51
|
Collins CP, Herzog C, Vick LV, Nielsen R, Harville YI, Longo DL, Arthur JM, Murphy WJ. Sequential SARS-CoV-2 mRNA Vaccination Induces Anti-Idiotype (Anti-ACE2) Antibodies in K18 Human ACE2 Transgenic Mice. Vaccines (Basel) 2025; 13:224. [PMID: 40266063 PMCID: PMC11946769 DOI: 10.3390/vaccines13030224] [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: 12/21/2024] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Novel mRNA vaccines have been successfully utilized to curtail the SARS-CoV-2 pandemic. However, the immunology underlying CoV2 vaccinations, particularly with repeated boosting, has not been properly characterized due to limitations in the preclinical modeling of SARS-CoV-2 infection/vaccinations as well as constantly changing vaccine formulations. The immunoregulatory aspects involved in such vaccine approaches remain unclear. Antibodies, due to inherent immunogenicity by VDJ gene rearrangement, have the potential to induce antibodies directed towards them called anti-idiotype antibodies, which can play a downregulatory role in responses. The paratope of some of these anti-idiotype antibodies can also act as a mirror to the original antigen, which, in the case of SARS-CoV-2 vaccines, would be to the spike protein and, therefore, also be capable of binding its target, ACE2, potentially causing adverse effects. METHODS To investigate if sequential SARS-CoV-2 mRNA vaccination can induce anti-idiotype antibody responses, K18 hACE2 transgenic mice were serially vaccinated with a SARS-CoV-2 mRNA construct to determine the kinetics of anti-spike and anti-ACE2 responses via custom-made ELISAs. RESULTS While sequential vaccination produced robust anti-spike responses, anti-ACE2 levels were also detected and gradually amplified with each boost. These anti-ACE2 antibodies persisted for 3 months after the final vaccination and showed evidence of hACE2 binding, as levels were lower in K18 mice in comparison to the wild type. CONCLUSIONS These data would suggest that sequential SARS-CoV-2 mRNA vaccination has the potential to induce anti-ACE2 antibodies in mice, with each boost amplifying the amount of antibody.
Collapse
Affiliation(s)
- Craig P. Collins
- School of Medicine, University of California, Davis, CA 95817, USA; (C.P.C.); (L.V.V.); (R.N.)
| | - Christian Herzog
- Department of Internal Medicine Nephrology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (C.H.); (Y.I.H.); (J.M.A.)
| | - Logan V. Vick
- School of Medicine, University of California, Davis, CA 95817, USA; (C.P.C.); (L.V.V.); (R.N.)
| | - Ryan Nielsen
- School of Medicine, University of California, Davis, CA 95817, USA; (C.P.C.); (L.V.V.); (R.N.)
| | - Yanping Izak Harville
- Department of Internal Medicine Nephrology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (C.H.); (Y.I.H.); (J.M.A.)
| | - Dan L. Longo
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA;
| | - John M. Arthur
- Department of Internal Medicine Nephrology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (C.H.); (Y.I.H.); (J.M.A.)
| | - William J. Murphy
- School of Medicine, University of California, Davis, CA 95817, USA; (C.P.C.); (L.V.V.); (R.N.)
- Department of Internal Medicine, Division of Hematology and Oncology, School of Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
52
|
Yanik S, Venkatesh V, Gordy JT, Alameh MG, Meza J, Li Y, Glass E, Flores-Garcia Y, Tam Y, Chaiyawong N, Sarkar D, Weissman D, Markham R, Srinivasan P. iDC-targeting PfCSP mRNA vaccine confers superior protection against Plasmodium compared to conventional mRNA. NPJ Vaccines 2025; 10:34. [PMID: 39971939 PMCID: PMC11840135 DOI: 10.1038/s41541-025-01089-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: 06/28/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
Malaria resurgence in 2022 saw 249 million clinical cases and 608,000 deaths, mostly in children under five. The WHO-approved circumsporozoite protein (CSP)-targeting vaccines, RTS,S and R21, remain limited in availability. Strong humoral responses are crucial for sporozoite neutralization before hepatocyte infection, yet first-generation vaccines provide suboptimal protection, necessitating improved strategies. With the success of mRNA-LNP vaccines against COVID-19, there is interest in leveraging this approach to malaria. Here, we developed a novel chemokine fusion mRNA vaccine targeting immature dendritic cells (iDC) to enhance immunity against P. falciparum CSP (PfCSP). Mice immunized with MIP3α-CSP mRNA-LNP exhibited stronger CD4 + T cell responses and higher anti-NANP6 antibody titers than conventional CSP mRNA-LNP. Importantly, upon P. berghei PfCSP transgenic sporozoite challenge, MIP3α-CSP mRNA provided significantly greater protection from liver infection, strongly associated with multifunctional CD4 + T cells and anti-NANP6 titers. This study underscores iDC targeting as a promising strategy to enhance malaria vaccine efficacy.
Collapse
Affiliation(s)
- Sean Yanik
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Varsha Venkatesh
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - James T Gordy
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | | | - Jacob Meza
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Yangchen Li
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Elizabeth Glass
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Nattawat Chaiyawong
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Deepti Sarkar
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Drew Weissman
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Markham
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA.
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA.
| |
Collapse
|
53
|
Chen S, Wang S, Liao S, Blakney AK. Exploring the Effects of Incorporating Different Bioactive Phospholipids into Messenger Ribonucleic Acid Lipid Nanoparticle (mRNA LNP) Formulations. ACS BIO & MED CHEM AU 2025; 5:154-165. [PMID: 39990935 PMCID: PMC11843334 DOI: 10.1021/acsbiomedchemau.4c00085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 02/25/2025]
Abstract
The current rapid advancement in ribonucleic acid (RNA) therapeutics research depends on innovations in drug delivery, especially the development of a lipid-nanoparticle (LNP)-based system. The conventional LNP formulation typically contains four components, including an ionizable cationic lipid, a phospholipid, cholesterol or a cholesterol derivative, and poly(ethylene glycol) (PEG)-lipid, with each contributing to the formulation's overall stability and effectiveness. Among these four types of lipids, the phospholipid component is often known to provide structural support for the nanoparticles but is also a class of bioactive molecules with strong cell signaling potential. This study explores the possibility of incorporating some known structurally related bioactive phospholipids as the fifth component of a conventional four-component LNP formulation and assesses the impacts of such an approach on the physicochemical properties and biological functions of the mRNA LNP formulation. We screened a library of mRNA LNP formulations containing 7 different structurally related bioactive phospholipids at molar concentrations of 5%, 15% and 30% in addition to a conventional four-component LNP formulation (base). We observed differences in physicochemical properties between the mRNA LNP formulations that could be attributed to both the types of phospholipids examined and the molar concentrations used. Cryo-EM analysis revealed structural similarity between the Base formulation and the other formulations. We also characterized the protein expression level in HeLa cells and picked up a distinct cytokine panel signature for each formulation in human peripheral blood mononuclear cells (hPBMCs). Further immunophenotyping analysis showed that most cells that were transfected were CD4+ T cells, and the addition of the different bioactive phospholipids slightly altered cellular tropism. This exploratory study illustrates how adding the bioactive phospholipid can be used to modulate the LNP function, further expanding the design space for RNA LNP formulations and potentiating LNPs for use as RNA therapeutics.
Collapse
Affiliation(s)
- Sunny
P. Chen
- School
of Biomedical Engineering, University of
British Columbia, Vancouver V6T 1Z3, Canada
- Michael
Smith Laboratories, University of British
Columbia, Vancouver V6T 1Z4, Canada
| | - Shuangyu Wang
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, Vancouver V6T 2A1, Canada
| | - Suiyang Liao
- School
of Biomedical Engineering, University of
British Columbia, Vancouver V6T 1Z3, Canada
- Michael
Smith Laboratories, University of British
Columbia, Vancouver V6T 1Z4, Canada
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, Vancouver V6T 2A1, Canada
| | - Anna K. Blakney
- School
of Biomedical Engineering, University of
British Columbia, Vancouver V6T 1Z3, Canada
- Michael
Smith Laboratories, University of British
Columbia, Vancouver V6T 1Z4, Canada
| |
Collapse
|
54
|
Ben-Akiva E, Chapman A, Mao T, Irvine DJ. Linking vaccine adjuvant mechanisms of action to function. Sci Immunol 2025; 10:eado5937. [PMID: 39951545 DOI: 10.1126/sciimmunol.ado5937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/23/2025] [Indexed: 02/16/2025]
Abstract
Vaccines deliver an immunogen in a manner designed to safely provoke an immune response, leading to the generation of memory T and B cells and long-lived antibody-producing plasma cells. Adjuvants play a critical role in vaccines by controlling how the immune system is exposed to the immunogen and providing inflammatory cues that enable productive immune priming. However, mechanisms of action underlying adjuvant function at the molecular, cell, and tissue levels are diverse and often poorly understood. Here, we review the current understanding of mechanisms of action underlying adjuvants used in subunit protein/polysaccharide vaccines and mRNA vaccines, discuss where possible how these mechanisms of action link to downstream effects on the immune response, and identify knowledge gaps that will be important to fill in order to enable the continued development of more effective adjuvants for challenging pathogens such as HIV and emerging threats.
Collapse
Affiliation(s)
- Elana Ben-Akiva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Asheley Chapman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Tianyang Mao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
- Broad Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
55
|
Boehm DT, Landreth KM, Kilic ES, Lee KS, Misra B, Bobbala S, Damron FH, Liu TW. Intratumoral administration of mRNA COVID-19 vaccine delays melanoma growth in mice. Sci Rep 2025; 15:5337. [PMID: 39948424 PMCID: PMC11825918 DOI: 10.1038/s41598-025-89930-0] [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: 08/29/2024] [Accepted: 02/10/2025] [Indexed: 02/16/2025] Open
Abstract
Immunotherapies are effective for cancer treatment but are limited in 'cold' tumor microenvironments due to a lack of infiltrating CD8+ T cells, key players in the anti-cancer immune response. The onset of the COVID-19 pandemic sparked the widespread use of mRNA-formulated vaccines and is well documented that vaccination induces a Th1-skewed immune response. Here, we evaluated the effects of an intratumoral injection of the mRNA COVID-19 vaccine in subcutaneous melanoma tumor mouse models. Tumor growth and survival studies following a single intratumoral injection of the COVID-19 vaccine showed significant tumor suppression and prolonged survival in established B16F10 subcutaneous tumor-bearing mice. mRNA vaccine treatment resulted in a significant increase in CD8+ T cell infiltration into the tumor microenvironment, as observed using intravital imaging and flow cytometry. Further tumor growth suppression was achieved using additional mRNA vaccine treatments. Combination administration of mRNA vaccine with immune checkpoint therapies demonstrated enhanced effects, further delaying tumor growth and improving the survival time of tumor-bearing mice. This study demonstrates that mRNA vaccines may be used as adjuvants for immunotherapies.
Collapse
Affiliation(s)
- Dylan T Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Kaitlyn M Landreth
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Emel Sen Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Katherine S Lee
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Tracy W Liu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
56
|
Pekarek MJ, Madapong A, Wiggins J, Weaver EA. Adenoviral-Vectored Multivalent Vaccine Provides Durable Protection Against Influenza B Viruses from Victoria-like and Yamagata-like Lineages. Int J Mol Sci 2025; 26:1538. [PMID: 40004004 PMCID: PMC11855595 DOI: 10.3390/ijms26041538] [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: 01/17/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Despite annual vaccines, Influenza B viruses (IBVs) continue to cause severe infections and have a significant impact and burden on the healthcare system. Improving IBV vaccine effectiveness is a key focus, with various strategies under investigation. In this research, we used a computational design to select wildtype sequences for a multivalent viral-vectored vaccine (rAd-Tri-Vic) targeting the Victoria-like (Vic) hemagglutinin (HA) protein. In mouse models, the vaccine induced strong antibody and T cell responses, providing complete protection against both lineage-specific and cross-lineage (Yamagata-like) lethal challenges. The immune responses remained robust for up to six months, which demonstrated sustained protection. These results highlight the potential of HA-targeted multivalent vaccines to enhance the IBV efficacy and address protection against antigenically diverse IBV strains.
Collapse
Affiliation(s)
- Matthew J. Pekarek
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| | - Adthakorn Madapong
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
| | - Joshua Wiggins
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| | - Eric A. Weaver
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| |
Collapse
|
57
|
Zhong Z, Deventer MH, Chen Y, Vanhee S, Lammens I, Deswarte K, Huang Y, Ye T, Wang H, Nuhn L, Vandeputte MM, Gontsarik M, Cui X, Sanders NN, Lienenklaus S, N Lambrecht B, Baptista AP, Stove CP, G De Geest B. A Fentanyl Hapten-Displaying Lipid Nanoparticle Vaccine that Non-Covalently Encapsulates a TLR7/8 Agonist and T-Helper Epitope Induces Protective Anti-Fentanyl Immunity. Angew Chem Int Ed Engl 2025; 64:e202419031. [PMID: 39441822 DOI: 10.1002/anie.202419031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 10/25/2024]
Abstract
Opioid use disorder - particularly involving fentanyl - has precipitated a public health crisis characterized by a significant increase in addiction and overdose-related deaths. Fentanyl-specific immunotherapy, which aims at inducing fentanyl-specific antibodies capable of binding fentanyl molecules in the bloodstream, preventing their entry in the central nervous system, is therefore gaining momentum. Conventional opioid designs rely on the covalent conjugation of fentanyl analogues to immunogenic carrier proteins that hold the inherent capacity of mounting immunodominant responses. Here, we present an alternative fentanyl vaccine design that utilizes a non-covalent assembly of lipid nanoparticles (LNPs) to deliver fentanyl haptens in conjunction with a CD4+ T-helper peptide epitope and an imidazoquinoline TLR7/8 agonist. Our results demonstrate that a single intramuscular administration of the LNP-based nanovaccine elicits fentanyl-specific antibodies, significantly mitigating the effects of opioid overdose in preclinical mouse models. Furthermore, we analyzed the immunobiological behavior of the vaccine in vivo in mouse models, providing evidence that covalent attachment of a fentanyl hapten to a carrier proteins or peptide epitope is not necessary for inducing an effective immune response. However, co-delivery - specifically, the physical assembly of all immune cues into an LNP - remains essential for inducing hapten-specific immunity.
Collapse
Affiliation(s)
- Zifu Zhong
- Department of Pharmaceutics, Ghent University, Belgium, Ottergemsesteenweg 460, Gent, 9000, Belgium
| | - Marie H Deventer
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000, Ghent, Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Belgium, Ottergemsesteenweg 460, Gent, 9000, Belgium
| | - Stijn Vanhee
- Department of Internal Medicine and Pediatrics, Ghent University, Technologiepark-Zwijnaarde 71, Gent, Ghent, 9052, Belgium
- Laboratory of immunoregulation and mucosal immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium Ghent, Belgium
- Department of Head and Skin, Ghent University, Gent, Belgium
| | - Inés Lammens
- Department of Internal Medicine and Pediatrics, Ghent University, Technologiepark-Zwijnaarde 71, Gent, Ghent, 9052, Belgium
- Laboratory of immunoregulation and mucosal immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium Ghent, Belgium
- Department of Head and Skin, Ghent University, Gent, Belgium
| | - Kim Deswarte
- Laboratory of immunoregulation and mucosal immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium Ghent, Belgium
| | - Yi Huang
- Department of Pharmaceutics, Ghent University, Belgium, Ottergemsesteenweg 460, Gent, 9000, Belgium
| | - Tingting Ye
- Department of Pharmaceutics, Ghent University, Belgium, Ottergemsesteenweg 460, Gent, 9000, Belgium
| | - Haixiu Wang
- Department of Pharmaceutics, Ghent University, Belgium, Ottergemsesteenweg 460, Gent, 9000, Belgium
| | - Lutz Nuhn
- Institute of Functional Materials and Biofabrication, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Würzburg, 97070, Germany
| | - Marthe M Vandeputte
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000, Ghent, Belgium
| | - Mark Gontsarik
- Department of Pharmaceutics, Ghent University, Belgium, Ottergemsesteenweg 460, Gent, 9000, Belgium
| | - Xiaole Cui
- Laboratory of Gene Therapy, Ghent University, Belgium, Heidestraat 19, Merelbeke, 9820, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Ghent University, Belgium, Heidestraat 19, Merelbeke, 9820, Belgium
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science and Institute of Immunology, Hannover Medical School, 30625, Hannover, Germany
| | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Technologiepark-Zwijnaarde 71, Gent, Ghent, 9052, Belgium
- Laboratory of immunoregulation and mucosal immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium Ghent, Belgium
| | - Antonio P Baptista
- Department of Internal Medicine and Pediatrics, Ghent University, Technologiepark-Zwijnaarde 71, Gent, Ghent, 9052, Belgium
- Laboratory of immunoregulation and mucosal immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium Ghent, Belgium
| | - Christophe P Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000, Ghent, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Belgium, Ottergemsesteenweg 460, Gent, 9000, Belgium
| |
Collapse
|
58
|
Wang Y, Wang S, Huang L, Mao W, Li F, Lin A, Zhao W, Zeng X, Zhang Y, Yang D, Han Y, Li Y, Ren L, Li Y, Zhang L, Yan F, Yang Y, Tang X. A nucleoside-modified rabies mRNA vaccine induces long-lasting and comprehensive immune responses in mice and non-human primates. Mol Ther 2025; 33:548-559. [PMID: 39741409 PMCID: PMC11853375 DOI: 10.1016/j.ymthe.2024.12.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/29/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Rabies is a lethal zoonotic infectious disease. Vaccines against the rabies virus have significantly reduced the number of deaths from the disease. However, all licensed rabies vaccines are inactivated vaccines, which have limited immunogenicity and complicated immunization procedures. A novel vaccine that provides sustained and comprehensive protection is urgently needed. Here, we developed a novel rabies mRNA vaccine candidate containing sequence-optimized mRNAs encoding full-length glycoprotein encapsulated in ionizable lipid nanoparticles. In mice and rhesus macaques, the rabies mRNA exhibited superior immunogenicity over licensed vaccines, especially in inducing long-lasting neutralizing antibodies and memory B cells. A single administration of 1.5 μg mRNA vaccine could provide complete protection against a lethal rabies virus challenge in mice. Additionally, the mRNA vaccine could robustly activate cellular immune responses with moderate release of several cytokines. In summary, our data demonstrated that the rabies mRNA vaccine outperformed approved inactivated vaccines in both mice and rhesus macaques. This highlights the potential of the mRNA platform in developing next-generation rabies vaccines.
Collapse
Affiliation(s)
- Yu Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China; Xuzhou Medical University, Xuzhou 221004, China
| | - Shen Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
| | - Lulu Huang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China
| | - Wenhao Mao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China
| | - Fangmeng Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China
| | - Ang Lin
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China; Institute of Translational Medicine, China Pharmaceutical University, Nanjing 211112, China
| | - Weijun Zhao
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing 211112, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211112, China
| | - Xianhuan Zeng
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211112, China
| | - Yue Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China
| | - Dingcao Yang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China
| | - Yuhong Han
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211112, China
| | - Yidan Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China
| | - Leyuan Ren
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China
| | - Ying Li
- Translational Medicine Research Institute, Yangzhou University, Yangzhou 225001, China
| | - Liang Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China.
| | - Yong Yang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China; Institute of Translational Medicine, China Pharmaceutical University, Nanjing 211112, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211112, China; Xuzhou Medical University, Xuzhou 221004, China.
| | - Xinying Tang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211112, China; Institute of Translational Medicine, China Pharmaceutical University, Nanjing 211112, China.
| |
Collapse
|
59
|
Kawai A, Shimizu T, Tanaka H, Shichinohe S, Anindita J, Hirose M, Kawahara E, Senpuku K, Shimooka M, Quynh Mai LT, Suzuki R, Nogimori T, Yamamoto T, Hirai T, Kato T, Watanabe T, Akita H, Yoshioka Y. Low-inflammatory lipid nanoparticle-based mRNA vaccine elicits protective immunity against H5N1 influenza virus with reduced adverse reactions. Mol Ther 2025; 33:529-547. [PMID: 39690742 PMCID: PMC11852987 DOI: 10.1016/j.ymthe.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024] Open
Abstract
Messenger RNA vaccines based on lipid nanoparticles (mRNA-LNPs) are promising vaccine modalities. However, mRNA-LNP vaccines frequently cause adverse reactions such as swelling and fever in humans, partly due to the inflammatory nature of LNP. Modification of the ionizable lipids used in LNPs is one approach to avoid these adverse reactions. Here, we report the development of mRNA-LNP vaccines with better protective immunity and reduced adverse reactions using LNPs, which contain a disulfide (SS)-cleavable bond and pH-activated lipid-like materials with oleic acid (ssPalmO) as an ionizable lipid (LNPssPalmO). We used mRNA expressing H5N1 subtype high-pathogenicity avian influenza virus-derived hemagglutinin or neuraminidase to generate mRNA-LNP vaccines against H5N1 influenza. Compared with conventional LNPs, mRNA-LNPssPalmO induced comparable antigen-specific antibodies and better interferon-γ (IFN-γ)-producing T helper type 1 responses in mice. Both mRNA-LNPssPalmO and conventional mRNA-LNPs conferred strong protection against homologous H5N1 virus challenge. In addition, mRNA-LNPssPalmO showed better cross-protection against heterologous H5N1 virus challenge compared with conventional mRNA-LNPs. Furthermore, we observed that mRNA-LNPssPalmO induced less-inflammatory responses (e.g., inflammatory cytokine production, vascular hyperpermeability) and fewer adverse reactions (e.g., weight loss, fever) compared with conventional mRNA-LNPs. These results suggest that mRNA-LNPssPalmO would be a safe alternative to conventional vaccines to overcome mRNA-LNP vaccine hesitancy.
Collapse
Affiliation(s)
- Atsushi Kawai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Taro Shimizu
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroki Tanaka
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Shintaro Shichinohe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jessica Anindita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Mika Hirose
- Laboratory for Cryo-EM Structural Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eigo Kawahara
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kota Senpuku
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Makoto Shimooka
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Le Thi Quynh Mai
- Department of Virology, National Institute of Hygiene and Epidemiology, No. 1 Yersin Street, Hanoi 100000, Vietnam
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Toshiro Hirai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kato
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory for Cryo-EM Structural Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tokiko Watanabe
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hidetaka Akita
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, The Research Foundation for Microbial Diseases of Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
60
|
Gnazzo V, Saleh H, Castro ÍA, Boon ACM, Pinto AK, Brien JD, López CB. DDO-adjuvanted influenza A virus nucleoprotein mRNA vaccine induces robust humoral and cellular type 1 immune responses and protects mice from challenge. mBio 2025; 16:e0358924. [PMID: 39692514 PMCID: PMC11796404 DOI: 10.1128/mbio.03589-24] [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: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024] Open
Abstract
A challenge in viral vaccine development is to produce vaccines that generate both neutralizing antibodies to prevent infection and cytotoxic CD8+ T-cells that target conserved viral proteins and can eliminate infected cells to control virus spread. mRNA technology offers an opportunity to design vaccines based on conserved CD8-targeting epitopes, but achieving robust antigen-specific CD8+ T-cells remains a challenge. Here, we tested the viral-derived oligonucleotide DDO268 as an adjuvant in the context of a model influenza A virus (IAV) nucleoprotein (NP) mRNA vaccine in C57BL/6 mice. DDO268 when co-packaged with mRNA in lipid nanoparticles is sensed by RIG I-like receptors and safely induces local type I interferon (IFN) production followed by dendritic cells type 1 activation and migration to the draining lymph nodes. This early response triggered by DDO268 improved the generation of IgG2c antibodies and antigen-specific Th1 CD4+ and CD8+ T-cells (IFNγ+TNFα+IL2+) that provided enhanced protection against lethal IAV challenge. In addition, the inclusion of DDO268 reduced the antigen dose required to achieve protection. These results highlight the potential of DDO268 as an effective mRNA vaccine adjuvant and show that an IAV NP mRNA/DDO268 vaccine is a promising approach for generating protective immunity against conserved internal IAV epitopes.IMPORTANCEVaccines that generate neutralizing antibodies and cytotoxic CD8+ T-cells targeting conserved epitopes are ideal for effective protection against viruses. mRNA vaccines combined with the right adjuvant offer a promising solution to this challenge. We show that the virus-derived oligonucleotide DDO268 enhances antibody and T-cell responses to an influenza A virus (IAV) nucleoprotein mRNA vaccine in mice. DDO268 safely induces local type I interferon production and stimulates dendritic cell activation providing enhanced protection against IAV challenge. In addition, the adjuvant activity of DDO268 allows for the use of lower antigen doses during vaccination.
Collapse
MESH Headings
- Animals
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Mice
- Mice, Inbred C57BL
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Influenza A virus/immunology
- Influenza A virus/genetics
- CD8-Positive T-Lymphocytes/immunology
- mRNA Vaccines/immunology
- mRNA Vaccines/administration & dosage
- Female
- Immunity, Humoral
- Immunity, Cellular
- Antibodies, Neutralizing/blood
- Nucleoproteins/immunology
- Nucleoproteins/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Nucleocapsid Proteins
- Adjuvants, Vaccine/administration & dosage
- RNA-Binding Proteins/immunology
- RNA-Binding Proteins/genetics
- Viral Core Proteins/immunology
- Viral Core Proteins/genetics
- RNA, Messenger/immunology
- RNA, Messenger/genetics
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Victoria Gnazzo
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Hanaa Saleh
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ítalo A. Castro
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Adrianus C. M. Boon
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Amelia K. Pinto
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - James D. Brien
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Carolina B. López
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
61
|
Li B, Jiang AY, Raji I, Atyeo C, Raimondo TM, Gordon AGR, Rhym LH, Samad T, MacIsaac C, Witten J, Mughal H, Chicz TM, Xu Y, McNamara RP, Bhatia S, Alter G, Langer R, Anderson DG. Enhancing the immunogenicity of lipid-nanoparticle mRNA vaccines by adjuvanting the ionizable lipid and the mRNA. Nat Biomed Eng 2025; 9:167-184. [PMID: 37679571 DOI: 10.1038/s41551-023-01082-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/27/2023] [Indexed: 09/09/2023]
Abstract
To elicit optimal immune responses, messenger RNA vaccines require intracellular delivery of the mRNA and the careful use of adjuvants. Here we report a multiply adjuvanted mRNA vaccine consisting of lipid nanoparticles encapsulating an mRNA-encoded antigen, optimized for efficient mRNA delivery and for the enhanced activation of innate and adaptive responses. We optimized the vaccine by screening a library of 480 biodegradable ionizable lipids with headgroups adjuvanted with cyclic amines and by adjuvanting the mRNA-encoded antigen by fusing it with a natural adjuvant derived from the C3 complement protein. In mice, intramuscular or intranasal administration of nanoparticles with the lead ionizable lipid and with mRNA encoding for the fusion protein (either the spike protein or the receptor-binding domain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) increased the titres of antibodies against SARS-CoV-2 tenfold with respect to the vaccine encoding for the unadjuvanted antigen. Multiply adjuvanted mRNA vaccines may improve the efficacy, safety and ease of administration of mRNA-based immunization.
Collapse
MESH Headings
- Animals
- Mice
- Nanoparticles/chemistry
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- RNA, Messenger/immunology
- RNA, Messenger/genetics
- RNA, Messenger/administration & dosage
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19/prevention & control
- COVID-19/immunology
- Lipids/chemistry
- Adjuvants, Immunologic
- Female
- mRNA Vaccines/immunology
- Antibodies, Viral/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Humans
- Mice, Inbred BALB C
- Immunogenicity, Vaccine
- Adjuvants, Vaccine
- Liposomes
Collapse
Affiliation(s)
- Bowen Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Allen Yujie Jiang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Idris Raji
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Theresa M Raimondo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Akiva G R Gordon
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luke H Rhym
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tahoura Samad
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Corina MacIsaac
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob Witten
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Haseeb Mughal
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Taras M Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Yue Xu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Sangeeta Bhatia
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Wyss Institute at Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
62
|
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
|
63
|
Li YR, Lyu Z, Shen X, Fang Y, Yang L. Boosting CAR-T cell therapy through vaccine synergy. Trends Pharmacol Sci 2025; 46:180-199. [PMID: 39755457 DOI: 10.1016/j.tips.2024.12.004] [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: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has transformed the treatment landscape for hematological cancers. However, achieving comparable success in solid tumors remains challenging. Factors contributing to these limitations include the scarcity of tumor-specific antigens (TSAs), insufficient CAR-T cell infiltration, and the immunosuppressive tumor microenvironment (TME). Vaccine-based strategies are emerging as potential approaches to address these challenges, enhancing CAR-T cell expansion, persistence, and antitumor efficacy. In this review, we explore diverse vaccine modalities, including mRNA, peptide, viral vector, and dendritic cell (DC)-based vaccines, and their roles in augmenting CAR-T cell responses. Special focus is given to recent clinical advancements combining mRNA-based vaccines with CAR-T therapy for the treatment of genitourinary cancers. In addition, we discuss crucial considerations for optimizing vaccine dosing, scheduling, and delivery to maximize CAR-T synergy, aiming to refine this combination strategy to improve treatment efficacy and safety.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinyuan Shen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
64
|
Phan AT, Aunins E, Cruz-Morales E, Dwivedi G, Bunkofske M, Eberhard JN, Aldridge DL, Said H, Banda O, Tam Y, Christian DA, Vonderheide RH, Kedl RM, Weissman D, Alameh MG, Hunter CA. The type I IFN-IL-27 axis promotes mRNA vaccine-induced CD8 + T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633383. [PMID: 39896632 PMCID: PMC11785111 DOI: 10.1101/2025.01.16.633383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The ability of lipid nanoparticle (LNP)-delivered mRNA vaccines to induce type I IFNs is critical to promote CD8 + T cell responses. The studies presented here indicate that immunization with nucleoside modified mRNA-LNP vaccines drives myeloid cell expression of the cytokine IL-27, which acts on antigen-specific CD8 + T cells to sustain T cell expansion. In vitro and in vivo studies revealed that type I IFN signaling is necessary for mRNA-LNP-induced IL-27 production, that immunization failed in IL-27 KO mice, and that immunization of IFNAR1-deficient mice with mRNA-LNP particles that also encode IL-27 mRNA restored antigen-specific CD8 + T cell responses. In addition, IL-27 mRNA-LNPs served as an adjuvant that improved cytolytic CD8 + T cell responses and the therapeutic efficacy of mRNA-LNPs to drive anti-pathogen and anti-tumor immunity. These studies highlight the central role of IL-27 in mRNA-LNP induced CD8 + T cell responses and the ability of this cytokine to augment the functionality of the CD8 + T cell response for prophylactic or therapeutic immunization.
Collapse
|
65
|
Imani S, Li X, Chen K, Maghsoudloo M, Jabbarzadeh Kaboli P, Hashemi M, Khoushab S, Li X. Computational biology and artificial intelligence in mRNA vaccine design for cancer immunotherapy. Front Cell Infect Microbiol 2025; 14:1501010. [PMID: 39902185 PMCID: PMC11788159 DOI: 10.3389/fcimb.2024.1501010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Messenger RNA (mRNA) vaccines offer an adaptable and scalable platform for cancer immunotherapy, requiring optimal design to elicit a robust and targeted immune response. Recent advancements in bioinformatics and artificial intelligence (AI) have significantly enhanced the design, prediction, and optimization of mRNA vaccines. This paper reviews technologies that streamline mRNA vaccine development, from genomic sequencing to lipid nanoparticle (LNP) formulation. We discuss how accurate predictions of neoantigen structures guide the design of mRNA sequences that effectively target immune and cancer cells. Furthermore, we examine AI-driven approaches that optimize mRNA-LNP formulations, enhancing delivery and stability. These technological innovations not only improve vaccine design but also enhance pharmacokinetics and pharmacodynamics, offering promising avenues for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Xiaoyan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Keyi Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Xiaoping Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
66
|
Lee WS, Selva KJ, Audsley J, Kent HE, Reynaldi A, Schlub TE, Cromer D, Khoury DS, Peck H, Aban M, Vu MN, Zheng MZ, Chung AW, Koutsakos M, Tan HX, Wheatley AK, Juno JA, Rockman S, Davenport MP, Barr I, Kent SJ. Randomized trial of same- versus opposite-arm coadministration of inactivated influenza and SARS-CoV-2 mRNA vaccines. JCI Insight 2025; 10:e187075. [PMID: 39786918 PMCID: PMC11949032 DOI: 10.1172/jci.insight.187075] [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: 09/13/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUNDThe immunogenicity of current influenza vaccines needs improvement. Inactivated influenza and COVID-19 mRNA vaccines can be coadministered, but randomized controlled trial data are lacking on whether the 2 vaccines are more immunogenic if given in the same arm or opposite arms. Murine studies suggest mRNA vaccines can adjuvant influenza vaccines when coformulated and codelivered.METHODSWe randomly assigned 56 adults to receive the Afluria quadrivalent inactivated influenza and Moderna monovalent SARS-CoV-2 XBB.1.5 mRNA vaccines, either in opposite arms or both in the same arm at the same site. The primary endpoint was the difference in median combined serum hemagglutination inhibition titer to the H1, H3, and B-Vic vaccine influenza strains after vaccination.RESULTSWe found no significant difference in hemagglutination inhibition antibody levels between the groups (P = 0.30), with the same-arm group having a 1.26-fold higher titer than the opposite-arm group. There were no differences in analyses of antibodies against individual influenza strains or in nasal or saliva antibody levels. While both binding and neutralizing antibody titers against SARS-CoV-2 were not significantly different between groups postvaccination, there was a higher fold-change in BA.5 and ancestral strain neutralizing antibodies in the opposite-arm group.CONCLUSIONInfluenza vaccination is equivalently immunogenic if given in the same arm or opposite arms as the SARS-CoV-2 vaccine, but it may be preferable to administer the SARS-CoV-2 vaccine at a different site from influenza vaccines.TRIAL REGISTRATIONAustralian New Zealand Clinical Trials Registry ACTRN12624000445572.FUNDINGAustralian National Health and Medical Research Council, Australian Medical Research Future Fund, and National Institutes of Health (UH2AI176172).
Collapse
Affiliation(s)
- Wen Shi Lee
- Department of Microbiology and Immunology and
| | | | - Jennifer Audsley
- Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | | | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Timothy E. Schlub
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Heidi Peck
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Malet Aban
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Mai Ngoc Vu
- Department of Microbiology and Immunology and
| | | | | | | | | | | | | | - Steven Rockman
- Department of Microbiology and Immunology and
- Vaccine Innovation Unit, CSL Seqirus Ltd, Parkville, Victoria, Australia
| | - Miles P. Davenport
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology and
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
67
|
Quadiri A, Prakash S, Zayou L, Dhanushkodi NR, Chilukuri A, Ryan G, Wang K, Vahed H, Chentoufi AA, BenMohamed L. A Spike-Based mRNA Vaccine Encapsulated in Phospholipid 1,2-Dioleoyl-sn-Glycero-3-PhosphoEthanolamine Containing Lipid Nanoparticles Induced Potent B- and T-Cell Responses Associated with Protection Against SARS-CoV-2 Infection and COVID-19-like Symptoms in Hamsters. Vaccines (Basel) 2025; 13:47. [PMID: 39852826 PMCID: PMC11769137 DOI: 10.3390/vaccines13010047] [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: 10/20/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNPs) have emerged as a promising vaccine strategy, especially for COVID-19. While the LNPs protect mRNA from degradation and efficiently deliver the mRNA to antigen-presenting cells, the effect of lipid composition on the immunogenicity and protective efficacy of mRNA/LNP vaccines is not well characterized. Studies on using the mRNA/LNP platform for vaccines have largely focused on the nucleic acid cargo with less attention paid to the LNP vehicle. Whether the composition and biophysical properties of LNPs impact vaccine performance remains to be fully elucidated. METHODS In the present study, we used SARS-CoV-2 Spike-mRNA as a prototype vaccine to study the effect of four different LNPs with various lipid compositions. RESULTS We demonstrate that when the same Spike-mRNA was delivered in the LNP4 formulation based on phospholipid 1,2-dioleoyl-sn-glycero-3-Phosphoethanolamine, it outperformed other LNPs (LNP1, LNP2, and LNP3) that are based on different lipids. Compared to the other three LNPs, LNP4 (i) enhanced the phenotypic and functional maturation of dendritic cells; (ii) induced strong T-cell responses; (iii) increased the secretion of proinflammatory cytokines and pro-follicular T helper (Tfh) cell cytokines; (iv) induced higher neutralization IgG titers; and (v) provided better protection against SARS-CoV-2 infection and COVID-19-like symptoms in the hamster model. Furthermore, we compared LNP-4 with the commercially available LNPs and found it to provide better T-cell immunity against COVID-19 in hamsters. CONCLUSION This study suggests mRNA vaccines encapsulated in Phospholipid 1,2-Dioleoyl-sn-Glycero-3-PhosphoEthanolamine containing LNPs induced Potent B- and T cell immunity. The mechanisms by which Phospholipid 1,2-Dioleoyl-sn-Glycero-3-PhosphoEthanolamine-based LNPs may activate protective B and T cells are discussed.
Collapse
Affiliation(s)
- Afshana Quadiri
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
| | - Swayam Prakash
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
| | - Latifa Zayou
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
| | - Nisha Rajeswari Dhanushkodi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
| | - Amruth Chilukuri
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
| | - Gemma Ryan
- Precision Nanosystems Inc., Vancouver, BC V6P 6T7, Canada; (G.R.); (K.W.)
| | - Kelly Wang
- Precision Nanosystems Inc., Vancouver, BC V6P 6T7, Canada; (G.R.); (K.W.)
| | - Hawa Vahed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
- Department of Vaccines and Immunotherapies, TechImmune LLC, University Lab Partners, Irvine, CA 92660, USA
| | - Aziz A. Chentoufi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; (A.Q.); (S.P.); (L.Z.); (N.R.D.); (A.C.); (H.V.); (A.A.C.)
- Department of Vaccines and Immunotherapies, TechImmune LLC, University Lab Partners, Irvine, CA 92660, USA
- Division of Infectious Diseases and Hospitalist Program, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
- Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
68
|
Pongma C, Keawvilai P, Boonmee A, Wongprom B, Pattarakankul T, Sittplangkoon C, Alameh MG, Kueanjinda P, Heyes J, Lam K, Saechue B, Ruxrungtham K, Palaga T. Effect of mRNA formulated with lipid nanoparticles on the transcriptomic and epigenetic profiles of F4/80 + liver-associated macrophages. Sci Rep 2025; 15:1146. [PMID: 39774150 PMCID: PMC11706949 DOI: 10.1038/s41598-025-85234-5] [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: 05/24/2024] [Accepted: 01/01/2025] [Indexed: 01/30/2025] Open
Abstract
Delivery of an mRNA formulated with lipid nanoparticles (LNPs) induces robust humoral and cell-mediated branches of the immune response. Depending on the LNP formula, mRNA encoding proteins can be detected in the liver upon intramuscular administration of mRNA/LNP in mice. This study investigated the impact of mRNA/LNP administration on liver-associated macrophages at the transcriptomic and epigenetic levels in a mouse model. An mRNA encoding ovalbumin (OVA) formulated with LNPs, was administered intramuscularly, and a robust OVA-specific antibody was detected in the serum on Day 7. F4/80+ liver-associated macrophages were isolated and subjected to RNA sequencing, which identified 554 genes whose expression levels were altered compared with those in the PBS control group. The expression of genes involved in macrophage inflammatory functions, such as Tnf, Il6 and Marco, were upregulated. Gene ontology enrichment analysis revealed that IL-6/JAK/STAT3 and TNFα/NF-κB hallmarks were significantly enriched, and mRNA/LNP-exposed liver-associated macrophages were characterized as M1-like cells based on the macrophage transcriptomic profiles. Enrichment of the active histone mark H3K4me3 showed that clusters of loci were highly increased in the mRNA/LNP group, indicating an impact of mRNA/LNPs on macrophage epigenetic profiles. The cis-regulatory regions of Tnf, Il6 and Marco showed enrichment of H3K4me3 marks, which correlated well with their increased transcription. Taken together, our data indicated that mRNA/LNP administration via the intramuscular route influences the gene expression and epigenetic profiles of liver-associated macrophages, reflecting its robust ability to induce an immune response.
Collapse
Affiliation(s)
- Chitsuda Pongma
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pornlapat Keawvilai
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Atsadang Boonmee
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Benjawan Wongprom
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thitiporn Pattarakankul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Materials and Biointerfaces, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
| | - Chutamath Sittplangkoon
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Patipark Kueanjinda
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Rama IV Road, Pathumwan, Bangkok, 10330, Thailand
| | - James Heyes
- Genevant Sciences Corporation, Vancouver, BC, Canada
| | - Kieu Lam
- Genevant Sciences Corporation, Vancouver, BC, Canada
| | - Benjawan Saechue
- One Health Research Unit, Faculty of Veterinary Science, Mahasarakham University, Mahasarakham, 44000, Thailand
| | - Kiat Ruxrungtham
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tanapat Palaga
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
69
|
Zhang M, Wang C, Pan J, Cui H, Zhao X. Advancing novel veterinary vaccines: From comprehensive antigen and adjuvant design to preparation process optimization. Int Immunopharmacol 2025; 145:113784. [PMID: 39672026 DOI: 10.1016/j.intimp.2024.113784] [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: 10/15/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
Vaccination stands as the paramount and cost-effective strategy for the prevention and management of animal infectious diseases. With the advances in biological technology, materials science and industrial optimization, substantial progress has been made in the development of innovative veterinary vaccines. A majority of the novel vaccines under current investigation tend to stimulate multiple immune pathways and to achieve long-term resistance against infectious diseases, yet it remains imperative to concentrate research efforts on the efficient utilization of vaccines, mitigating toxic side effects, and ensuring safe production processes. This article presents an overview of research progress in veterinary vaccines, encompassing comprehensive antigen design, adjuvant formulation advancements, preparation process optimization, and rigorous immune efficacy evaluation. It summarizes cutting-edge vaccines derived from in vitro synthesis and in vivo application, emphasizing immunogenic components and immune response mechanisms. It also highlights novel biological adjuvants that enhance immune efficacy, diversify raw materials, and possess targeted functions, while comprehensively exploring advancements in production methodologies and compatible vaccine products. By establishing a foundation for the integrated use of these innovative veterinary vaccines, this work facilitates future interdisciplinary cooperation in their advancement, aiming to accelerate the achievement of herd immunity through concerted efforts.
Collapse
Affiliation(s)
- Meng Zhang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Chunxin Wang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Junqian Pan
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Haixin Cui
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
70
|
Smith OA, Fujimoto B, Wong TAS, To A, Odo T, Ball A, Haun BK, Muramatsu H, Tam YK, Pardi N, Lehrer AT. Impact of Metabolic States on SARS-CoV-2 Vaccine Responses in Mouse Models of Obesity and Diabetes. COVID 2025; 5:2. [PMID: 40406709 PMCID: PMC12097603 DOI: 10.3390/covid5010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
The emergence of SARS-CoV-2 has resulted in a significant impact on public health, particularly for individuals with underlying health conditions such as obesity and diabetes. While vaccination efforts have played a crucial role in reducing hospitalizations, it remains unclear whether the effectiveness of these vaccines varies among different population groups. In this study, we investigated the immune responses generated by various SARS-CoV-2 vaccine platforms in mouse models with obesity and diabetes, focusing on both cell-mediated and humoral immune responses. Our findings revealed diminished immune responses in diabetic and obese mice compared to healthy counterparts. After vaccination with adjuvanted subunit or mRNA lipid nanoparticle (LNP) vaccines, both humoral and cell-mediated responses were significantly reduced in diabetic mice. Obese mice also exhibited decreased immunogenicity, albeit to a lesser extent. However, it should be noted that mRNA vaccines demonstrated strong neutralizing responses across all metabolic states, while adjuvanted subunit vaccines elicited higher antibody avidity in mice with type 2 diabetes (T2D) and obesity compared to healthy mice. These results suggest that the impaired humoral and cell-mediated responses observed in altered metabolic states may be linked to chronic inflammation associated with obesity and suboptimal glycemic control in diabetes. Understanding the impact of these metabolic disturbances on vaccine immunogenicity is crucial for developing optimized vaccines that can effectively enhance immune responses and provide long-lasting protection against SARS-CoV-2, even in individuals with obesity and diabetes. By contributing these findings, we support efforts to improve vaccine efficacy in populations affected by metabolic disorders, advancing effective immunization against SARS-CoV-2.
Collapse
Affiliation(s)
- Olivia A. Smith
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Brent Fujimoto
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Teri Ann S. Wong
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Albert To
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Troy Odo
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Aquena Ball
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Brien K. Haun
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
- Cell and Molecular Biology Graduate Program, University of Hawaii Manoa, Honolulu, HI 96813, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Axel T. Lehrer
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii Manoa, Honolulu, HI 96813, USA
| |
Collapse
|
71
|
Grovenstein P, Bhatnagar N, Kim KH, Pal SS, Le CTT, Raha JR, Liu R, Shin CH, Park BR, Du L, Subbiah J, Wang BZ, Kang SM. Influenza 5xM2e mRNA lipid nanoparticle vaccine confers broad immunity and significantly enhances the efficacy of inactivated split vaccination when coadministered. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:104-114. [PMID: 40073270 PMCID: PMC11844137 DOI: 10.1093/jimmun/vkae013] [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: 07/22/2024] [Accepted: 10/29/2024] [Indexed: 03/14/2025]
Abstract
Current influenza vaccines are not effective in conferring protection against antigenic variants and pandemics. To improve cross-protection of influenza vaccination, we developed a 5xM2e messenger RNA (mRNA) vaccine encoding the tandem repeat conserved ectodomain (M2e) of ion channel protein M2 derived from human, swine, and avian influenza A viruses. The lipid nanoparticle (LNP)-encapsulated 5xM2e mRNA vaccine was immunogenic, eliciting high levels of M2e-specific IgG antibodies, IFN-γ+ T cells, T follicular helper cells, germinal center phenotypic B cells, and plasma cells. The mice with 5xM2e mRNA vaccination were broadly protected against lethal infection regardless of hemagglutinin (H1, H3, H5) subtypes by preventing severe weight loss. Injection of 5xM2e mRNA LNP vaccine induced acute innate responses recruiting monocytes, macrophages, and diverse subsets of dendritic cells. A single dose of combined 5xM2e mRNA LNP and split vaccines resulted in significantly enhanced and sustainable IgG antibody responses to viral antigens and protection against homologous and heterologous viruses. This study provides a new strategy of combined mRNA and seasonal vaccination, significantly enhancing vaccine protective efficacy.
Collapse
Affiliation(s)
- Phillip Grovenstein
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Chau Thuy Tien Le
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Rong Liu
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Chong Hyun Shin
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Bo Ryoung Park
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Lanying Du
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Jeeva Subbiah
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
72
|
Lin W, Shen C, Li M, Ma S, Liu C, Huang J, Ren Z, Yang Y, Zhao M, Xie Q, Guo S, Wang W, Wang K, Ma Q, Jiang Y, Zheng J, Liao Y. Programmable Macrophage Vesicle Based Bionic Self-Adjuvanting Vaccine for Immunization against Monkeypox Virus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408608. [PMID: 39513669 PMCID: PMC11714231 DOI: 10.1002/advs.202408608] [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: 07/25/2024] [Revised: 11/01/2024] [Indexed: 11/15/2024]
Abstract
The emergence of monkeypox has become a global health threat after the COVID-19 pandemic. Due to the lack of available specifically treatment against MPV, developing an available vaccine is thus the most prospective and urgent strategy. Herein, a programmable macrophage vesicle based bionic self-adjuvanting vaccine (AM@AEvs-PB) is first developed for defending against monkeypox virus (MPV). Based on MPV-related antigen-stimulated macrophage-derived vesicles, the nanovaccine is constructed by loading the mature virion (MV)-related intracellular protein (A29L/M1R) and simultaneously modifying with the enveloped virion (EV) antigen (B6R), enabling them to effectively promote antigen presentation and enhance adaptive immune through self-adjuvant strategy. Owing to the synergistic properties of bionic vaccine coloaded MV and EV protein in defensing MPV, the activation ratio of antigen-presenting cells is nearly four times than that of single antigen in the same dose, resulting in stronger immunity in host. Notably, intramuscular injection uptake of AM@AEvs-PB demonstrated vigorous immune-protective effects in the mouse challenge attempt, offering a promising strategy for pre-clinical monkeypox vaccine development.
Collapse
Affiliation(s)
- Weiqiang Lin
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
- Institute for Engineering MedicineKunming Medical UniversityKunming650500P. R. China
| | - Chenguang Shen
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Mengjun Li
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Shengchao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Chenxin Liu
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhou510515P. R. China
| | - Jialin Huang
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Zuning Ren
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Yuechao Yang
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Minghai Zhao
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Qiulin Xie
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Shuang Guo
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Wei Wang
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Kaiyuan Wang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingapore119074Singapore
| | - Qiang Ma
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhou510515P. R. China
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Judun Zheng
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Yuhui Liao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
- Institute for Engineering MedicineKunming Medical UniversityKunming650500P. R. China
| |
Collapse
|
73
|
Yong L, Hutchings C, Barnes E, Klenerman P, Provine NM. Distinct Requirements for CD4 + T Cell Help for Immune Responses Induced by mRNA and Adenovirus-Vector SARS-CoV-2 Vaccines. Eur J Immunol 2025; 55:e202451142. [PMID: 39604225 PMCID: PMC11739681 DOI: 10.1002/eji.202451142] [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: 03/15/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
CD4+ T cells have been established as central orchestrators of cellular and humoral immune responses to infection or vaccination. However, the need for CD4+ T cell help to generate primary CD8+ T cell responses is variable depending on the infectious agent or vaccine and yet consistently required for the recall of CD8+ T cell memory responses or antibody responses. Given the deployment of new vaccine platforms such as nucleoside-modified mRNA vaccines, we sought to elucidate the requirement for CD4+ T cell help in the induction of cellular and antibody responses to mRNA and adenovirus (Ad)-vectored vaccines against SARS-CoV-2. Using antibody-mediated depletion of CD4+ T cells in a mouse immunization model, we observed that CD4+ T cell help was dispensable for both primary and secondary CD8+ T cell responses to the BNT162b2 and mRNA-1273 mRNA vaccines but required for the AZD1222 Ad-vectored vaccine. Nonetheless, CD4+ T cell help was needed by both mRNA and Ad-vectored vaccine platforms for the generation of antibodies, demonstrating the centrality of CD4+ T cells in vaccine-induced protective immunity against SARS-CoV-2. Ultimately, this highlights the shared and distinct regulation of humoral and cellular responses induced by these vaccine platforms.
Collapse
Affiliation(s)
- Lyn Yong
- Pandemic Sciences InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine–Experimental MedicineUniversity of OxfordOxfordUK
| | - Claire Hutchings
- Peter Medawar Building for Pathogen Research, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Eleanor Barnes
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine–Experimental MedicineUniversity of OxfordOxfordUK
- Peter Medawar Building for Pathogen Research, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Paul Klenerman
- Pandemic Sciences InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine–Experimental MedicineUniversity of OxfordOxfordUK
- Peter Medawar Building for Pathogen Research, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Nicholas M. Provine
- Pandemic Sciences InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
- Centre for Human GeneticsNuffield Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
74
|
Zhang Y, Zhai S, Qin S, Chen Y, Chen K, Huang Z, Lan X, Luo Y, Li G, Li H, He X, Chen M, Zhang Z, Peng X, Jiang X, Huang H, Song X. MHCI trafficking signal-based mRNA vaccines strengthening immune protection against RNA viruses. Bioeng Transl Med 2025; 10:e10709. [PMID: 39801759 PMCID: PMC11711215 DOI: 10.1002/btm2.10709] [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: 12/18/2023] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 01/16/2025] Open
Abstract
The major histocompatibility complex class I (MHCI) trafficking signal (MITD) plays a pivotal role in enhancing the efficacy of mRNA vaccines. However, there was a lack of research investigating its efficacy in enhancing immune responses to RNA virus infections. Here, we have developed an innovative strategy for the formulation of mRNA vaccines. This approach involved the integration of MITD into the mRNA sequence encoding the virus antigen. Mechanistically, MITD-based mRNA vaccines can strengthen immune protection by mimicking the dynamic trafficking properties of MHCI molecule and thus expand the memory specific B and T cells. The model MITD-based mRNA vaccines encoding binding receptor-binding domain (RBD) of SARS-CoV-2 were indeed found to achieve protective duration, optimal storage stability, broad efficacy, and high safety.
Collapse
Affiliation(s)
- Yupei Zhang
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Songhui Zhai
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
- Department of PediatricsWest China Second University Hospital, Sichuan UniversityChengduSichuanChina
| | - Shugang Qin
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yuting Chen
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Kepan Chen
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Zhiying Huang
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xing Lan
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yaoyao Luo
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Guohong Li
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Hao Li
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xi He
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of MacauMacauChina
| | - Zhongwei Zhang
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xingchen Peng
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xin Jiang
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Hai Huang
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xiangrong Song
- Department of Critical Care MedicineFrontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
75
|
Cersosimo A, Di Pasquale M, Arabia G, Metra M, Vizzardi E. COVID myocarditis: a review of the literature. Monaldi Arch Chest Dis 2024; 94. [PMID: 37930657 DOI: 10.4081/monaldi.2023.2784] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023] Open
Abstract
Myocarditis is a potentially fatal complication of coronavirus disease 2019 (COVID-19), which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. COVID-19 myocarditis appears to have distinct inflammatory characteristics that distinguish it from other viral etiologies. COVID-19 myocarditis can present with symptoms ranging from dyspnea and chest pain to acute heart failure and death. It is critical to detect any cases of myocarditis, especially fulminant myocarditis, which can be characterized by signs of heart failure and arrhythmias. Serial troponins, echocardiography, and electrocardiograms should be performed as part of the initial workup for suspected myocarditis. The second step in detecting myocarditis is cardiac magnetic resonance imaging and endomyocardial biopsy. Treatment for COVID-19 myocarditis is still debatable; however, combining intravenous immunoglobulins and corticosteroids may be effective, especially in cases of fulminant myocarditis. Overall, more research is needed to determine the incidence of COVID-19 myocarditis, and the use of intravenous immunoglobulins and corticosteroids in combination requires large randomized controlled trials to determine efficacy. The purpose of this review is to summarize current evidence on the subject.
Collapse
Affiliation(s)
- Angelica Cersosimo
- Cardiology Unit, Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia
| | - Mattia Di Pasquale
- Cardiology Unit, Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia
| | - Gianmarco Arabia
- Cardiology Unit, Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia
| | - Marco Metra
- Cardiology Unit, Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia
| | - Enrico Vizzardi
- Cardiology Unit, Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia
| |
Collapse
|
76
|
Grant M, Ni Lee L, Chinnakannan S, Tong O, Kwok J, Cianci N, Tillman L, Saha A, Pereira Almeida V, Leung C. Unlocking cancer vaccine potential: What are the key factors? Hum Vaccin Immunother 2024; 20:2331486. [PMID: 38564321 PMCID: PMC11657071 DOI: 10.1080/21645515.2024.2331486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer is a global health challenge, with changing demographics and lifestyle factors producing an increasing burden worldwide. Screening advancements are enabling earlier diagnoses, but current cancer immunotherapies only induce remission in a small proportion of patients and come at a high cost. Cancer vaccines may offer a solution to these challenges, but they have been mired by poor results in past decades. Greater understanding of tumor biology, coupled with the success of vaccine technologies during the COVID-19 pandemic, has reinvigorated cancer vaccine development. With the first signs of efficacy being reported, cancer vaccines may be beginning to fulfill their potential. Solid tumors, however, present different hurdles than infectious diseases. Combining insights from previous cancer vaccine clinical development and contemporary knowledge of tumor immunology, we ask: who are the 'right' patients, what are the 'right' targets, and which are the 'right' modalities to maximize the chances of cancer vaccine success?
Collapse
|
77
|
Essink BJ, Shapiro C, Isidro MGD, Bradley P, Pragalos A, Bloch M, Santiaguel J, Frias MV, Miyakis S, Alves de Mesquita M, Berrè S, Servais C, Waugh N, Hoffmann C, Baba E, Schönborn-Kellenberger O, Wolz OO, Koch SD, Ganyani T, Boutet P, Mann P, Mueller SO, Ramanathan R, Gaudinski MR, Vanhoutte N. Safety and immunogenicity of a modified mRNA-lipid nanoparticle vaccine candidate against COVID-19: Results from a phase 1, dose-escalation study. Hum Vaccin Immunother 2024; 20:2408863. [PMID: 39422261 PMCID: PMC11492660 DOI: 10.1080/21645515.2024.2408863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
This phase 1, open-label, dose-escalation, multi-center study (NCT05477186) assessed the safety and immunogenicity of a booster dose of an mRNA COVID-19 vaccine (CV0501) encoding the SARS-CoV-2 Omicron BA.1 spike protein. Participants aged ≥ 18 years previously vaccinated with ≥ 2 doses of an mRNA COVID-19 vaccine received CV0501 doses ranging from 12 to 200 μg. After assessment of safety and immunogenicity of the 12 μg dose in 30 adults, 30 adults ≤ 64 years were randomized to receive either a 3 or 6 μg dose. Solicited adverse events (AEs) were collected for 7 days, unsolicited AEs for 28 days, and serious AEs (SAEs), medically attended AEs (MAAEs), and AEs of special interest (AESIs) until day (D) 181 post-vaccination. Serum neutralizing titers specific to SARS-CoV-2 BA.1, wild-type, Delta, and additional Omicron subvariants were assessed at D1, D15, D29, D91, and D181. Of 180 vaccinated participants (mean age: 49.3 years; 57.8% women), 70.6% had prior SARS-CoV-2 infection. Most solicited local (98.1%) and systemic (96.7%) AEs were of mild-to-moderate severity; the most common were injection site pain (57.5%; 33.3-73.3% across groups) and myalgia (36.9%; 13.3-56.7%). Unsolicited AEs were reported by 14.4% (6.7-26.7%) of participants (mild-to-moderate severity in 88.5% of the participants). Three participants (1.7%) reported SAEs, 16.7% (6.7-30.0%) reported MAAEs, and 8.3% (0.0-13.3%) reported AESIs (15 COVID-19 cases), none related to vaccination. Geometric means of serum neutralizing titers increased from baseline to D15 and D29 (dose-dependent), and then decreased over time. The safety and immunogenicity results supported advancement to a phase 2 trial.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark Bloch
- Holdsworth House Medical Practice Sydney, Darlinghurst, New South Wales, Australia
| | - Joel Santiaguel
- Quirino Memorial Medical Center, Quezon, Philippines
- University of the Philippines College of Medicine, Manila, Philippines
| | - Melchor Victor Frias
- De La Salle Medical and Health Sciences Institute, Dasmariñas, Cavite, Philippines
| | - Spiros Miyakis
- Wollongong Hospital, Wollongong, New South Wales, Australia
- Graduate School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
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
|
79
|
Jo H, Jeoung J, Kim W, Jeoung D. Regulating Immune Responses Induced by PEGylated Messenger RNA-Lipid Nanoparticle Vaccine. Vaccines (Basel) 2024; 13:14. [PMID: 39852793 PMCID: PMC11768904 DOI: 10.3390/vaccines13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Messenger RNA (mRNA)-based therapeutics have shown remarkable progress in the treatment and prevention of diseases. Lipid nanoparticles (LNPs) have shown great successes in delivering mRNAs. After an mRNA-LNP vaccine enters a cell via an endosome, mRNA is translated into an antigen, which can activate adaptive immunity. mRNAs can bind to various pattern recognition receptors (PRRs), including toll-like receptors (TLRs), and increase the production of inflammatory cytokines. This review summarizes mechanisms of innate immunity induced by mRNAs. Polyethylene glycol (PEG) has been employed as a component of the mRNA-LNP vaccine. PEGylated nanoparticles display enhanced stability by preventing aggregation of particles. However, PEGylation can cause adverse reactions, including blood clearance (ABC) of nanoparticles via complement activation and anaphylaxis. Mechanisms of PEG-induced ABC phenomenon and anaphylaxis are presented and discussed. There have been studies aimed at reducing immune responses associated with PEG to make safe and effective vaccines. Effects of modifying or replacing PEG in reducing immune responses associated with PEGylated nanoparticles are also discussed. Modifying mRNA can induce immune tolerance, which can prevent hypersensitivity reactions induced by PEGylated mRNA-LNP vaccines. Current progress of immune tolerance induction in association with mRNA-LNP is also summarized. This review might be helpful for developing safe and effective PEGylated mRNA-LNP vaccines.
Collapse
Affiliation(s)
| | | | | | - Dooil Jeoung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (H.J.); (J.J.); (W.K.)
| |
Collapse
|
80
|
Peng S, Zhang Y, Zhao X, Wang Y, Zhang Z, Zhang X, Li J, Zheng H, Zhang Y, Shi H, Li H, Liu L. Pathologic Tissue Injury and Inflammation in Mice Immunized with Plasmid DNA-Encapsulated DOTAP-Based Lipid Nanoparticles. Bioconjug Chem 2024; 35:2015-2026. [PMID: 39656061 DOI: 10.1021/acs.bioconjchem.4c00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Ionizable cationic lipids have been developed to mitigate the toxicity of quaternary ammonium lipids, such as DOTAP. Despite its toxicity, DOTAP can promote localization of lipid nanoparticles (LNPs) in target tissues, serving as one of the ionizable cationic helper lipids. Notably, DOTAP-based nanoadjuvants prepared via microfluidic methods showed a better T-cell response. Previous studies showed that DOTAP-based LNPs prepared by the lipid-film method resulted in obvious adverse events. Therefore, our research focused on evaluating the tissue localization and adverse toxicity of a DOTAP-based delivery system prepared through microfluidic techniques. We assessed the delivery efficacy, biodistribution, inflammatory response, and pathological injury in various tissues. In our study, the plasmid DNA encoding the receptor-binding domain (RBD) of SARS-CoV-2 was encapsulated using a mixture of lipids that included DOTAP, DOPE, cholesterol, and DMG-PEG2000 via microfluidic mixing. The LNP-RBDs were smaller than those prepared via the traditional lipid membrane system. We found that LNP-DNA complexes can be effectively delivered and expressed in muscle tissue, with specific antibodies in serum induced postimmunization. Initial distribution of the liposomes was observed in the muscle and liver. Interestingly, both LNPs and DNA showed sustained presence in the lungs and spleen in the group immunized with DNA-encapsulated DOTAP-based LNPs, whereas lower amounts of DNA were detected in the group immunized with dissociative DNA. We detected obvious inflammatory responses and pathological injuries in the muscle, heart, and liver, and the side effects decreased when the immunization dose decreased. These findings suggest that DOTAP-based LNPs have obvious advantages for targeting the lungs and spleen. Additionally, inflammatory responses and pathological injuries occur in a dose-dependent manner in the muscles, heart, and liver. In conclusion, these findings contribute to the development of an LNP delivery system with DOTAP, highlighting its potential to enhance tissue localization and promote high levels of expression when coordinated with ionizable lipids.
Collapse
Affiliation(s)
- Shasha Peng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Yifan Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| | - Xin Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Yibin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Zihan Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Xin Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Jiali Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Huiwen Zheng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Ying Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Haijing Shi
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Heng Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| | - Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| |
Collapse
|
81
|
Brandi R, Paganelli A, D’Amelio R, Giuliani P, Lista F, Salemi S, Paganelli R. mRNA Vaccines Against COVID-19 as Trailblazers for Other Human Infectious Diseases. Vaccines (Basel) 2024; 12:1418. [PMID: 39772079 PMCID: PMC11680146 DOI: 10.3390/vaccines12121418] [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: 11/06/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
mRNA vaccines represent a milestone in the history of vaccinology, because they are safe, very effective, quick and cost-effective to produce, easy to adapt should the antigen vary, and able to induce humoral and cellular immunity. METHODS To date, only two COVID-19 mRNA and one RSV vaccines have been approved. However, several mRNA vaccines are currently under development for the prevention of human viral (influenza, human immunodeficiency virus [HIV], Epstein-Barr virus, cytomegalovirus, Zika, respiratory syncytial virus, metapneumovirus/parainfluenza 3, Chikungunya, Nipah, rabies, varicella zoster virus, and herpes simplex virus 1 and 2), bacterial (tuberculosis), and parasitic (malaria) diseases. RESULTS RNA viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV)-2, HIV, and influenza, are characterized by high variability, thus creating the need to rapidly adapt the vaccines to the circulating viral strain, a task that mRNA vaccines can easily accomplish; however, the speed of variability may be higher than the time needed for a vaccine to be adapted. mRNA vaccines, using lipid nanoparticles as the delivery system, may act as adjuvants, thus powerfully stimulating innate as well as adaptive immunity, both humoral, which is rapidly waning, and cell-mediated, which is highly persistent. Safety profiles were satisfactory, considering that only a slight increase in prognostically favorable anaphylactic reactions in young females and myopericarditis in young males has been observed. CONCLUSIONS The COVID-19 pandemic determined a shift in the use of RNA: after having been used in medicine as micro-RNAs and tumor vaccines, the new era of anti-infectious mRNA vaccines has begun, which is currently in great development, to either improve already available, but unsatisfactory, vaccines or develop protective vaccines against infectious agents for which no preventative tools have been realized yet.
Collapse
Affiliation(s)
- Rossella Brandi
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | | | | | - Paolo Giuliani
- Poliambulatorio Montezemolo, Ente Sanitario Militare del Ministero Della Difesa Presso la Corte dei Conti, 00195 Rome, Italy;
| | - Florigio Lista
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | - Simonetta Salemi
- Division of Internal Medicine, Azienda Ospedaliero-Universitaria S. Andrea, 00189 Rome, Italy
| | - Roberto Paganelli
- Internal Medicine, Faculty of Medicine and Surgery, Unicamillus, International School of Medicine, 00131 Rome, Italy
| |
Collapse
|
82
|
Abt ER, Lam AK, Noguchi M, Rashid K, McLaughlin J, Teng PL, Tran W, Cheng D, Nesterenko PA, Mao Z, Creech AL, Burton Sojo G, Jeyachandran AV, Tam YK, Henley JE, Comai L, Pardi N, Arumugaswami V, Witte ON, Radu CG, Wu TT. Staggered immunization with mRNA vaccines encoding SARS-CoV-2 polymerase or spike antigens broadens the T cell epitope repertoire. Proc Natl Acad Sci U S A 2024; 121:e2406332121. [PMID: 39589869 PMCID: PMC11626164 DOI: 10.1073/pnas.2406332121] [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: 03/27/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Combining a T cell-targeting mRNA vaccine encoding the conserved SARS-CoV-2 RNA-dependent RNA polymerase, RdRp, with a Spike-encoding mRNA vaccine may offer an additional pathway toward COVID-19 protection. Here, we show that a nucleoside-modified RdRp mRNA vaccine raises robust and durable CD8+ T cell responses in mice. Immunization drives a CD8+ T cell response enriched toward a specific RdRp epitope. Unexpectedly, coadministration of mRNA vaccines encoding RdRp or the Spike Receptor Binding Domain (RBD) dampens RBD-specific immune responses. Contralateral administration reduces the suppression of RBD-specific T cell responses while type I interferon signaling blockade restores RBD-specific antibodies. A staggered immunization strategy maintains both RBD vaccine-mediated antibody and T cell responses as well as protection against lethal SARS-CoV-2 challenge in human ACE2 transgenic mice. In HLA-A2.1 transgenic mice, the RdRp vaccine elicits CD8+ T cell responses against HLA-A*02:01-restricted epitopes recognized by human donor T cells. These results highlight RdRp as a candidate antigen for COVID-19 vaccines. The findings also offer insights into crafting effective multivalent mRNA vaccines to broaden CD8+ T cell responses against SARS-CoV-2 and potentially other viruses with pandemic potential.
Collapse
Affiliation(s)
- Evan R. Abt
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Alex K. Lam
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Miyako Noguchi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Khalid Rashid
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pu-Lin Teng
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Wendy Tran
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pavlo A. Nesterenko
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Zhiyuan Mao
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Amanda L. Creech
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Giselle Burton Sojo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, BCV6T 1Z3, Canada
| | - Jill E. Henley
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Lucio Comai
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | | | - Owen N. Witte
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA90095
- Parker Institute for Cancer Immunotherapy, University of California Los Angeles, Los Angeles, CA90095
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- AIDS Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA90095
| |
Collapse
|
83
|
Dowell W, Dearborn J, Languon S, Miller Z, Kirch T, Paige S, Garvin O, Kjendal L, Harby E, Zuchowski AB, Clark E, Lescieur-Garcia C, Vix J, Schumer A, Mistri SK, Snoke DB, Doiron AL, Freeman K, Toth MJ, Poynter ME, Boyson JE, Majumdar D. Distinct Inflammatory Programs Underlie the Intramuscular Lipid Nanoparticle Response. ACS NANO 2024; 18:33058-33072. [PMID: 39563529 DOI: 10.1021/acsnano.4c08490] [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] [Indexed: 11/21/2024]
Abstract
Developments in mRNA/lipid nanoparticle (LNP) technology have advanced the fields of vaccinology and gene therapy, raising questions about immunogenicity. While some mRNA/LNPs generate an adjuvant-like environment in muscle tissue, other mRNA/LNPs are distinct in their capacity for multiple rounds of therapeutic delivery. We evaluate the adjuvancy of components of mRNA/LNPs by phenotyping cellular infiltrate at injection sites, tracking uptake by immune cells, and assessing the inflammatory state. Delivery of 9 common, but chemically distinct, LNPs to muscle revealed two classes of inflammatory gene expression programs: inflammatory (Class A) and noninflammatory (Class B). We find that intramuscular injection with Class A, but not Class B, empty LNPs (eLNPs) induce robust neutrophil infiltration into muscle within 2 h and a diverse myeloid population within 24 h. Single-cell RNA sequencing revealed SM-102-mediated expression of inflammatory chemokines by myeloid infiltrates within muscle 1 day after injection. Surprisingly, we found direct transfection of muscle infiltrating myeloid cells and splenocytes 24 h after intramuscular mRNA/LNP administration. Transfected myeloid cells within the muscle exhibit an activated phenotype 24 h after injection. Similarly, directly transfected splenic lymphocytes and dendritic cells (DCs) are differentially activated by Class A or Class B containing mRNA/LNP. Within the splenic DC compartment, type II conventional DCs (cDC2s) are directly transfected and activated by Class A mRNA/LNP. Together, we show that mRNA and LNPs work synergistically to provide the necessary innate immune stimuli required for effective vaccination. Importantly, this work provides a design framework for vaccines and therapeutics alike.
Collapse
Affiliation(s)
- William Dowell
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Jacob Dearborn
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Sylvester Languon
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Zachary Miller
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Tylar Kirch
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Stephen Paige
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Olivia Garvin
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Lily Kjendal
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Ethan Harby
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Adam B Zuchowski
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Emily Clark
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Carlos Lescieur-Garcia
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Jesse Vix
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Amy Schumer
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Somen K Mistri
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Deena B Snoke
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Amber L Doiron
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Kalev Freeman
- Department of Emergency Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Michael J Toth
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Matthew E Poynter
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Jonathan E Boyson
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Devdoot Majumdar
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| |
Collapse
|
84
|
Taaffe J, Ostrowsky JT, Mott J, Goldin S, Friede M, Gsell P, Chadwick C. Advancing influenza vaccines: A review of next-generation candidates and their potential for global health impact. Vaccine 2024; 42:126408. [PMID: 39369576 DOI: 10.1016/j.vaccine.2024.126408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/20/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Influenza vaccines are an essential tool for influenza prevention, control and preparedness. However, demand for them and their programmatic suitability globally is significantly influenced by their variable effectiveness against influenza illness annually, limited duration of protection and need for yearly updating and vaccination. As such, the World Health Organization and major funders, such as the United States National Institute of Allergy and Infectious Diseases and Bill and Melinda Gates Foundation, have strongly encouraged developing influenza vaccines with increased efficacy, breadth and duration of protection. Here, we review the next-generation influenza vaccine pipeline, focusing on products in clinical development, and compare their characteristics to currently approved seasonal influenza vaccines. METHODS To identify and characterize next-generation influenza vaccine candidates, we conducted a comprehensive literature review, using the CIDRAP Universal Influenza Vaccine Technology Landscape as a primary reference source and extracting additional information from peer-reviewed manuscripts, clinical trial records and other media in the public domain. RESULTS Our analysis reveals a robust clinical development pipeline for next-generation influenza vaccines, featuring a diversity of approaches to address existing vaccine challenges and several candidates in advanced stages of development. mRNA vaccines emerged as a predominant platform, as evidenced by the number of candidates focused on improved seasonal protection as well as combination vaccine candidates targeting additional respiratory viruses. CONCLUSION While still early in development, results from universal or broadly protective products are promising and warrant continued investment from funders. As most Phase 3 candidates are mRNA-based and include combination vaccines, it is critical to begin considering how these new products may become integrated into the current global influenza vaccine strain selection and manufacturing ecosystems, and existing immunization programmes.
Collapse
Affiliation(s)
| | - Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, USA
| | - Joshua Mott
- World Health Organization, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
85
|
Chatterjee D, Kurup D, Smeyne RJ. Environmental exposures and familial background alter the induction of neuropathology and inflammation after SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626375. [PMID: 39677638 PMCID: PMC11642758 DOI: 10.1101/2024.12.02.626375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Basal ganglia disease has been reported as a post-infection sequela of several viruses, with documentation of this phenomenon from the H1N1 Spanish flu to the recent COVID-19 (SARS-CoV-2) pandemic. SARS-CoV-2 infection leads to multisystem deficits, including those affecting the nervous system. Here, we investigated whether a SARS-CoV-2 infection alone increases the susceptibility to develop parkinsonian phenotypes in C57BL/6J mice expressing the human ACE2 receptor, or in addition to two well-known toxin exposures, MPTP and paraquat. Additionally, we examined mice carrying a G2019S mutation in the LRRK2 gene. We also examined if vaccination with either an mRNA- or protein-based vaccine can alter any observed neuropathology. We find that the infection with the WA-1/2020 (alpha) or omicron B1.1.529 strains in ACE2 and G2019S LRRK2 mice both synergize with a subtoxic exposure to the mitochondrial toxin MPTP to induce neurodegeneration and neuroinflammation in the substantia nigra. This synergy appears toxin-dependent since we do not observe this following exposure to the direct redox-inducing compound paraquat. This synergistic neurodegeneration and neuroinflammation is rescued in WT mice that were vaccinated using either mRNA- and protein- based vaccines directed against the Spike protein of the SARS-CoV-2 virus. However, in the G2019S LRRK2 mutant mice, we find that only the protein-based vaccine but not the mRNA- based vaccine resulted in a rescue of the SARS-CoV-2 mediated neuropathology. Taken together, our results highlight the role of both environmental exposures and familial background on the development of parkinsonian pathology secondary to viral infection and the benefit of vaccines in reducing these risks.
Collapse
|
86
|
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
|
87
|
Ye T, Zhong Z, Cappellesso F, Deswarte K, Chen Y, Lauwers H, De Lombaerde E, Gontsarik M, Lienenklaus S, Van Lysebetten D, Sanders NN, Lambrecht BN, De Koker S, Laoui D, De Geest BG. CO-DELIVERY of glutamic acid-extended peptide antigen and imidazoquinoline TLR7/8 agonist via ionizable lipid nanoparticles induces protective anti-tumor immunity. Biomaterials 2024; 311:122693. [PMID: 38996672 DOI: 10.1016/j.biomaterials.2024.122693] [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: 02/20/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024]
Abstract
Cancer vaccines aim at generating cytotoxic CD8+ T cells that kill cancer cells and confer durable tumor regression. Hereto, CD8+ peptide epitopes should be presented by antigen presenting cells to CD8+ T cells in lymphoid tissue. Unfortunately, in unformulated soluble form, peptide antigens are poorly taken up by antigen presenting cells and do not efficiently reach lymph nodes. Hence, the lack of efficient delivery remains a major limitation for successful clinical translation of cancer vaccination using peptide antigens. Here we propose a generic peptide nanoformulation strategy by extending the amino acid sequence of the peptide antigen epitope with 10 glutamic acid residues. The resulting overall anionic charge of the peptide allows encapsulation into lipid nanoparticles (peptide-LNP) by electrostatic interaction with an ionizable cationic lipid. We demonstrate that intravenous injection of peptide-LNP efficiently delivers the peptide to immune cells in the spleen. Peptide-LNP that co-encapsulate an imidazoquinoline TLR7/8 agonist (IMDQ) induce robust innate immune activation in a broad range of immune cell subsets in the spleen. Peptide-LNP containing the minimal CD8+ T cell epitope of the HPV type 16 E7 oncoprotein and IMDQ induces high levels of antigen-specific CD8+ T cells in the blood, and can confer protective immunity against E7-expressing tumors in both prophylactic and therapeutic settings.
Collapse
Affiliation(s)
- Tingting Ye
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Zifu Zhong
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Federica Cappellesso
- Lab of Cellular and Molecular Immunology, Brussel Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
| | - Kim Deswarte
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Heleen Lauwers
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | | | - Mark Gontsarik
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Stefan Lienenklaus
- Institute of Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | | | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Ghent University, Merelbeke, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Damya Laoui
- Lab of Cellular and Molecular Immunology, Brussel Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium.
| | | |
Collapse
|
88
|
Huang L, Zhao T, Zhao W, Shao A, Zhao H, Ma W, Gong Y, Zeng X, Weng C, Bu L, Di Z, Sun S, Dai Q, Sun M, Wang L, Liu Z, Shi L, Hu J, Fang S, Zhang C, Zhang J, Wang G, Loré K, Yang Y, Lin A. Herpes zoster mRNA vaccine induces superior vaccine immunity over licensed vaccine in mice and rhesus macaques. Emerg Microbes Infect 2024; 13:2309985. [PMID: 38258878 PMCID: PMC10860463 DOI: 10.1080/22221751.2024.2309985] [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: 08/18/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Herpes zoster remains an important global health issue and mainly occurs in aged and immunocompromised individuals with an early exposure history to Varicella Zoster Virus (VZV). Although the licensed vaccine Shingrix has remarkably high efficacy, undesired reactogenicity and increasing global demand causing vaccine shortage urged the development of improved or novel VZV vaccines. In this study, we developed a novel VZV mRNA vaccine candidate (named as ZOSAL) containing sequence-optimized mRNAs encoding full-length glycoprotein E encapsulated in an ionizable lipid nanoparticle. In mice and rhesus macaques, ZOSAL demonstrated superior immunogenicity and safety in multiple aspects over Shingrix, especially in the induction of strong T-cell immunity. Transcriptomic analysis revealed that both ZOSAL and Shingrix could robustly activate innate immune compartments, especially Type-I IFN signalling and antigen processing/presentation. Multivariate correlation analysis further identified several early factors of innate compartments that can predict the magnitude of T-cell responses, which further increased our understanding of the mode of action of two different VZV vaccine modalities. Collectively, our data demonstrated the superiority of VZV mRNA vaccine over licensed subunit vaccine. The mRNA platform therefore holds prospects for further investigations in next-generation VZV vaccine development.
Collapse
Affiliation(s)
- Lulu Huang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Tongyi Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Weijun Zhao
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Andong Shao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, People’s Republic of China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, People’s Republic of China
| | - Wenxuan Ma
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yingfei Gong
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Xianhuan Zeng
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Changzhen Weng
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Lingling Bu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Zhenhua Di
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Shiyu Sun
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Qinsheng Dai
- Targeted Discovery Center, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Minhui Sun
- Targeted Discovery Center, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Limei Wang
- Advanced Medical Research Institute, Shandong University, Jinan, People’s Republic of China
| | - Zhenguang Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People’s Republic of China
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jiesen Hu
- Firestone Biotechnologies, Shanghai, People’s Republic of China
| | - Shentong Fang
- School of Biopharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Cheng Zhang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, People’s Republic of China
| | - Jian Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, People’s Republic of China
| | - Guan Wang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, People’s Republic of China
| | - Karin Loré
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Yong Yang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Ang Lin
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
89
|
Liu J, Sun J, Ding X, Liu W, Wang Y, Wang Z, Peng H, Zhang Y, Su W, Jiang C. A nucleoside-modified mRNA vaccine forming rabies virus-like particle elicits strong cellular and humoral immune responses against rabies virus infection in mice. Emerg Microbes Infect 2024; 13:2389115. [PMID: 39129566 PMCID: PMC11328811 DOI: 10.1080/22221751.2024.2389115] [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: 11/29/2023] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
Rabies is a lethal zoonotic disease that threatens human health. As the only viral surface protein, the rabies virus (RABV) glycoprotein (G) induces main neutralizing antibody (Nab) responses; however, Nab titre is closely correlated with the conformation of G. Virus-like particles (VLP) formed by the co-expression of RABV G and matrix protein (M) improve retention and antigen presentation, inducing broad, durable immune responses. RABV nucleoprotein (N) can elicit humoral and cellular immune responses. Hence, we developed a series of nucleoside-modified RABV mRNA vaccines encoding wild-type G, soluble trimeric RABV G formed by an artificial trimer motif (tG-MTQ), membrane-anchored prefusion-stabilized G (preG). Furthermore, we also developed RABV VLP mRNA vaccine co-expressing preG and M to generate VLPs, and VLP/N mRNA vaccine co-expressing preG, M, and N. The RABV mRNA vaccines induced higher humoral and cellular responses than inactivated rabies vaccine, and completely protected mice against intracerebral challenge. Additionally, the IgG and Nab titres in RABV preG, VLP and VLP/N mRNA groups were significantly higher than those in G and tG-MTQ groups. A single administration of VLP or VLP/N mRNA vaccines elicited protective Nab responses, the Nab titres were significantly higher than that in inactivated rabies vaccine group at day 7. Moreover, RABV VLP and VLP/N mRNA vaccines showed superior capacities to elicit potent germinal centre, long-lived plasma cell and memory B cell responses, which linked to high titre and durable Nab responses. In summary, our data demonstrated that RABV VLP and VLP/N mRNA vaccines could be promising candidates against rabies.
Collapse
MESH Headings
- Animals
- Rabies Vaccines/immunology
- Rabies Vaccines/administration & dosage
- Rabies Vaccines/genetics
- Rabies/prevention & control
- Rabies/immunology
- Rabies virus/immunology
- Rabies virus/genetics
- Mice
- Immunity, Humoral
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Immunity, Cellular
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Female
- mRNA Vaccines/immunology
- Mice, Inbred BALB C
- Nucleosides/immunology
- Glycoproteins/immunology
- Glycoproteins/genetics
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Viral Matrix Proteins/immunology
- Viral Matrix Proteins/genetics
- Antigens, Viral/immunology
- Antigens, Viral/genetics
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/genetics
- RNA, Messenger/genetics
- RNA, Messenger/immunology
Collapse
Affiliation(s)
- Jie Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Jie Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Xue Ding
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Wenhao Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Yipeng Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Zihan Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Hanyu Peng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Weiheng Su
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, People's Republic of China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
90
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
91
|
Tu Z, Wang Y, Liang J, Liu J. Helicobacter pylori-targeted AI-driven vaccines: a paradigm shift in gastric cancer prevention. Front Immunol 2024; 15:1500921. [PMID: 39669583 PMCID: PMC11634812 DOI: 10.3389/fimmu.2024.1500921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024] Open
Abstract
Helicobacter pylori (H. pylori), a globally prevalent pathogen Group I carcinogen, presents a formidable challenge in gastric cancer prevention due to its increasing antimicrobial resistance and strain diversity. This comprehensive review critically analyzes the limitations of conventional antibiotic-based therapies and explores cutting-edge approaches to combat H. pylori infections and associated gastric carcinogenesis. We emphasize the pressing need for innovative therapeutic strategies, with a particular focus on precision medicine and tailored vaccine development. Despite promising advancements in enhancing host immunity, current Helicobacter pylori vaccine clinical trials have yet to achieve long-term efficacy or gain approval regulatory approval. We propose a paradigm-shifting approach leveraging artificial intelligence (AI) to design precision-targeted, multiepitope vaccines tailored to multiple H. pylori subtypes. This AI-driven strategy has the potential to revolutionize antigen selection and optimize vaccine efficacy, addressing the critical need for personalized interventions in H. pylori eradication efforts. By leveraging AI in vaccine design, we propose a revolutionary approach to precision therapy that could significantly reduce H. pylori -associated gastric cancer burden.
Collapse
Affiliation(s)
| | | | | | - Jinping Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
92
|
Catenacci L, Rossi R, Sechi F, Buonocore D, Sorrenti M, Perteghella S, Peviani M, Bonferoni MC. Effect of Lipid Nanoparticle Physico-Chemical Properties and Composition on Their Interaction with the Immune System. Pharmaceutics 2024; 16:1521. [PMID: 39771501 PMCID: PMC11728546 DOI: 10.3390/pharmaceutics16121521] [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: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid nanoparticles (LNPs) have shown promise as a delivery system for nucleic acid-based therapeutics, including DNA, siRNA, and mRNA vaccines. The immune system plays a critical role in the response to these nanocarriers, with innate immune cells initiating an early response and adaptive immune cells mediating a more specific reaction, sometimes leading to potential adverse effects. Recent studies have shown that the innate immune response to LNPs is mediated by Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs), which recognize the lipid components of the nanoparticles. This recognition can trigger the activation of inflammatory pathways and the production of cytokines and chemokines, leading to potential adverse effects such as fever, inflammation, and pain at the injection site. On the other hand, the adaptive immune response to LNPs appears to be primarily directed against the protein encoded by the mRNA cargo, with little evidence of an ongoing adaptive immune response to the components of the LNP itself. Understanding the relationship between LNPs and the immune system is critical for the development of safe and effective nucleic acid-based delivery systems. In fact, targeting the immune system is essential to develop effective vaccines, as well as therapies against cancer or infections. There is a lack of research in the literature that has systematically studied the factors that influence the interaction between LNPs and the immune system and further research is needed to better elucidate the mechanisms underlying the immune response to LNPs. In this review, we discuss LNPs' composition, physico-chemical properties, such as size, shape, and surface charge, and the protein corona formation which can affect the reactivity of the immune system, thus providing a guide for the research on new formulations that could gain a favorable efficacy/safety profile.
Collapse
Affiliation(s)
- Laura Catenacci
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Rachele Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Francesca Sechi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Daniela Buonocore
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Milena Sorrenti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Marco Peviani
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Maria Cristina Bonferoni
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| |
Collapse
|
93
|
Wang L, Wan J, He W, Wang Z, Wu Q, Zhou M, Fu ZF, Zhao L. IL-7 promotes mRNA vaccine-induced long-term immunity. J Nanobiotechnology 2024; 22:716. [PMID: 39550592 PMCID: PMC11568559 DOI: 10.1186/s12951-024-02993-5] [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] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
Messenger RNA (mRNA) vaccines are a key technology in combating existing and emerging infectious diseases. However, improving the immunogenicity and durability of mRNA vaccines remains a challenge. To elicit optimal immune responses, integrating antigen-encoded mRNA and immunostimulatory adjuvants into a single formulation is a promising approach to enhancing the efficacy of mRNA vaccines. Here, we report an adjuvant strategy to enhance the efficacy of mRNA vaccines by co-loading mRNA encoding the antigen (rabies virus glycoprotein, RABV-G) and mRNA encoding IL-7 into lipid nanoparticles, achieving co-delivery to the same antigen-presenting cells. A single immunization with G&IL-7 mRNA vaccine elicited robust humoral immune responses in mice and conferred complete protection against RABV challenge. Notably, the high levels of neutralizing antibody induced by the G&IL-7 mRNA vaccine were maintained for at least 6 months, providing mice with long-term significant and complete protection against RABV. Additionally, IL-7 also enhanced antibody responses against the SARS-CoV-2. These data demonstrate that IL-7 is a potent mRNA vaccine adjuvant that can provide the required immune stimulation in various mRNA vaccine formulations.
Collapse
Affiliation(s)
- Lingli Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jiawu Wan
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenna He
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zongmei Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qiong Wu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ming Zhou
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhen F Fu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ling Zhao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.
- Hubei Hongshan Laboratory, Wuhan, 430070, China.
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China.
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
94
|
Brook B, Checkervarty AK, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The BNT162b2 mRNA vaccine demonstrates reduced age-associated T H1 support in vitro and in vivo. iScience 2024; 27:111055. [PMID: 39569372 PMCID: PMC11576392 DOI: 10.1016/j.isci.2024.111055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 07/05/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024] Open
Abstract
mRNA vaccines demonstrate impaired immunogenicity and durability in vulnerable older populations. We hypothesized that human in vitro modeling and proteomics could elucidate age-specific mRNA vaccine actions. BNT162b2-stimulation changed the plasma proteome of blood samples from young (18-50Y) and older adult (≥60Y) participants, assessed by mass spectrometry, proximity extension assay, and multiplex. Young adult up-regulation (e.g., PSMC6, CPN1) contrasted reduced induction in older adults (e.g., TPM4, APOF, APOC2, CPN1, PI16). 30-85% lower TH1-polarizing cytokines and chemokines were induced in elderly blood (e.g., IFNγ, CXCL10). Analytes lower in older adult samples included human in vivo mRNA immunogenicity biomarkers (e.g., IFNγ, CXCL10, CCL4, IL-1RA). BNT162b2 also demonstrated reduced CD4+ TH1 responses in aged vs. young adult mice. Our study demonstrates the utility of human in vitro platforms modeling age-specific mRNA vaccine immunogenicity, highlights impaired support of TH1 polarization in older adults, and provides a rationale for precision mRNA vaccine adjuvantation to induce greater immunogenicity.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC V6Z 2K5, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amy C Sherman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine- Chair of Pediatrics, University of Rome, 00133 Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dheeraj Soni
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hanno Steen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
95
|
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
|
96
|
Manzo C, Isetta M, Castagna A. Infective agents and polymyalgia rheumatica: key discussion points emerging from a narrative review of published literature. Reumatologia 2024; 62:360-367. [PMID: 39677882 PMCID: PMC11635615 DOI: 10.5114/reum/194687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/16/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction The aetiology of polymyalgia rheumatica (PMR) is unknown. Recently, reports on cases of PMR following the coronavirus disease 2019 (COVID-19) have revived the role of infection as an aetiological or triggering factor. It is estimated that patients with PMR have manifestations of giant cell arteritis (GCA) in < 20% of cases. To date, little is known on the potential role of infectious agents in facilitating this association. Given this background, we performed a review of published literature. Our first aim was to review and discuss the relationship between PMR and infective agents. Secondly, we compared data of PMR-only patients with PMR and overlapping GCA to seek any commonalities or differences regarding the type of infectious agent in these two subgroups. Material and methods We performed a non-systematic literature search on Embase and Medline (COVID interface) with the following search terms: "polymyalgia rheumatica" AND "infections" OR "infectious agents", both MESH headings and free-text (in each language they were written). Each paper's reference list was scanned for additional publications meeting this study's aim. When papers reported data partially presented in previous articles, we referred to the most recent published data. Abstracts submitted at conferences or from non-peer-reviewed sources were not included. Polymyalgia rheumatica following vaccinations was an additional exclusion criterion. Results Several infectious agents have been held responsible for PMR. However, no definite causal link has been identified so far. According to our review, the search for a specific infectious agent, however intriguing, appears to be stagnating. Genetic background and epigenetic regulation probably play a key role. However, topical studies are lacking. Polymyalgia rheumatica as an adverse event following immunization should be kept methodologically distinct from PMR following an acute infection, as the adjuvants in the vaccine can make a significant difference. Conclusions Finally, some infectious agents are able to replicate in human arteries or have an endothelium tropism. Whilst these can theoretically trigger GCA, their role in isolated PMR seems minimal.
Collapse
Affiliation(s)
- Ciro Manzo
- Department of Internal and Geriatric Medicine, Azienda Sanitaria Locale Napoli 3 sud, Rheumatologic Outpatient Clinic, Health District No. 59, Naples, Sant’Agnello, Italy
| | - Marco Isetta
- Central and North West London NHS Trust, England
| | - Alberto Castagna
- Department of Primary Care, Health District of Soverato, Azienda Sanitaria Provinciale Catanzaro, Italy
| |
Collapse
|
97
|
Mebrahtu A, Laurén I, Veerman R, Akpinar GG, Lord M, Kostakis A, Astorga-Wells J, Dahllund L, Olsson A, Andersson O, Persson J, Persson H, Dönnes P, Rockberg J, Mangsbo S. A bispecific CD40 agonistic antibody allowing for antibody-peptide conjugate formation to enable cancer-specific peptide delivery, resulting in improved T proliferation and anti-tumor immunity in mice. Nat Commun 2024; 15:9542. [PMID: 39500897 PMCID: PMC11538452 DOI: 10.1038/s41467-024-53839-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
Current antibody-based immunotherapy depends on tumor antigen shedding for proper T cell priming. Here we select a novel human CD40 agonistic drug candidate and generate a bispecific antibody, herein named BiA9*2_HF, that allows for rapid antibody-peptide conjugate formation. The format is designed to facilitate peptide antigen delivery to CD40 expressing cells combined with simultaneous CD40 agonistic activity. In vivo, the selected bispecific antibody BiA9*2_HF loaded with peptide cargos induces improved antigen-specific proliferation of CD8+ (10-15 fold) and CD4+ T cells (2-7 fold) over control in draining lymph nodes. In both virus-induced and neoantigen-based mouse tumor models, BiA9*2_HF demonstrates therapeutic efficacy and elevated safety profile, with complete tumor clearance, as well as measured abscopal impact on tumor growth. The BiA9*2_HF drug candidate can thus be utilized to tailor immunotherapeutics for cancer patients.
Collapse
Affiliation(s)
- Aman Mebrahtu
- KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
- Strike Pharma AB, Uppsala, Sweden
| | - Ida Laurén
- Strike Pharma AB, Uppsala, Sweden
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | - Martin Lord
- Strike Pharma AB, Uppsala, Sweden
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexandros Kostakis
- Strike Pharma AB, Uppsala, Sweden
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Juan Astorga-Wells
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Leif Dahllund
- KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
- Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden
| | - Anders Olsson
- KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
- Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden
| | - Oscar Andersson
- KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
- Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden
| | - Jonathan Persson
- KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
- Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden
| | - Helena Persson
- KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
- Science for Life Laboratory, Drug Discovery and Development, Stockholm, Sweden
| | - Pierre Dönnes
- Strike Pharma AB, Uppsala, Sweden
- SciCross AB, Skövde, Sweden
| | - Johan Rockberg
- KTH Royal Institute of Technology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden.
- Strike Pharma AB, Uppsala, Sweden.
| | - Sara Mangsbo
- Strike Pharma AB, Uppsala, Sweden.
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
98
|
Wang Q, Su T, Cheng F, Zhou S, Liu X, Wang M, Xu Y, Tang R, Liao S, Dailey J, Xiao G, Yang C, Wen H, Zheng W, Wen B, Tyc KM, Liu J, Sun D, Wang S, Zhu G. Proteolysis-targeting vaccines (PROTAVs) for robust combination immunotherapy of melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.01.616069. [PMID: 39574605 PMCID: PMC11580958 DOI: 10.1101/2024.10.01.616069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Protein/peptide subunit vaccines are promising to promote the tumor therapeutic efficacy of immune checkpoint blockade (ICB). However, current protein/peptide vaccines elicit limited antitumor T cell responses, leading to suboptimal therapeutic efficacy. Here, we present proteolysis-targeting vaccines (PROTAVs) that facilitate antigen proteolytic processing and cross-presentation to potentiate T cell responses for robust ICB combination immunotherapy of melanoma. PROTAVs are modular conjugates of protein/peptide antigens, E3 ligase-binding ligands, and linkers. In antigen-presenting cells (APCs), PROTAVs bind to E3 ligases to rapidly ubiquitinate PROTAV antigens, facilitating antigen proteolytic processing by proteasome, and thereby promoting antigen cross-presentation to T cells and potentiating CD8+ T cell responses. We developed a melanoma PROTAV using a tandem peptide of trivalent melanoma-associated antigens. Co-delivered by lipid nanoparticles (LNPs) with bivalent immunostimulant adjuvants, this PROTAV promotes the quantity and quality of melanoma-specific CD8+ T cells in mice. Further, combining PROTAV and ICB ameliorates the immunosuppressive melanoma microenvironment. As a result, PROTAV and ICB combination enhances melanoma complete regression rates and eradicated 100% large Braf V600E melanoma without recurrence in syngeneic mice. PROTAVs hold the potential for robust tumor combination immunotherapy.
Collapse
Affiliation(s)
- Qiyan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ting Su
- Department of Pharmaceutics and Center for Pharmaceutical Engineering, College of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Furong Cheng
- Department of Pharmaceutics and Center for Pharmaceutical Engineering, College of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Shurong Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiang Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mi Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - You Xu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ri Tang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shimiao Liao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jordan Dailey
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guolan Xiao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chunpeng Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hanning Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Weijia Zheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katarzyna M Tyc
- Department of Biostatistics, School of Public Health; Bioinformatics Shared Resource, Massey Comprehensive Cancer Center; Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jinze Liu
- Department of Biostatistics, School of Public Health; Bioinformatics Shared Resource, Massey Comprehensive Cancer Center; Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shaomeng Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guizhi Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Bioinnovations in Brain Cancer, Biointerfaces Institute; Center for RNA Biomedicine. University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
99
|
Ramadan E, Ahmed A, Naguib YW. Advances in mRNA LNP-Based Cancer Vaccines: Mechanisms, Formulation Aspects, Challenges, and Future Directions. J Pers Med 2024; 14:1092. [PMID: 39590584 PMCID: PMC11595619 DOI: 10.3390/jpm14111092] [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: 09/21/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
After the COVID-19 pandemic, mRNA-based vaccines have emerged as a revolutionary technology in immunization and vaccination. These vaccines have shown remarkable efficacy against the virus and opened up avenues for their possible application in other diseases. This has renewed interest and investment in mRNA vaccine research and development, attracting the scientific community to explore all its other applications beyond infectious diseases. Recently, researchers have focused on the possibility of adapting this vaccination approach to cancer immunotherapy. While there is a huge potential, challenges still remain in the design and optimization of the synthetic mRNA molecules and the lipid nanoparticle delivery system required to ensure the adequate elicitation of the immune response and the successful eradication of tumors. This review points out the basic mechanisms of mRNA-LNP vaccines in cancer immunotherapy and recent approaches in mRNA vaccine design. This review displays the current mRNA modifications and lipid nanoparticle components and how these factors affect vaccine efficacy. Furthermore, this review discusses the future directions and clinical applications of mRNA-LNP vaccines in cancer treatment.
Collapse
Affiliation(s)
- Eslam Ramadan
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, H-6720 Szeged, Hungary;
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Ali Ahmed
- Department of Clinical Pharmacy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
| | - Youssef Wahib Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
100
|
Jia Z, Zhou X, Liu J, De X, Li Y, Yang Z, Duan H, Wang F, Ge J. Immune enhancement of rhamnolipid/manganese calcium phosphate mineralized nanoparticle: A promising subunit antigen delivery system. Int J Biol Macromol 2024; 282:137239. [PMID: 39491710 DOI: 10.1016/j.ijbiomac.2024.137239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
The use of biomimetic mineralization strategy is promising to solve the problem of poor stability and immune effect of subunit antigens. However, non-specifically inducing protein mineralization is still a challenge. we hypothesized that rhamnolipids with both protein and metal binding capacity could be used to develop more functional and biocompatible calcium mineralized nanoparticle (RMCP). The results show that rhamnolipids synergistically enhanced the mineralization of protein with manganese ions and improved 21 % the loading antigens of RMCP compared to manganese calcium phosphate nanoparticles. Transmission electron microscopy (TEM) and Dynamic Light Scattering (DLS) showed particle size of RMCP is 260 ± 12.1 nm with spherical morphology. In vitro experiments have shown that RMCP effectively activate immune cells through the cGAS-STING and NLRP3 pathways and demonstrated a higher level of cytokines in RAW264.7 Macrophages. In vivo, RMCP triggered an increased IgG titer with 16.5-fold IgG2a/IgG1 ratio compared to the aluminum adjuvant which improved the recovery status after challenge in mice. We used biological surfactants for the first time to enhance the biomimetic mineralization process of subunit antigen, which provides a new approach for constructing calcium-based biocompatible antigen delivery vectors, helping to develop a new generation of stable, efficient, and safe subunit vaccines.
Collapse
Affiliation(s)
- Zheng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Xinyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Jingjing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Xinqi De
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Yifan Li
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Zaixing Yang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Haoyuan Duan
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Fang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China.
| |
Collapse
|