.K.a.R.); (T.Z.)
| | - Marcus Odendahl
- Medical Faculty Carl Gustav Carus, Technical University Dresden, Experimental Transfusion Medicine, 01307 Dresden, Germany;
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, 01307 Dresden, Germany
| | - Dirk Schriefer
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (H.S.); (D.S.); (U.P.); (G.K.a.R.); (T.Z.)
| | - Undine Proschmann
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (H.S.); (D.S.); (U.P.); (G.K.a.R.); (T.Z.)
| | - Georges Katoul al Rahbani
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (H.S.); (D.S.); (U.P.); (G.K.a.R.); (T.Z.)
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (H.S.); (D.S.); (U.P.); (G.K.a.R.); (T.Z.)
| | - Katja Akgün
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (H.S.); (D.S.); (U.P.); (G.K.a.R.); (T.Z.)
| |
Collapse
|
20
|
Khan A, Zhu Y, Babcock HM, Busse LW, Duggal A, Exline MC, Gaglani M, Gibbs KW, Gong MN, Ginde AA, Hager DN, Hope AA, Hyde J, Johnson NJ, Kwon JH, Mohr NM, O'Rourke M, Peltan ID, Mallow C, Qadir N, Reddy R, Safdar B, Shapiro NI, Sohn I, Steingrub JS, Wilson JG, Baughman A, Womack KN, Rhoads JP, Self WH, Stubblefield WB. COVID-19 and influenza vaccine Hesitancy among adults hospitalized in the United States, 2019-2022. Vaccine 2025; 48:126806. [PMID: 39884913 DOI: 10.1016/j.vaccine.2025.126806] [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: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Understanding similarities and differences between hesitancy for influenza and COVID-19 vaccines could facilitate strategies to improve public receptivity toward vaccination. METHODS We compared hesitancy for COVID-19 vaccines during the first 13 months of availability (January 2021-January 2022) with hesitancy for influenza vaccines in the 15 months prior to COVID-19 vaccine availability (October 2019-December 2020) among adults hospitalized with acute respiratory illness at 21 hospitals in the United States. We interviewed patients regarding vaccination status, willingness to be vaccinated, and perceptions of vaccine safety and efficacy. We used multivariate logistic regression to identify factors associated with vaccine hesitancy. RESULTS Among 12,292 patients enrolled during the COVID-19 vaccine period, 5485 (44.6 %) were unvaccinated. Patient characteristics associated with not receiving the COVID-19 vaccine included younger age, female sex, higher BMI, lack of health insurance, absence of chronic comorbid medical conditions, no or rare influenza vaccination in prior years, higher CDC social vulnerability index (SVI), a measure of external stresses that may negatively impact health, living in the Midwest or southern US, lack of college or higher education, and not wearing a mask. Among 983 patients enrolled during the influenza vaccination period, 381(37.8 %) were unvaccinated. Characteristics associated with not receiving the influenza vaccine included no or one chronic comorbid medical condition, no or rare influenza vaccination in prior years, being a current smoker, and higher SVI. Discussion with healthcare providers was a reason for vaccination for 27.7 % (167) for influenza and 8.3 % (564) for COVID-19 and to decline vaccination for 0.5 % Ten great public health achievements-United States (2011) (2) for influenza and 2.2 % (118) for COVID-19. CONCLUSIONS We found that higher SVI scores and lack of prior influenza vaccination were associated with hesitancy for both COVID-19 and influenza vaccines. There were regional variations in COVID-19 vaccine acceptance and discussions with HCPs significantly influenced acceptance for both vaccines.
Collapse
Affiliation(s)
- Akram Khan
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Oregon Health and Sciences University, Portland, OR, USA.
| | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Hilary M Babcock
- Department of Medicine, Washington University in St. Louis, MO, USA.
| | - Laurence W Busse
- Department of Medicine, Emory University, Atlanta, Georgia Emory Critical Care Center, Emory Healthcare, Atlanta, GA, USA.
| | - Abhijit Duggal
- Department of Critical Care, Integrated Hospital Care Institute. Cleveland Clinic, Cleveland, OH, USA.
| | - Matthew C Exline
- Department of Medicine, The Ohio State University, Columbus, OH, USA.
| | - Manjusha Gaglani
- Baylor Scott and White Health, Temple and Dallas, Texas, and Texas A&M University College of Medicine, Temple, TX, USA.
| | - Kevin W Gibbs
- Department of Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Michelle N Gong
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Adit A Ginde
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| | - David N Hager
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Aluko A Hope
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Oregon Health and Sciences University, Portland, OR, USA.
| | - Jessica Hyde
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Oregon Health and Sciences University, Portland, OR, USA.
| | - Nicholas J Johnson
- Department of Emergency Medicine and Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA.
| | - Jennie H Kwon
- Department of Medicine, Washington University, St. Louis, MO, USA.
| | | | - Mary O'Rourke
- Department of Emergency Medicine, Hennepin County Medical Center, Minneapolis, MN, USA.
| | - Ithan D Peltan
- Department of Medicine, Intermountain Medical Center, Murray, Utah and University of Utah, Salt Lake City, UT, USA.
| | - Christopher Mallow
- Department of Medicine, University of Miami, Miami, Flo Department of Medicine, University of Miami, Miami, FL, USA.
| | - Nida Qadir
- Department of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
| | - Raju Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Oregon Health and Sciences University, Portland, OR, USA.
| | - Basmah Safdar
- Department of Emergency Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Nathan I Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Ine Sohn
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jay S Steingrub
- Department of Medicine, Baystate Medical Center, Springfield, MA, USA.
| | - Jennifer G Wilson
- Department of Emergency Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Adrienne Baughman
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Kelsey N Womack
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jillian P Rhoads
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Wesley H Self
- Vanderbilt Institute for Clinical and Translational Research, and Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William B Stubblefield
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
21
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
22
|
Nag R, Srivastava S, Rizvi S, Ahmed S, Raza ST. Innovations in vaccine design: Computational tools and techniques. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:375-391. [PMID: 40175050 DOI: 10.1016/bs.apha.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
The advancements in computational tools have revolutionized vaccine development by organizing and analyzing large-scale immunological data through immuno-informatics. This field combines computational and mathematical approaches to model molecular interactions during antigen presentation and processing. These tools have significantly accelerated vaccine development, making it more efficient and cost-effective. Applications such as SCWRL and SCAP help in side chain and backbone modeling to improve antibodies and forecast secondary structures. Multi-graft and multivalent scaffolds present antigens to elicit strong immune responses; antibodyomics studies the sequences of antibodies to find antibodies that can neutralize. It is another traditional way of doing vaccines where the pathogen's genome is scanned by diacide such as Vaxign to identify the likely vaccine agents. Codon optimization, as implemented with the aid of COOL and OPTIMIZER tools, enhances the output of proteins among which vaccines are needed. These tools also allow for predicting epitope structures the more accurately, or so. Prediction tools that include immunogenicity screening tests that map B-cell epitope and T-cell epitope such as ElliPro and DiscoTope aid in drug design, while the application of Fusion technologies facilitates vaccine development and kit diagnostics. The percentage of time trying to identify possible vaccine candidates is reduced alongside the costs with the application of these tools allowing the improvement in the prediction of vaccine candidates. The purpose of this chapter is to emphasize the invention of computational tools and methods that together are revolutionizing vaccine design and development and to underline the importance of tissue engineering and immunology advances.
Collapse
Affiliation(s)
- Riya Nag
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Sanchita Srivastava
- Department of Biotechnology, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Saliha Rizvi
- Department of Biotechnology, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Samar Ahmed
- Department of Clinical Science, Dubai Medical College of Girls, Dubai, United Arab Emirates
| | - Syed Tasleem Raza
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India.
| |
Collapse
|
23
|
Fatima M, Park PG, Hong KJ. Clinical advancements in mRNA vaccines against viral infections. Clin Immunol 2025; 271:110424. [PMID: 39734036 DOI: 10.1016/j.clim.2024.110424] [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: 11/18/2024] [Revised: 12/15/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
Over the last decade, mRNA vaccines development has shown significant advancement, particularly during the COVID-19 pandemic. This comprehensive review examines the efficacy of pivotal vaccines against emerging COVID-19 variants and strategies for enhancing vaccine effectiveness. It also explores the versatility of mRNA technology in addressing other infectious diseases such as influenza, respiratory syncytial virus, HIV, cytomegalovirus, Ebola, Zika, Rabies, and Nipah viruses. The analysis includes safety and clinical progress of mRNA vaccines and evaluates their potential in combination vaccine strategies. Additionally, it addresses challenges related to delivery and scalability while highlighting opportunities for future advancements in the field. Recent advances in mRNA optimization, biomaterial-based delivery and thermostable designs offer promising solutions. It is essential to gain insights into the evolving landscape of mRNA vaccine technology to maximize its vital role in addressing diverse viral threats, advancing vaccinology and enhancing public health preparedness for future pandemic.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Pil-Gu Park
- Department of Microbiology, Gachon University College of Medicine, Incheon, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea; Korea mRNA Vaccine Initiative, Gachon University, Seongnam, Republic of Korea.
| |
Collapse
|
24
|
Yi L, Zhang Z, He T, Li Y, Yao W, Xie G, Li W. Improving lipid nanoparticles delivery efficiency of macrophage cells by using immunomodulatory small molecules. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 64:102809. [PMID: 39904398 DOI: 10.1016/j.nano.2025.102809] [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: 02/21/2024] [Revised: 07/13/2024] [Accepted: 01/09/2025] [Indexed: 02/06/2025]
Abstract
Nucleic acid drug delivery remains a key challenge in the development of nucleic acid therapy. How to improve the efficiency of nucleic acid delivery is still an important strategy for Lipofectamine 3000 and LNP development. Here, we screened 248 inhibitors or agonists related to immune modulation and identified three small molecules (BP-1-102, SCH58261, and Bropirimine) that could enhance the transfection efficiency to 2-fold to 5-fold of Lipofectamine 3000 and LNP, all of which are currently approved for clinical use in the treatment of the most common malignant tumors. In addition, we used high-throughput RNA sequencing technology to analyze the mechanisms and found that they were mainly associated with the receptor-mediated endocytosis pathway. Our findings have yielded novel insights that can contribute to the advancement of nucleic acid drugs, enhancing both their efficacy and precision in targeting cancer therapy.
Collapse
Affiliation(s)
- Lirong Yi
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226000, China
| | - Ziyan Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226000, China
| | - Tian He
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226000, China
| | - Yating Li
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226000, China
| | - Weiwei Yao
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
| | - Gangcai Xie
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226000, China.
| | - Wenqing Li
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226000, China.
| |
Collapse
|
25
|
Márquez-Escobar VA, Alonso-Cerda MJ, Rosales-Mendoza S, Betancourt-Mendiola MDL. Evaluation of Zein Nanoparticles as Delivery Agents of SARS-CoV-2 Antigens. Vaccines (Basel) 2025; 13:139. [PMID: 40006685 PMCID: PMC11860552 DOI: 10.3390/vaccines13020139] [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: 12/20/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Nanovaccines have significant potential to enhance immunization strategies by improving efficacy, safety, and cost-effectiveness. In particular, organic nanoparticles hold promise for the generation of low-cost nanovaccines obtained by environmentally friendly methods. In this study, the feasibility of using zein nanoparticles (NPs) as carriers for an antigenic peptide (p30) and the receptor binding domain (RBD) from SARS-CoV-2 spike protein was explored. METHODS A synthesis method for zein NPs was established by combining previously reported techniques, and the resulting NPs were characterized in terms of morphology, particle size, polydispersity index (PDI), surface charge, and colloidal stability using dynamic light scattering (DLS) and transmission electron microscopy (TEM). Tween 20 was employed as a surfactant to enhance particle stability and prevent aggregation. RESULTS The zein NPs were deemed safe based on an in vitro cytotoxicity assay using Vero cells. Immunogenicity assessments demonstrated that zein NPs:p30 and zein NPs:RBD induced IgG responses in test mice, whose magnitude was comparable to those achieved with alum as an adjuvant. CONCLUSIONS These findings support the use of zein NPs as promising vaccine delivery vehicles with adjuvant effects due to their ease and environmentally friendly synthesis, high stability, and low cost.
Collapse
Affiliation(s)
- Verónica Araceli Márquez-Escobar
- Biotechnology Section, Center for Research in Health Science and Biomedicine, Autonomous University of San Luis Potosí, Av. Sierra Leona 550, Lomas de San Luis, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (M.J.A.-C.)
- Recombinant Biopharmaceuticals Laboratory, School of Chemical Sciences, Autonomous University of San Luis Potosí, Manuel Nava 6, Av. Dr. Manuel Nava, San Luis Potosí 78210, Mexico
| | - María José Alonso-Cerda
- Biotechnology Section, Center for Research in Health Science and Biomedicine, Autonomous University of San Luis Potosí, Av. Sierra Leona 550, Lomas de San Luis, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (M.J.A.-C.)
- Recombinant Biopharmaceuticals Laboratory, School of Chemical Sciences, Autonomous University of San Luis Potosí, Manuel Nava 6, Av. Dr. Manuel Nava, San Luis Potosí 78210, Mexico
| | - Sergio Rosales-Mendoza
- Biotechnology Section, Center for Research in Health Science and Biomedicine, Autonomous University of San Luis Potosí, Av. Sierra Leona 550, Lomas de San Luis, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (M.J.A.-C.)
- Recombinant Biopharmaceuticals Laboratory, School of Chemical Sciences, Autonomous University of San Luis Potosí, Manuel Nava 6, Av. Dr. Manuel Nava, San Luis Potosí 78210, Mexico
| | - María de Lourdes Betancourt-Mendiola
- Biotechnology Section, Center for Research in Health Science and Biomedicine, Autonomous University of San Luis Potosí, Av. Sierra Leona 550, Lomas de San Luis, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (M.J.A.-C.)
- Recombinant Biopharmaceuticals Laboratory, School of Chemical Sciences, Autonomous University of San Luis Potosí, Manuel Nava 6, Av. Dr. Manuel Nava, San Luis Potosí 78210, Mexico
| |
Collapse
|
26
|
Al-Mana NM, Zareef TA, Albathi FA, Awney HA, Baeshen F, Abdullah R. Exploring lifestyle and dietary pattern shifts among Saudi adults during COVID-19 pandemic: insights from a cross-sectional examination. Front Nutr 2025; 11:1489160. [PMID: 39834457 PMCID: PMC11743356 DOI: 10.3389/fnut.2024.1489160] [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: 08/31/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Background Since the emergence of COVID-19 and the subsequent imposition of lockdown and movement restrictions, the world has witnessed fundamental lifestyle changes including alterations in dietary patterns and food consumption habits. Here, we investigated how the COVID-19 lockdown impacted dietary patterns and eating behaviors in the Saudi population. Methodology This cross-sectional study enrolled 427 participants aged 18 years or more, with 258 of them completing the survey. The survey included questions about demographic and dietary patterns during the COVID-19 lockdown. Data were collected and dietary behaviors before and during the lockdown in Jeddah, Saudi Arabia, were analyzed. Results The number of participants who considered lunch as their primary meal significantly decreased (p < 0.001) during the COVID-19 lockdown (74%), compared to before it (86%). By contrast, the number of participants who considered dinner as their primary meal remained almost unchanged (p = 0.079) during (79.1%) and before (84.1%) the lockdown. However, snack consumption significantly increased (p < 0.001) while fast-food consumption significantly decreased (p < 0.01) during the lockdown period. Our results also revealed a significant increase (p < 0.01) in water and coffee intake during the lockdown, with a significant rise in dessert consumption (p < 0.01). Conclusion Our results demonstrate that the COVID-19 lockdown caused a marked shift in dietary patterns and eating behaviors among the Saudi population. Notable changes were observed in overall food preferences after the lockdowns were imposed, with reduced consumption of fast foods and increased fluid intake.
Collapse
Affiliation(s)
- Najlaa M. Al-Mana
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Tahani A. Zareef
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Jeddah, Saudi Arabia
| | - Fatmah A. Albathi
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Hala A. Awney
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Farah Baeshen
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Renad Abdullah
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| |
Collapse
|
27
|
Binici B, Borah A, Watts JA, McLoughlin D, Perrie Y. The influence of citrate buffer molarity on mRNA-LNPs: Exploring factors beyond general critical quality attributes. Int J Pharm 2025; 668:124942. [PMID: 39537041 DOI: 10.1016/j.ijpharm.2024.124942] [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: 09/03/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Lipid nanoparticles (LNPs) are crucial in delivering mRNA vaccines and therapeutics. The properties of LNPs can be influenced by the choice of lipids and the manufacturing conditions, such as mixing parameters, lipid concentration, and the type and concentration of the aqueous buffer used. In this study, we investigated the impact of the citrate buffer molarity, the buffer commonly used to dissolve mRNA in the preparation of mRNA-LNPs. We prepared SM-102 LNPs containing firefly luciferase mRNA using citrate buffers at molarities of 50 mM, 100 mM, or 300 mM. Our findings revealed that varying the molarity of the citrate buffer did not significantly affect the particle size when considering the average diameter (z-average or Mode). All formulations exhibited low polydispersity index (PDI) and high encapsulation efficiency. Detailed analysis of particle size sub-populations (D10, D50, and D90) and morphology indicated that citrate buffer concentration might influence lipid packing during LNP production, though these differences were subtle. However, using higher citrate molarity (300 mM) to produce LNPs notably reduced cellular internalisation and in vitro transfection efficiency. This trend was also observed in vivo, where similar expression levels were noted in mice receiving the 50 mM and 100 mM LNP formulations, but lower expression was seen for the 300 mM formulation. Our study highlights the importance of buffer molarity in the aqueous phase during mRNA-based LNP preparation and that generally reported critical quality attributes (CQAs) for LNPs may not detect subtle formulation differences.
Collapse
Affiliation(s)
- Burcu Binici
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral St, University of Strathclyde, Glasgow, Scotland, G4 0RE, UK
| | - Ankita Borah
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral St, University of Strathclyde, Glasgow, Scotland, G4 0RE, UK
| | - Julie A Watts
- School of Pharmacy, University of Nottingham, University Park, Nottingham, England NG7 2RD, UK
| | - Daragh McLoughlin
- Center for Process Innovation, The Coxon Building, John Walker Road, Sedgefield, England TS21 3FE, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral St, University of Strathclyde, Glasgow, Scotland, G4 0RE, UK.
| |
Collapse
|
28
|
Ursino M, Villacampa G, Rekowski J, Dimairo M, Solovyeva O, Ashby D, Berlin J, Boix O, Calvert M, Chan AW, Coschi CH, Evans TRJ, Garrett-Mayer E, Golub RM, Guo C, Hayward KS, Hopewell S, Isaacs JD, Ivy SP, Jaki T, Kholmanskikh O, Kightley A, Lee S, Liu R, Mander A, Marshall LV, Matcham J, Patel D, Peck R, Rantell KR, Richards DP, Rouhifard M, Seymour L, Tanaka Y, Weir CJ, de Bono J, Yap C. SPIRIT-DEFINE explanation and elaboration: recommendations for enhancing quality and impact of early phase dose-finding clinical trials protocols. EClinicalMedicine 2025; 79:102988. [PMID: 39877554 PMCID: PMC11773215 DOI: 10.1016/j.eclinm.2024.102988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/31/2025] Open
Abstract
Transparent and accurate reporting in early phase dose-finding (EPDF) clinical trials is crucial for informing subsequent larger trials. The SPIRIT statement, designed for trial protocol content, does not adequately cover the distinctive features of EPDF trials. Recent findings indicate that the protocol contents in past EPDF trials frequently lacked completeness and clarity. To address this gap, the international consensus-driven SPIRIT-DEFINE checklist was developed through a robust methodological framework for guideline development, with the aim to improve completeness and clarity in EPDF trial protocols. The checklist builds on the SPIRIT statement, adding 17 new items and modifying 15 existing ones.The SPIRIT-DEFINE explanation and elaboration (E&E) document provides comprehensive information to enhance understanding and usability of the SPIRIT-DEFINE checklist when writing an EPDF trial protocol. Each new or modified checklist item is accompanied by a detailed description, its rationale with supportive evidence, and examples of good reporting curated from EPDF trial protocols covering a range of therapeutic areas and interventions. We recommend utilising this paper alongside the SPIRIT statement, and any relevant extensions, to enhance the development and review of EPDF trial protocols.By facilitating adoption of the SPIRIT-DEFINE statement for EPDF trials, this E&E document can promote enhancement of methodological rigour, patient safety, transparency, and facilitate the generation of high-quality, reproducible evidence that will strengthen the foundation of early phase research and ultimately improve patient outcomes. Funding This work is a further extension of the SPIRIT-DEFINE study, which obtained no external funding. The principal investigator (CY) used internal staff resources, together with additional resources from external partners, to conduct this study. The SPIRIT-DEFINE study is a component of the DEFINE project, which also developed the MRC/NIHR funded CONSORT-DEFINE guidance. ICR-CTSU receives programmatic infrastructure funding from Cancer Research UK (C1491/A25351; CTUQQR-Dec22/100004), which has contributed to accelerating the advancement and successful completion of this work.
Collapse
Affiliation(s)
- Moreno Ursino
- ReCAP/F CRIN, INSERM, 5400, Nancy, France
- Unit of Clinical Epidemiology, University Hospital Centre Robert Debré, Université Paris Cité, Paris, France
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- HeKA Team, Centre Inria, Paris, France
| | - Guillermo Villacampa
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, UK
- Statistics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Jan Rekowski
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, UK
| | - Munyaradzi Dimairo
- Division of Population Health, Sheffield Centre for Health and Related Research, University of Sheffield, Sheffield, UK
| | - Olga Solovyeva
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, UK
| | - Deborah Ashby
- School of Public Health, Imperial College London, St Mary's Hospital, London, UK
| | | | | | - Melanie Calvert
- Centre for Patient Reported Outcomes Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Applied Research Collaboration West Midlands, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Precision Transplant and Cellular Therapeutics, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Birmingham Biomedical Research Centre, NIHR Birmingham Biomedical Research Centre, Institute of Translational Medicine, University Hospital NHS Foundation Trust, Birmingham, UK
| | - An-Wen Chan
- Department of Medicine, Women's College Research Institute, University of Toronto, Toronto, Canada
| | | | - Thomas R. Jeffry Evans
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Elizabeth Garrett-Mayer
- Center for Research and Analytics, American Society of Clinical Oncology, Alexandria, VA, USA
| | - Robert M. Golub
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Christina Guo
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Kathryn S. Hayward
- Departments of Physiotherapy and Medicine, University of Melbourne, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Sally Hopewell
- Oxford Clinical Research Unit, NDORMS, University of Oxford, Oxford, UK
| | - John D. Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Freeman Hospital, Newcastle upon Tyne, UK
| | - S. Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Institute of Health, Bethesda, MD, USA
| | - Thomas Jaki
- MRC Biostatistics Unit, Cambridge University, Cambridge, UK
- Computational Statistics Group, University of Regensburg, Regensburg, Germany
| | | | - Andrew Kightley
- Patient and Public Involvement and Engagement (PPIE) Lead, Lichfield, UK
| | - Shing Lee
- Columbia University Mailman School of Public Health, New York, NY, USA
| | | | - Adrian Mander
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Lynley V. Marshall
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - James Matcham
- Strategic Consulting, Cytel (Australia), Perth, WA, Australia
| | - Dhrusti Patel
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, UK
| | - Richard Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Hoffmann-La Roche, Basel, Switzerland
| | | | | | - Mahtab Rouhifard
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, UK
| | | | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Christopher J. Weir
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Johann de Bono
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Christina Yap
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, UK
| |
Collapse
|
29
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore IN, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. PLoS Pathog 2025; 21:e1012456. [PMID: 39847599 PMCID: PMC11793774 DOI: 10.1371/journal.ppat.1012456] [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: 07/31/2024] [Revised: 02/04/2025] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather than the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n = 6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Sarah E. Scheuermann
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Ian N. Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
30
|
Rekowski J, Guo C, Solovyeva O, Dimairo M, Rouhifard M, Patel D, Alger E, Ashby D, Berlin J, Boix O, Calvert M, Chan AW, Coschi CH, de Bono J, Evans TRJ, Garrett–Mayer E, Golub RM, Hayward KS, Hopewell S, Isaacs JD, Ivy SP, Jaki T, Kholmanskikh O, Kightley A, Lee S, Liu R, Maia I, Mander A, Marshall LV, Matcham J, Peck R, Rantell KR, Richards DP, Seymour L, Tanaka Y, Ursino M, Weir CJ, Yap C. CONSORT-DEFINE explanation and elaboration: recommendations for enhancing reporting quality and impact of early phase dose-finding clinical trials. EClinicalMedicine 2025; 79:102987. [PMID: 39877553 PMCID: PMC11773258 DOI: 10.1016/j.eclinm.2024.102987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/31/2025] Open
Abstract
Early phase dose-finding (EPDF) trials are key in the development of novel therapies, with their findings directly informing subsequent clinical development phases and providing valuable insights for reverse translation. Comprehensive and transparent reporting of these studies is critical for their accurate and critical interpretation, which may improve and expedite therapeutic development. However, quality of reporting of design characteristics and results from EPDF trials is often variable and incomplete. The international consensus-based CONSORT-DEFINE (Consolidated Standards for Reporting Trials Dose-finding Extension) statement, an extension of the CONSORT statement for randomised trials, was developed to improve the reporting of EPDF trials. The CONSORT-DEFINE statement introduced 21 new items and modified 19 existing CONSORT items.This CONSORT-DEFINE Explanation and Elaboration (E&E) document provides important information to enhance understanding and facilitate the implementation of the CONSORT-DEFINE checklist. For each new or modified checklist item, we provide a detailed description and its rationale with supporting evidence, and present examples from EPDF trial reports published in peer-reviewed scientific journals. When reporting the results of EPDF trials, authors are encouraged to consult the CONSORT-DEFINE E&E document, together with the CONSORT and CONSORT-DEFINE statement papers, and adhere to their recommendations. Widespread adoption of the CONSORT-DEFINE statement is likely to enhance the reporting quality of EPDF trials, thus facilitating the peer review of such studies and their appraisal by researchers, regulators, ethics committee members, and funders. Funding This work is a further extension of the CONSORT-DEFINE study, which was funded by the UK Medical Research Council (MRC)-National Institute for Health and Care Research (NIHR) Methodology Research Programme (MR/T044934/1). The Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU) receives programmatic infrastructure funding from Cancer Research UK (C1491/A25351; CTUQQR-Dec 22/100 004), which has contributed to accelerating the advancement and successful completion of this work.
Collapse
Affiliation(s)
- Jan Rekowski
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Christina Guo
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Olga Solovyeva
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Munyaradzi Dimairo
- Division of Population Health, Sheffield Centre for Health and Related Research, University of Sheffield, Sheffield, UK
| | - Mahtab Rouhifard
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Dhrusti Patel
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Emily Alger
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Deborah Ashby
- School of Public Health, Imperial College London, St Mary's Hospital, London, UK
| | | | | | - Melanie Calvert
- Centre for Patient Reported Outcomes Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Applied Research Collaboration West Midlands, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Precision Transplant and Cellular Therapeutics, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Birmingham Biomedical Research Centre, NIHR Birmingham Biomedical Research Centre, Institute of Translational Medicine, University Hospital NHS Foundation Trust, Birmingham, UK
| | - An-Wen Chan
- Department of Medicine, Women's College Research Institute, University of Toronto, Toronto, Canada
| | | | - Johann de Bono
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Thomas R. Jeffry Evans
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Elizabeth Garrett–Mayer
- Center for Research and Analytics, American Society of Clinical Oncology, Alexandria, VA, USA
| | - Robert M. Golub
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathryn S. Hayward
- Departments of Physiotherapy and Medicine, University of Melbourne, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Sally Hopewell
- Oxford Clinical Research Unit, NDORMS, University of Oxford, Oxford, UK
| | - John D. Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Freeman Hospital, Newcastle upon Tyne, UK
| | - S. Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Institute of Health, Bethesda, MD, USA
| | - Thomas Jaki
- MRC Biostatistics Unit, Cambridge University, Cambridge, UK
- Computational Statistics Group, University of Regensburg, Regensburg, Germany
| | | | - Andrew Kightley
- Patient and Public Involvement and Engagement (PPIE) Lead, Lichfield, UK
| | - Shing Lee
- Columbia University Mailman School of Public Health, New York, NY, USA
| | | | | | - Adrian Mander
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Lynley V. Marshall
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - James Matcham
- Strategic Consulting, Cytel (Australia), Perth, WA, Australia
| | - Richard Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Hoffmann-La Roche, Basel, Switzerland
| | | | | | | | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Moreno Ursino
- ReCAP/F CRIN, INSERM, 5400, Nancy, France
- Unit of Clinical Epidemiology, University Hospital Centre Robert Debré, Université Paris Cité, Paris, France
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- HeKA Team, Centre Inria, Paris, France
| | - Christopher J. Weir
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Christina Yap
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| |
Collapse
|
31
|
Isa F, Gonzalez Ortiz AM, Meyer J, Hamilton JD, Olenchock BA, Brackin T, Ganguly S, Forleo-Neto E, Faria L, Heirman I, Marovich M, Hutter J, Polakowski L, Irvin SC, Thakur M, Hooper AT, Baum A, Petro CD, Fakih FA, McElrath MJ, De Rosa SC, Cohen KW, Williams LD, Hellman CA, Odeh AJ, Patel AH, Tomaras GD, Geba GP, Kyratsous CA, Musser B, Yancopoulos GD, Herman GA. Effect of timing of casirivimab and imdevimab administration relative to mRNA-1273 COVID-19 vaccination on vaccine-induced SARS-CoV-2 neutralising antibody responses: a prospective, open-label, phase 2, randomised controlled trial. THE LANCET. INFECTIOUS DISEASES 2025; 25:52-67. [PMID: 39236733 DOI: 10.1016/s1473-3099(24)00421-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Deeper insight is needed on how monoclonal antibodies (mAbs) affect vaccine-mediated immune responses when targeting the same protein. We describe the first prospective randomised trial designed to understand mAb-mediated alterations in vaccine-induced immune responses to SARS-CoV-2 spike protein epitopes. METHODS This randomised, open-label, parallel-group study assessed the potential interaction of a mAb combination, casirivimab and imdevimab, with a vaccine, Moderna's mRNA-1273, in healthy SARS-CoV-2 immunologically naive, seronegative adults at six centres in the USA. Participants were randomly assigned (per prespecified randomisation ratios within enrolment waves) according to a computer-generated randomisation scheme, stratified by age (<65 years and ≥65 years), to various intravenous or subcutaneous doses of casirivimab and imdevimab before, after, or at the same time as mRNA-1273 or to mRNA-1273 only. The doses of casirivimab and imdevimab were chosen to mimic various time intervals between receipt of 1200 mg of the mAb and the first dose of a primary series with mRNA-1273. The primary endpoint was vaccine-induced 50% inhibitory dilution neutralising antibody titres to SARS-CoV-2 spike protein, 56 days after the first vaccination. Secondary endpoints included vaccine-induced total antibodies to SARS-CoV-2 antigens and incidence of treatment-emergent adverse events. Exploratory endpoints included blood-derived T-cell and B-cell responses. The per-protocol set was used for the analysis of the primary endpoint and included all randomly assigned participants who received both doses of the vaccine and completed the injection or infusion of casirivimab and imdevimab per protocol, had no evidence of SARS-CoV-2 infection in the past or in the 56 days after the first dose of vaccine, and did not receive any intervention outside of the study that could alter the immune response. Safety was assessed in the safety analysis set, which included all randomly assigned participants who had received one or more doses of mRNA-1273 or any study drug, and analysed based on treatment received. The study is registered with ClinicalTrials.gov, NCT04852978, and is complete. FINDINGS Between April 29, 2021, and Nov 21, 2022, 807 participants were assessed for eligibility and 295 were randomly assigned. 293 participants were included in the safety analysis set and 260 were included in the per-protocol set. All vaccinated participants developed neutralising antibodies to SARS-CoV-2, with median titres above the published protective threshold (100 IU/mL) against the SARS-CoV-2 D614G variant (considered a reference strain at the time the initial COVID-19 vaccines were developed). Titres were decreased up to 4-fold (median titres 280-450 IU/mL for casirivimab and imdevimab vs 1160 IU/mL for vaccine only on day 56) when casirivimab and imdevimab was given 85 days or less before vaccination (150-1200 mg intravenously) or co-administered subcutaneously (600 mg or 1200 mg) with vaccination. Minimal reduction in neutralisation titres was observed in the 48 mg and 12 mg intravenous groups, corresponding to receipt of casirivimab and imdevimab 113 days and 169 days, respectively, before vaccination, and when administering the vaccine 6 days before the mAb. Across all groups, mAbs had a minimal effect on vaccine-induced total antibodies and T-cell responses to the spike protein. Casirivimab and imdevimab plus mRNA-1273 was generally well tolerated; a slight increase in treatment-emergent adverse events was observed in the casirivimab and imdevimab plus vaccine groups versus the vaccine-only group. INTERPRETATION Casirivimab and imdevimab administration before or at the time of COVID-19 vaccination reduced the elicitation of SARS-CoV-2 neutralising antibodies, but minimal effect was observed when vaccination occurred before mAb administration. Although the clinical significance of this decrease in neutralisation is unclear, this evidence suggests that further investigation of potential interactions could be warranted before concurrent clinical use of mAbs and vaccines targeting the same viral proteins as their main modes of action for the prevention or treatment of infectious diseases. FUNDING Regeneron Pharmaceuticals and F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Flonza Isa
- Regeneron Pharmaceuticals, Tarrytown, NY, USA.
| | | | | | | | | | | | | | | | - Lori Faria
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Mary Marovich
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Julia Hutter
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Laura Polakowski
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | | | | | - Alina Baum
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Faisal A Fakih
- Clinical Site Partners, d/b/a CSP Orlando, Winter Park, FL, USA
| | - M Juliana McElrath
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Stephen C De Rosa
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | | | | | | | - Ahmad J Odeh
- Duke University School of Medicine, Durham, NC, USA
| | | | | | | | | | - Bret Musser
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | |
Collapse
|
32
|
Jung M, Kim H, Choi E, Shin MK, Shin SJ. Enhancing vaccine effectiveness in the elderly to counter antibiotic resistance: The potential of adjuvants via pattern recognition receptors. Hum Vaccin Immunother 2024; 20:2317439. [PMID: 39693178 DOI: 10.1080/21645515.2024.2317439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 12/20/2024] Open
Abstract
Vaccines are an effective way to prevent the emergence and spread of antibiotic resistance by preventing diseases and establishing herd immunity. However, the reduced effectiveness of vaccines in the elderly due to immunosenescence is one of the significant contributors to the increasing antibiotic resistance. To counteract this decline and enhance vaccine effectiveness in the elderly, adjuvants play a pivotal role. Adjuvants are designed to augment the effectiveness of vaccines by activating the innate immune system, particularly through pattern recognition receptors on antigen-presenting cells. To improve vaccine effectiveness in the elderly using adjuvants, it is imperative to select the appropriate adjuvants based on an understanding of immunosenescence and the mechanisms of adjuvant functions. This review demonstrates the phenomenon of immunosenescence and explores various types of adjuvants, including their mechanisms and their potential in improving vaccine effectiveness for the elderly, thereby contributing to developing more effective vaccines for this vulnerable demographic.
Collapse
Affiliation(s)
- Myunghwan Jung
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunsol Choi
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
33
|
Hsieh MJ, Tsai PH, Chiang PH, Kao ZK, Zhuang ZQ, Hsieh AR, Ho HL, Chiou SH, Liang KH, Chen YC. Genomic insights into mRNA COVID-19 vaccines efficacy: Linking genetic polymorphisms to waning immunity. Hum Vaccin Immunother 2024; 20:2399382. [PMID: 39254005 PMCID: PMC11404610 DOI: 10.1080/21645515.2024.2399382] [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: 06/25/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Genetic polymorphisms have been linked to the differential waning of vaccine-induced immunity against COVID-19 following vaccination. Despite this, evidence on the mechanisms behind this waning and its implications for vaccination policy remains limited. We hypothesize that specific gene variants may modulate the development of vaccine-initiated immunity, leading to impaired immune function. This study investigates genetic determinants influencing the sustainability of immunity post-mRNA vaccination through a genome-wide association study (GWAS). Utilizing a hospital-based, test negative case-control design, we enrolled 1,119 participants from the Taiwan Precision Medicine Initiative (TPMI) cohort, all of whom completed a full mRNA COVID-19 vaccination regimen and underwent PCR testing during the Omicron outbreak. Participants were classified into breakthrough and protected groups based on PCR results. Genetic samples were analyzed using SNP arrays with rigorous quality control. Cox regression identified significant single nucleotide polymorphisms (SNPs) associated with breakthrough infections, affecting 743 genes involved in processes such as antigenic protein translation, B cell activation, and T cell function. Key genes identified include CD247, TRPV1, MYH9, CCL16, and RPTOR, which are vital for immune responses. Polygenic risk score (PRS) analysis revealed that individuals with higher PRS are at greater risk of breakthrough infections post-vaccination, demonstrating a high predictability (AUC = 0.787) in validating population. This finding confirms the significant influence of genetic variations on the durability of immune responses and vaccine effectiveness. This study highlights the importance of considering genetic polymorphisms in evaluating vaccine-induced immunity and proposes potential personalized vaccination strategies by tailoring regimens to individual genetic profiles.
Collapse
Affiliation(s)
- Min-Jia Hsieh
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pin-Hsuan Chiang
- Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Zih-Kai Kao
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Zi-Qing Zhuang
- Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ai-Ru Hsieh
- Department of Statistics, Tamkang University, New Taipei, Taiwan
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kung-Hao Liang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Biosafety level 3 laboratory, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chun Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan
- School of medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital Yuli Branch, Hualien, Taiwan
| |
Collapse
|
34
|
Cheng J, Jian L, Chen Z, Li Z, Yu Y, Wu Y. In Vivo Delivery Processes and Development Strategies of Lipid Nanoparticles. Chembiochem 2024; 25:e202400481. [PMID: 39101874 DOI: 10.1002/cbic.202400481] [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: 05/31/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/06/2024]
Abstract
Lipid nanoparticles (LNPs) represent an advanced and highly efficient delivery system for RNA molecules, demonstrating exceptional biocompatibility and remarkable delivery efficiency. This is evidenced by the clinical authorization of three LNP formulations: Patisiran, BNT162b2, and mRNA-1273. To further maximize the efficacy of RNA-based therapy, it is imperative to develop more potent LNP delivery systems that can effectively protect inherently unstable and negatively charged RNA molecules from degradation by nucleases, while facilitating their cellular uptake into target cells. Therefore, this review presents feasible strategies commonly employed for the development of efficient LNP delivery systems. The strategies encompass combinatorial chemistry for large-scale synthesis of ionizable lipids, rational design strategy of ionizable lipids, functional molecules-derived lipid molecules, the optimization of LNP formulations, and the adjustment of particle size and charge property of LNPs. Prior to introducing these developing strategies, in vivo delivery processes of LNPs, a crucial determinant influencing the clinical translation of LNP formulations, is described to better understand how to develop LNP delivery systems.
Collapse
Affiliation(s)
- Jiashun Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lina Jian
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhaolin Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhuoyuan Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaobang Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
35
|
Molina E, Tejero M, Duzenli OF, Kuoch H, Caine C, Krotova K, Paulaitis M, Aslanidi G. Insights in AAV-mediated antigen-specific immunity and a strategy for AAV vaccine dose reduction through AAV-extracellular vesicle association. Mol Ther Methods Clin Dev 2024; 32:101358. [PMID: 39559560 PMCID: PMC11570487 DOI: 10.1016/j.omtm.2024.101358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024]
Abstract
We previously showed therapeutic advantages of using a capsid-modified and encoded antigen-optimized AAV-based cancer vaccine to initiate strong antigen-specific immune responses and increase survival in a syngeneic mouse model of melanoma. In this study, we further explore AAV vaccine dose reduction and possible mechanisms of the immune response. Immunization with extracellular vesicle (EV)-associated AAV6-S663V encoded ovalbumin (OVA) or tyrosinase-related protein 1 (TRP-1) induced significantly higher levels of antigen-specific CD8+ T cells compared with standard AAV in mice. Importantly, a higher number of specific CD8+ T cells was achieved with EV-AAV several logs lower than optAAV-based doses. EV-optAAV-OVA was used in a dose 100 times lower, and EV-optTRP-1 10 times lower than optOVA and optTRP-1 correspondingly. Our data suggest that significant dose reduction for optimized AAV-based vaccines is possible without sacrificing efficiency. In addition, we studied the role of conventional type 1 dendritic cells (cDC1) in optimized AAV-based immunization using a C57BL/6-Irf8em1Kmm (Irf8 + 32-/-) mouse model lacking cDC1. Interestingly, we found that cDC1 are not essential for conveying effector T cell responses to AAV-encoded tumor antigens.
Collapse
Affiliation(s)
- Ester Molina
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Marcos Tejero
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | | | - Hisae Kuoch
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Colin Caine
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Karina Krotova
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Michael Paulaitis
- Department of Ophthalmology, Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - George Aslanidi
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 5455, USA
| |
Collapse
|
36
|
Hurst JR, Naghibosadat M, Budowski P, Liu J, Samaan P, Budiman F, Kurtesi A, Qi F, Menon H, Krishnan R, Abioye J, Gingras AC, Ostrowski M, Orozco NM, Kozak RA. Comparison of a SARS-CoV-2 mRNA booster immunization containing additional antigens to a spike-based mRNA vaccine against Omicron BA.5 infection in hACE2 mice. PLoS One 2024; 19:e0314061. [PMID: 39625929 PMCID: PMC11614295 DOI: 10.1371/journal.pone.0314061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
The emergence of SARS-CoV-2 variants presents challenges to vaccine effectiveness, underlining the necessity for next-generation vaccines with multiple antigens beyond the spike protein. Here, we investigated a multiantigenic booster containing spike and a chimeric construct composed of nucleoprotein (N) and membrane (M) proteins, comparing its efficacy to a spike-only booster against Omicron BA.5 in K18-hACE2 mice. Initially, mice were primed and boosted with Beta (B.1.351) spike-only mRNA, showing strong spike-specific T cell responses and neutralizing antibodies, albeit with limited cross-neutralization to Omicron variants. Subsequently, a spike-NM multiantigenic vaccine was then examined as a second booster dose for protection in hACE2-transgenic mice. Mice receiving either homologous spike-only or heterologous spike-NM booster had nearly complete inhibition of infectious virus shedding in oral swabs and reduced viral burdens in both lung and nasal tissues following BA.5 challenge. Examination of lung pathology further revealed that both spike-only and spike-NM boosters provided comparable protection against inflammatory infiltrates and fibrosis. Moreover, the spike-NM booster demonstrated neutralization efficacy in a pseudovirus assay against Wuhan-Hu-1, Beta, and Omicron variants akin to the spike-only booster. These findings indicate that supplementing spike with additional SARS-CoV-2 targets in a booster immunization confers equivalent immunity and protection against Omicron BA.5. This work highlights a promising strategy for individuals previously vaccinated with spike-only vaccines, potentially offering enhanced protection against emerging coronaviruses.
Collapse
Affiliation(s)
- Jacklyn R. Hurst
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Maedeh Naghibosadat
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Patrick Budowski
- Institute of Medical Sciences, University of Toronto, Ontario, Canada
| | - Jun Liu
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Frans Budiman
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alexandra Kurtesi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Fredo Qi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Haritha Menon
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | | | - Jumai Abioye
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mario Ostrowski
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Robert A. Kozak
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
- Division of Microbiology, Sunnybrook Health Sciences Centre, Department of Laboratory Medicine and Molecular Diagnostics, Toronto, ON, Canada
| |
Collapse
|
37
|
Wang B, Shen B, Xiang W, Shen H. Advances in the study of LNPs for mRNA delivery and clinical applications. Virus Genes 2024; 60:577-591. [PMID: 39172354 DOI: 10.1007/s11262-024-02102-6] [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: 04/22/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
Messenger ribonucleic acid (mRNA) was discovered in 1961 as an intermediary for transferring genetic information from DNA to ribosomes for protein synthesis. The COVID-19 pandemic brought worldwide attention to mRNA vaccines. The emergency use authorization of two COVID-19 mRNA vaccines, BNT162b2 and mRNA-1273, were major achievements in the history of vaccine development. Lipid nanoparticles (LNPs), one of the most superior non-viral delivery vectors available, have made many exciting advances in clinical translation as part of the COVID-19 vaccine and therefore has the potential to accelerate the clinical translation of many gene drugs. In addition, due to these small size, biocompatibility and excellent biodegradability, LNPs can efficiently deliver nucleic acids into cells, which is particularly important for current mRNA therapeutic regimens. LNPs are composed cationic or pH-dependent ionizable lipid bilayer, polyethylene glycol (PEG), phospholipids, and cholesterol, represents an advanced system for the delivery of mRNA vaccines. Furthermore, optimization of these four components constituting the LNPs have demonstrated enhanced vaccine efficacy and diminished adverse effects. The incorporation of biodegradable lipids enhance the biocompatibility of LNPs, thereby improving its potential as an efficacious therapeutic approach for a wide range of challenging and intricate diseases, encompassing infectious diseases, liver disorders, cancer, cardiovascular diseases, cerebrovascular conditions, among others. Consequently, this review aims to furnish the scientific community with the most up-to-date information regarding mRNA vaccines and LNP delivery systems.
Collapse
Affiliation(s)
- Bili Wang
- National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Biao Shen
- Hangzhou Cybernax Biotechnology Co. Ltd, Hangzhou, 311202, China
| | - Wenqing Xiang
- National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Hongqiang Shen
- National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
38
|
Li K, Wu Y, Zhang H, Chen S, Wu B, Li T, Li E, Luo F, Jin A, Zhang B, Zhang Y, Gong R, Zhang H, Chiu S. A Novel Circular Delta-XBB15 RBD Dimeric Protein Subunit Vaccine Mediated by Split Intein Elicits an Immune Response and Protection Against Multiple SARS-CoV-2 Variants in Mice. J Med Virol 2024; 96:e70134. [PMID: 39711477 DOI: 10.1002/jmv.70134] [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: 08/05/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/24/2024]
Abstract
SARS-CoV-2 continues to mutate, leading to breakthrough infections. The development of new vaccine strategies to combat various strains is crucial. Protein cyclization can enhance thermal stability and may improve immunogenicity. Here, we designed a cyclic tandem dimeric receptor-binding domain protein (cirRBD2) via the split intein Cth-Ter. Cyclization does not affect the antigen epitopes of RBD but results in better thermal stability than that of its linear counterpart (linRBD2). Compared with the mice immunized with linRBD2, those immunized with two doses of 5 μg of cirRBD2 produced significantly greater levels of broad-spectrum neutralizing antibodies, and generated a considerable cellular immune response. In the VEEV-VRP-hACE2-transduced mouse model, two doses of 5 μg of cirRBD2 provided protection against infection with BA.5, XBB.1.9, and partial protection against EG.5 which has more mutations. This study developed a novel circular RBD dimer subunit vaccine for SARS-CoV-2 that exhibits broad-spectrum neutralizing activity against various variants. A similar strategy can be applied to develop vaccines for other pathogens, especially for thermally stable vaccines.
Collapse
MESH Headings
- Animals
- Mice
- COVID-19 Vaccines/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Inteins
- Mice, Inbred BALB C
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/chemistry
- Humans
- Female
- Immunity, Cellular
Collapse
Affiliation(s)
- Kangyin Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Yan Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Hongqing Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shaohong Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Bihao Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Tingting Li
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Feiyang Luo
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, China
| | - Aishun Jin
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, China
| | - Bo Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yanan Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Rui Gong
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Huajun Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| |
Collapse
|
39
|
Bellavite P, Donzelli A, Isidoro C. The WHO Algorithm for Causality Assessment of Adverse Effects Following Immunization with Genetic-Based Anti-COVID-19 Vaccines: Pitfalls and Suggestions for Improvement. J Clin Med 2024; 13:7291. [PMID: 39685749 DOI: 10.3390/jcm13237291] [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: 09/24/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Clarifying and differentiating the causes of diseases is an essential step in any clinical activity, but it takes on particular relevance and complexity in the case that arise following vaccinations. The WHO has proposed a protocol that uses a list of specific questions about vaccine-related adverse events and an algorithm for making a judgement. Here, we analyze and discuss the important limitations of this protocol when applied to the new genetic-based anti-COVID-19 vaccines, particularly once dealing with rare and unexpected pathological events. The main controversial aspects concern: (a) the prevailing consideration of other possible causes; (b) the biological plausibility and the choice of an appropriate time window to consider adverse effects possibly caused by vaccines; (c) the reference to scientific literature, which may be very limited and often controversial in early stages of introducing new vaccines because of the short period of observation; (d) the final classification of the algorithm into only three classes, which leaves ample space for the "indeterminate" category. Failure to address these issues may lead to distorted pharmacovigilance reports with significant consequences on the benefit/harm assessment. In anticipation of possible future pandemics managed with new vaccines, the WHO algorithm needs to be revised with appropriate protocols for monitoring and evaluation of adverse effects that take into account the novel mechanism of action and real-world epidemiological data.
Collapse
Affiliation(s)
| | - Alberto Donzelli
- Foundation Allineare Sanità e Salute, via Ricordi 4, 20131 Milano, Italy
| | - Ciro Isidoro
- Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy
| |
Collapse
|
40
|
Chen L, Shao C, Li J, Zhu F. Impact of Immunosenescence on Vaccine Immune Responses and Countermeasures. Vaccines (Basel) 2024; 12:1289. [PMID: 39591191 PMCID: PMC11598585 DOI: 10.3390/vaccines12111289] [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: 10/13/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The biological progression of aging encompasses complex physiological processes. As individuals grow older, their physiological functions gradually decline, including compromised immune responses, leading to immunosenescence. Immunosenescence significantly elevates disease susceptibility and severity in older populations while concurrently compromising vaccine-induced immune responses. This comprehensive review aims to elucidate the implications of immunosenescence for vaccine-induced immunity and facilitate the development of optimized vaccination strategies for geriatric populations, with specific focus on COVID-19, influenza, pneumococcal, herpes zoster, and respiratory syncytial virus (RSV) vaccines. This review further elucidates the relationship between immunosenescence and vaccine-induced immunity. This review presents a systematic evaluation of intervention strategies designed to enhance vaccine responses in older populations, encompassing adjuvant utilization, antigen doses, vaccination frequency modification, inflammatory response modulation, and lifestyle interventions, including physical activity and nutritional modifications. These strategies are explored for their potential to improve current vaccine efficacy and inform the development of next-generation vaccines for geriatric populations.
Collapse
Affiliation(s)
- Li Chen
- School of Public Health, Southeast University, Nanjing 210096, China; (L.C.); (C.S.)
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Chengwei Shao
- School of Public Health, Southeast University, Nanjing 210096, China; (L.C.); (C.S.)
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Jingxin Li
- School of Public Health, Southeast University, Nanjing 210096, China; (L.C.); (C.S.)
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Fengcai Zhu
- School of Public Health, Southeast University, Nanjing 210096, China; (L.C.); (C.S.)
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| |
Collapse
|
41
|
Shi Y, Shi M, Wang Y, You J. Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct Target Ther 2024; 9:322. [PMID: 39543114 PMCID: PMC11564800 DOI: 10.1038/s41392-024-02002-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: 06/02/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
In the last decade, messenger ribonucleic acid (mRNA)-based drugs have gained great interest in both immunotherapy and non-immunogenic applications. This surge in interest can be largely attributed to the demonstration of distinct advantages offered by various mRNA molecules, alongside the rapid advancements in nucleic acid delivery systems. It is noteworthy that the immunogenicity of mRNA drugs presents a double-edged sword. In the context of immunotherapy, extra supplementation of adjuvant is generally required for induction of robust immune responses. Conversely, in non-immunotherapeutic scenarios, immune activation is unwanted considering the host tolerability and high expression demand for mRNA-encoded functional proteins. Herein, mainly focused on the linear non-replicating mRNA, we overview the preclinical and clinical progress and prospects of mRNA medicines encompassing vaccines and other therapeutics. We also highlight the importance of focusing on the host-specific variations, including age, gender, pathological condition, and concurrent medication of individual patient, for maximized efficacy and safety upon mRNA administration. Furthermore, we deliberate on the potential challenges that mRNA drugs may encounter in the realm of disease treatment, the current endeavors of improvement, as well as the application prospects for future advancements. Overall, this review aims to present a comprehensive understanding of mRNA-based therapies while illuminating the prospective development and clinical application of mRNA drugs.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Meixing Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, P. R. China.
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, P. R. China.
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, P. R. China.
| |
Collapse
|
42
|
Haghighi E, Abolmaali SS, Dehshahri A, Mousavi Shaegh SA, Azarpira N, Tamaddon AM. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. J Nanobiotechnology 2024; 22:710. [PMID: 39543630 PMCID: PMC11566655 DOI: 10.1186/s12951-024-02972-w] [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: 08/29/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
RNA therapeutics, such as mRNA, siRNA, and CRISPR-Cas9, present exciting avenues for treating diverse diseases. However, their potential is commonly hindered by vulnerability to degradation and poor cellular uptake, requiring effective delivery systems. Lipid nanoparticles (LNPs) have emerged as a leading choice for in vivo RNA delivery, offering protection against degradation, enhanced cellular uptake, and facilitation of endosomal escape. However, LNPs encounter numerous challenges for targeted RNA delivery in vivo, demanding advanced particle engineering, surface functionalization with targeting ligands, and a profound comprehension of the biological milieu in which they function. This review explores the structural and physicochemical characteristics of LNPs, in-vivo fate, and customization for RNA therapeutics. We highlight the quality-by-design (QbD) approach for targeted delivery beyond the liver, focusing on biodistribution, immunogenicity, and toxicity. In addition, we explored the current challenges and strategies associated with LNPs for in-vivo RNA delivery, such as ensuring repeated-dose efficacy, safety, and tissue-specific gene delivery. Furthermore, we provide insights into the current clinical applications in various classes of diseases and finally prospects of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
- Elahe Haghighi
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Azarpira
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
43
|
Omidi Y, Pourseif MM, Ansari RA, Barar J. Design and development of mRNA and self-amplifying mRNA vaccine nanoformulations. Nanomedicine (Lond) 2024; 19:2699-2725. [PMID: 39535127 DOI: 10.1080/17435889.2024.2419815] [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: 04/15/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
The rapid evolution of mRNA vaccines, highlighted by Pfizer-BioNTech and Moderna's COVID-19 vaccines, has transformed vaccine development and therapeutic approaches. Self-amplifying mRNA (saRNA) vaccines, a groundbreaking advancement in RNA-based vaccines, offer promising possibilities for disease prevention and treatment, including potential applications in cancer and neurodegenerative diseases. This review explores the complex design and development of these innovative vaccines, with a focus on their nanoscale formulations that utilize nanotechnology to improve their delivery and effectiveness. It articulates the fundamental principles of mRNA and saRNA vaccines, their mechanisms of action, and the role of synthetic mRNA in eliciting immune responses. The review further elaborates on various nanoscale delivery systems (e.g., lipid nanoparticles, polymeric nanoparticles and other nanocarriers), emphasizing their advantages in enhancing mRNA stability and cellular uptake. It addresses advanced nanoscale delivery techniques such as microfluidics and discusses the challenges in formulating mRNA and saRNA vaccines. By incorporating the latest technologies and current research, this review provides a thorough overview of recent mRNA and saRNA nanovaccines advancements, highlighting their potential to revolutionize vaccine technology and broaden clinical applications.
Collapse
Affiliation(s)
- Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Mohammad M Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Engineered Biomaterial Research Center, Khazar University, Baku, Azerbaijan
| | - Rais A Ansari
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
44
|
Zickler AM, Liang X, Gupta D, Mamand DR, De Luca M, Corso G, Errichelli L, Hean J, Sen T, Elsharkasy OM, Kamei N, Niu Z, Zhou G, Zhou H, Roudi S, Wiklander OPB, Görgens A, Nordin JZ, Castilla‐Llorente V, EL Andaloussi S. Novel Endogenous Engineering Platform for Robust Loading and Delivery of Functional mRNA by Extracellular Vesicles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407619. [PMID: 39246205 PMCID: PMC11558116 DOI: 10.1002/advs.202407619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Messenger RNA (mRNA) has emerged as an attractive therapeutic molecule for a plethora of clinical applications. For in vivo functionality, mRNA therapeutics require encapsulation into effective, stable, and safe delivery systems to protect the cargo from degradation and reduce immunogenicity. Here, a bioengineering platform for efficient mRNA loading and functional delivery using bionormal nanoparticles, extracellular vesicles (EVs), is established by expressing a highly specific RNA-binding domain fused to CD63 in EV producer cells stably expressing the target mRNA. The additional combination with a fusogenic endosomal escape moiety, Vesicular Stomatitis Virus Glycoprotein, enables functional mRNA delivery in vivo at doses substantially lower than currently used clinically with synthetic lipid-based nanoparticles. Importantly, the application of EVs loaded with effective cancer immunotherapy proves highly effective in an aggressive melanoma mouse model. This technology addresses substantial drawbacks currently associated with EV-based nucleic acid delivery systems and is a leap forward to clinical EV applications.
Collapse
Affiliation(s)
- Antje M. Zickler
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Xiuming Liang
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Cancer Research LaboratoryShandong University‐Karolinska Institutet collaborative LaboratorySchool of Basic Medical ScienceShandong UniversityNo. 44, Wenhua Xi RoadJi'nanShandong250012P. R. China
| | - Dhanu Gupta
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics.University of OxfordOld Road Campus, Roosevelt Dr, HeadingtonOxfordOX3 7TYUK
| | - Doste R. Mamand
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Breast Center, Karolinska Comprehensive Cancer CenterKarolinska University HospitalStockholm14186Sweden
| | - Mariacristina De Luca
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
- Human TechnopoleViale Rita Levi Montalcini, 1Milan20157Italy
| | - Giulia Corso
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Evercyte GmbHLeberstrasse 20Vienna1110Austria
| | - Lorenzo Errichelli
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
| | - Justin Hean
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
| | - Titash Sen
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
- Lonza BiologicsChesterford Research ParkCambridgeCB10 1XLUK
| | - Omnia M. Elsharkasy
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Noriyasu Kamei
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Laboratory of Drug Delivery SystemsFaculty of Pharmaceutical SciencesKobe Gakuin University1‐1‐3 Minatojima, Chuo‐kuKobeHyogo650‐8586Japan
| | - Zheyu Niu
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Department of Hepatobiliary SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityNo. 324, Five Jing RoadJi'nanShandong250012P. R. China
| | - Guannan Zhou
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Department of GynecologyThe Obstetrics and Gynecology Hospital of Fudan UniversityNo. 419, Fangxie RoadShanghai200011P. R. China
| | - Houze Zhou
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Samantha Roudi
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| | - Oscar P. B. Wiklander
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Breast Center, Karolinska Comprehensive Cancer CenterKarolinska University HospitalStockholm14186Sweden
| | - André Görgens
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐Essen45147EssenGermany
| | - Joel Z. Nordin
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Clinical Immunology and Transfusion Medicine (KITM)Karolinska University HospitalStockholm14186Sweden
| | - Virginia Castilla‐Llorente
- Evox Therapeutics Ltd.Oxford Science ParkMedawar CentreRobert Robinson AvenueOxfordOX4 4HGUK
- Uncommon BioCambridge TechnoparkNewmarket RdCambridgeCB5 8PBUK
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm14186Sweden
- Karolinska ATMP Center, Karolinska InstitutetANA FuturaAlfred‐Nobels‐Allé 8, HuddingeStockholm14152Sweden
| |
Collapse
|
45
|
Carvalho HMB, Fidalgo TAS, Acúrcio RC, Matos AI, Satchi-Fainaro R, Florindo HF. Better, Faster, Stronger: Accelerating mRNA-Based Immunotherapies With Nanocarriers. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2017. [PMID: 39537215 DOI: 10.1002/wnan.2017] [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: 06/15/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Messenger ribonucleic acid (mRNA) therapeutics are attracting attention as promising tools in cancer immunotherapy due to their ability to leverage the in vivo expression of all known protein sequences. Even small amounts of mRNA can have a powerful effect on cancer vaccines by promoting the synthesis of tumor-specific antigens (TSA) or tumor-associated antigens (TAA) by antigen-presenting cells (APC). These antigens are then presented to T cells, eliciting strong antitumor immune stimulation. The potential of mRNA can be further enhanced by expressing immunomodulatory agents, such as cytokines, antibodies, and chimeric antigen receptors (CAR), enhancing tumor immunity. Recent research also explores mRNA-encoded tumor death inducers or tumor microenvironment (TME) modulators. Despite its promise, the clinical translation of mRNA-based anticancer strategies faces challenges, including inefficient targeted delivery in vivo, failure of endosomal escape, and inadequate intracellular mRNA release, resulting in poor transfection efficiencies. Inspired by the approval of lipid nanoparticle-loaded mRNA vaccines against coronavirus disease 2019 (COVID-19) and the encouraging outcomes of mRNA-based cancer therapies in trials, innovative nonviral nanotechnology delivery systems have been engineered. These aim to advance mRNA-based cancer immunotherapies from research to clinical application. This review summarizes recent preclinical and clinical progress in lipid and polymeric nanomedicines for delivering mRNA-encoded antitumor therapeutics, including cytokines and antibody-based immunotherapies, cancer vaccines, and CAR therapies. It also addresses advanced delivery systems for direct oncolysis or TME reprogramming and highlights key challenges in translating these therapies to clinical use, exploring future perspectives, including the role of artificial intelligence and machine learning in their development.
Collapse
Affiliation(s)
- Henrique M B Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago A S Fidalgo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ana I Matos
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
46
|
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
|
47
|
Eslami SM, Lu X. Recent advances in mRNA-based cancer vaccines encoding immunostimulants and their delivery strategies. J Control Release 2024; 376:S0168-3659(24)00708-9. [PMID: 39437963 DOI: 10.1016/j.jconrel.2024.10.035] [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: 05/30/2024] [Revised: 09/01/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
The high prevalence of drug resistance, relapse, and unfavorable response rate of conventional cancer therapies necessitate the development of more efficient treatment modalities. Immunotherapy represents a novel therapeutic approach to cancer treatment in which the immune system's potential is harnessed to recognize and eliminate tumor cells. mRNA cancer vaccines, as a burgeoning field of immunotherapy, have recently drawn particular attention, and among mRNAs encoding tumor-associated antigens, tumor-specific antigens, and immune stimulatory factors, the latter has been relatively less explored. These immunostimulatory mRNAs encode a range of proteins, including stimulatory ligands, receptors, enzymes, pro-inflammatory cytokines, and inhibitory binding proteins, which collectively augment the host immune system's ability against cancerous cells. In this review, we aimed to provide a comprehensive account of mRNA-based cancer vaccines encoding immune stimulants, encompassing their current status, mechanisms of action, delivery strategies employed, as well as recent advances in preclinical and clinical studies. The potential challenges, strategies and future perspectives have also been discussed.
Collapse
Affiliation(s)
- Seyyed Majid Eslami
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA.
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
48
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
49
|
Acúrcio RC, Kleiner R, Vaskovich‐Koubi D, Carreira B, Liubomirski Y, Palma C, Yeheskel A, Yeini E, Viana AS, Ferreira V, Araújo C, Mor M, Freund NT, Bacharach E, Gonçalves J, Toister‐Achituv M, Fabregue M, Matthieu S, Guerry C, Zarubica A, Aviel‐Ronen S, Florindo HF, Satchi‐Fainaro R. Intranasal Multiepitope PD-L1-siRNA-Based Nanovaccine: The Next-Gen COVID-19 Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404159. [PMID: 39116324 PMCID: PMC11515909 DOI: 10.1002/advs.202404159] [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: 04/21/2024] [Revised: 07/28/2024] [Indexed: 08/10/2024]
Abstract
The first approved vaccines for human use against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are nanotechnology-based. Although they are modular, rapidly produced, and can reduce disease severity, the currently available vaccines are restricted in preventing infection, stressing the global demand for novel preventive vaccine technologies. Bearing this in mind, we set out to develop a flexible nanovaccine platform for nasal administration to induce mucosal immunity, which is fundamental for optimal protection against respiratory virus infection. The next-generation multiepitope nanovaccines co-deliver immunogenic peptides, selected by an immunoinformatic workflow, along with adjuvants and regulators of the PD-L1 expression. As a case study, we focused on SARS-CoV-2 peptides as relevant antigens to validate the approach. This platform can evoke both local and systemic cellular- and humoral-specific responses against SARS-CoV-2. This led to the secretion of immunoglobulin A (IgA), capable of neutralizing SARS-CoV-2, including variants of concern, following a heterologous immunization strategy. Considering the limitations of the required cold chain distribution for current nanotechnology-based vaccines, it is shown that the lyophilized nanovaccine is stable for long-term at room temperature and retains its in vivo efficacy upon reconstitution. This makes it particularly relevant for developing countries and offers a modular system adaptable to future viral threats.
Collapse
Affiliation(s)
- Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Ron Kleiner
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Daniella Vaskovich‐Koubi
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Bárbara Carreira
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Yulia Liubomirski
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Carolina Palma
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Adva Yeheskel
- The Blavatnik Center for Drug DiscoveryTel Aviv UniversityTel Aviv6997801Israel
| | - Eilam Yeini
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Ana S. Viana
- Center of Chemistry and BiochemistryFaculty of SciencesUniversity of LisbonLisbon1749‐016Portugal
| | - Vera Ferreira
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Carlos Araújo
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Michael Mor
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Natalia T. Freund
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Eran Bacharach
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - João Gonçalves
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | | | - Manon Fabregue
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Solene Matthieu
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Capucine Guerry
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Ana Zarubica
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | | | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Ronit Satchi‐Fainaro
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv6997801Israel
| |
Collapse
|
50
|
Asare AF, Sabblah GT, Buabeng RO, Alhassan Y, Asamoa-Amoakohene A, Amponsa-Achiano K, Mohammed NT, Darko DM, Bonful HA. Adverse events following COVID-19 vaccination: A comprehensive analysis of spontaneous reporting data in Ghana. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003770. [PMID: 39331603 PMCID: PMC11432875 DOI: 10.1371/journal.pgph.0003770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/04/2024] [Indexed: 09/29/2024]
Abstract
Vaccines are important public health tools and formed part of the fight against the COVID-19 pandemic. Five COVID-19 vaccines were given Emergency Use Authorization in Ghana and deployed during the pandemic. Early phase trials of the vaccines were mostly not conducted in Africans. This study examines safety data during their deployment under real-life conditions in Ghana. This study analysed secondary data on COVID-19 vaccine-related adverse events following immunization (AEFI) reported to the Ghana Food and Drugs Authority (GFDA) between March 2021 and June 2022 using STATA. AEFIs were coded with their Preferred Terms using the Medical Dictionary for Regulatory Activities, version 24.0. Statistical tests examined associations between demographic characteristics, vaccine types, seriousness, and AEFI outcomes. Binary logistic regression model assessed factors associated with serious AEFIs, while the GFDA's Joint COVID-19 Vaccine Safety Review Committee provided causality assessments of serious AEFIs. Overall cumulative incidence of AEFIs was about 25 per 100,000 persons vaccinated. Across the five vaccines, majority of the AEFIs reported were not serious (98.7%) with higher incidences in those below 50 years (74.0%) and females (51.2%). The most common AEFIs recorded were headache (52.9%), pains (44.4%), pyrexia (35.1%), chills (16.7%) and injection site pain (15.6%). Relative to those 50 years and above, the odds of serious AEFI were 60% less among those aged <30 years (aOR = 0.40, CI: [0.19, 0.86], p = 0.019). However, a causality assessment of the 57 serious AEFIs indicated only 8 (14%) were vaccine product-related. There was a low incidence of AEFIs following deployment of the vaccines in Ghana with a much lower incidence of serious AEFIs. Informing the public about the safety of the vaccines and potential side effects may increase trust and acceptance, decreasing hesitancy in current and future vaccination programmes.
Collapse
Affiliation(s)
| | | | | | - Yakubu Alhassan
- Department of Biostatistics, School of Public Health, University of Ghana, Legon, Accra, Ghana
| | | | | | | | | | - Harriet Affran Bonful
- Department of Epidemiology and Disease Control, School of Public Health, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|