1
|
Ahmadivand S, Fux R, Palić D. Role of T Follicular Helper Cells in Viral Infections and Vaccine Design. Cells 2025; 14:508. [PMID: 40214462 PMCID: PMC11987902 DOI: 10.3390/cells14070508] [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: 01/25/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
T follicular helper (Tfh) cells are a specialized subset of CD4+ T lymphocytes that are essential for the development of long-lasting humoral immunity. Tfh cells facilitate B lymphocyte maturation, promote germinal center formation, and drive high-affinity antibody production. Our current knowledge of Tfh interactions with the humoral immune system effectors suggests that they have a critical role in supporting the immune response against viral infections. This review discusses the mechanisms through which Tfh cells influence anti-viral immunity, highlighting their interactions with B cells and their impact on antibody quality and quantity. We explore the role of Tfh cells in viral infections and examine how vaccine design can be improved to enhance Tfh cell responses. Innovative vaccine platforms, such as mRNA vaccines and self-assembling protein nanoplatforms (SAPNs), are promising strategies to enhance Tfh cell activation. Their integration and synergistic combination could further enhance immunity and Tfh responses (SAPN-RNA vaccines). In summary, we provide a comprehensive overview of the current insights into Tfh cells' role during viral infections, emphasizing their potential as strategic targets for innovative vaccine development.
Collapse
Affiliation(s)
- Sohrab Ahmadivand
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University Munich, 80539 Munich, Germany;
| | - Dušan Palić
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| |
Collapse
|
2
|
Liu X. Opportunities and challenges of mRNA technologies in development of dengue virus vaccine. Front Immunol 2025; 16:1520968. [PMID: 40109333 PMCID: PMC11919880 DOI: 10.3389/fimmu.2025.1520968] [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: 10/31/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
Dengue virus (DENV) is a mosquito-borne virus with a significant human health concern. With 390 million infections annually and 96 million showing clinical symptoms, severe dengue can lead to life-threatening conditions like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). The only FDA-approved vaccine, Dengvaxia, has limitations due to antibody-dependent enhancement (ADE), necessitating careful administration. The recent pre-approval of TAK-003 by WHO in 2024 highlights ongoing efforts to improve vaccine options. This review explores recent advancements in dengue vaccine development, emphasizing potential utility of mRNA-based vaccines. By examining current clinical trial data and innovations, we aim to identify promising strategies to address the limitations of existing vaccines and enhance global dengue prevention efforts.
Collapse
Affiliation(s)
- Xiaoyang Liu
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
3
|
Liu S, Wang J, Li Y, Wang M, Du P, Zhang Z, Li W, Sun R, Fan M, Yang M, Yin H. A Multivalent mRNA Therapeutic Vaccine Exhibits Breakthroughs in Immune Tolerance and Virological Suppression of HBV by Stably Presenting the Pre-S Antigen on the Cell Membrane. Pharmaceutics 2025; 17:211. [PMID: 40006578 PMCID: PMC11859219 DOI: 10.3390/pharmaceutics17020211] [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: 12/13/2024] [Revised: 01/14/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: In chronic hepatitis B infection (CHB), the hepatitis B surface antigen (HBsAg) continuously exhausts the hepatitis B surface antibody (HBsAb), which leads to the formation of immune tolerance. Accordingly, the hepatitis B virus (HBV) infection can be blocked by inhibiting the binding of the hepatitis B surface pre-S1/pre-S2 antigen to the hepatocyte receptor NTCP, but the clinical cure rate of pre-S-based vaccines for CHB is limited. Methods: In this study, we designed and prepared multivalent hepatitis B therapeutic mRNA vaccines encoding three hepatitis B surface antigen proteins (L, M, and S) at the cell membrane, verified via in vitro transfection and expression experiments. An in vivo immunization experiment in HBV transgenic (Tg) mice was first completed. Subsequently, an adeno-associated virus plasmid vector carrying the HBV1.2-fold genome (pAAV HBV1.2) model and the adeno-associated virus vector carrying HBV1.3-fold genome (rAAV HBV1.3) model were constructed and immunized with mRNA vaccines. The HBV antigen, antibodies, and HBV DNA in serum were detected. Indirect (enzyme-linked immunosorbent assay) ELISA were made to analyze the activated antigen-specific IgG in HBV Tg mice. Antigen-dependent T-cell activation experiments were carried out, as well as the acute toxicity tests in mice. Results: The L protein/pre-S antigens could be stably presented at the cell membrane with the support of the S protein (and M protein). After vaccinations, the vaccines effectively reactivated the production of high levels of HBsAb, disrupted immune tolerance, and activated the production of high-affinity antibodies against structural pre-S antigen in HBV Tg mice. The HBsAg seroconversion and serum HBV DNA clearance were achieved in two HBV mice models. Furthermore, pre-S antigen-dependent T-cell response against HBV infection was confirmed. The therapeutic vaccine also showed safety in mice. Conclusions: A novel therapeutic mRNA vaccine was developed to break through HBsAg-mediated immune tolerance and treat CHB by stably presenting the pre-S antigen at the membrane, and the vaccine has great potential for the functional cure of CHB.
Collapse
Affiliation(s)
- Shang Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Jie Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Yunxuan Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Muhan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Pei Du
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Zhijie Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Wenguo Li
- Jiangsu Cell Tech Medical Research Institute Co., Ltd., Nanjing 211100, China;
| | - Rongchen Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Mingtao Fan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| | - Meijia Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
- Jiangsu Cell Tech Medical Research Institute Co., Ltd., Nanjing 211100, China;
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (S.L.); (J.W.); (Y.L.); (M.W.); (P.D.); (Z.Z.); (R.S.); (M.F.)
| |
Collapse
|
4
|
Song X, Li Y, Wu H, Qiu H, Sun Y. T-Cell Epitope-Based Vaccines: A Promising Strategy for Prevention of Infectious Diseases. Vaccines (Basel) 2024; 12:1181. [PMID: 39460347 PMCID: PMC11511246 DOI: 10.3390/vaccines12101181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/06/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
With the development of novel vaccine strategies, T-cell epitope-based vaccines have become promising prophylactic and therapeutic tools against infectious diseases that cannot be controlled via traditional vaccines. T-cell epitope-based vaccines leverage specific immunogenic peptides to elicit protective T-cell responses against infectious pathogens. Compared to traditional vaccines, they provide superior efficacy and safety, minimizing the risk of adverse side effects. In this review, we summarized and compared the prediction and identification methods of T-cell epitopes. By integrating bioinformatic prediction and experimental validation, efficient and precise screening of T-cell epitopes can be achieved. Importantly, we delved into the development approaches to diverse T-cell epitope-based vaccines, comparing their merits and demerits, as well as discussing the prevalent challenges and perspectives in their applications. This review offers fresh perspectives for the formulation of safe and efficacious epitope-based vaccines for the devastating diseases against which no vaccines are currently available.
Collapse
Affiliation(s)
| | | | | | - Huaji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.S.); (Y.L.); (H.W.)
| | - Yuan Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.S.); (Y.L.); (H.W.)
| |
Collapse
|
5
|
Jung HG, Jeong S, Kang MJ, Hong I, Park YS, Ko E, Kim JO, Choi DY. Molecular Design of Encapsulin Protein Nanoparticles to Display Rotavirus Antigens for Enhancing Immunogenicity. Vaccines (Basel) 2024; 12:1020. [PMID: 39340050 PMCID: PMC11435836 DOI: 10.3390/vaccines12091020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Rotavirus considerably threatens global health, particularly for children <5 years. Current, licensed oral attenuated vaccine formulations have limitations including insufficient efficacy in children in low- and middle-income countries, warranting urgent development of novel vaccines with improved efficacy and safety profiles. Herein, we present a novel approach utilizing an encapsulin (ENC) nanoparticle (NP)-based non-replicating rotavirus vaccine. ENC, originating from bacteria, offers a self-assembling scaffold that displays rotavirus VP8* antigens on its surface. To enhance the correct folding and soluble expression of monomeric antigens and their subsequent assembly into NP, we adopted an RNA-interacting domain (RID) of mammalian transfer RNA synthetase as an expression tag fused to the N-terminus of the ENC-VP8* fusion protein. Using the RID-ENC-VP8* tripartite modular design, insertion of linkers of appropriate length and sequence and the universal T cell epitope P2 remarkably improved the production yield and immunogenicity. Cleavage of the RID rendered a homogenous assembly of ENC-P2-VP8* into protein NPs. Immunization with ENC-P2-VP8* induced markedly higher levels of VP8*-specific antibodies and virus neutralization titers in mice than those induced by P2-VP8* without ENC. Altogether, these results highlight the potential of the designed ENC NP-based rotavirus vaccine as an effective strategy against rotavirus disease to address global health challenges.
Collapse
Affiliation(s)
| | - Seonghun Jeong
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Min-Ji Kang
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Ingi Hong
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Young-Shin Park
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Eunbyeol Ko
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Jae-Ouk Kim
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | | |
Collapse
|
6
|
Uprety T, Soni S, Sreenivasan C, Hause BM, Naveed A, Ni S, Graves AJ, Morrow JK, Meade N, Mellits KH, Adam E, Kennedy MA, Wang D, Li F. Genetic and antigenic characterization of two diarrhoeicdominant rotavirus A genotypes G3P[12] and G14P[12] circulating in the global equine population. J Gen Virol 2024; 105:002016. [PMID: 39163114 PMCID: PMC11335307 DOI: 10.1099/jgv.0.002016] [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: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
Equine rotavirus species A (ERVA) G3P[12] and G14P[12] are two dominant genotypes that cause foal diarrhoea with a significant economic impact on the global equine industry. ERVA can also serve as a source of novel (equine-like) rotavirus species A (RVA) reassortants with zoonotic potential as those identified previously in 2013-2019 when equine G3-like RVA was responsible for worldwide outbreaks of severe gastroenteritis and hospitalizations in children. One hurdle to ERVA research is that the standard cell culture system optimized for human rotavirus replication is not efficient for isolating ERVA. Here, using an engineered cell line defective in antiviral innate immunity, we showed that both equine G3P[12] and G14P[12] strains can be rapidly isolated from diarrhoeic foals. The genome sequence analysis revealed that both G3P[12] and G14P[12] strains share the identical genotypic constellation except for VP7 and VP6 segments in which G3P[12] possessed VP7 of genotype G3 and VP6 of genotype I6 and G14P[12] had the combination of VP7 of genotype G14 and VP6 of genotype I2. Further characterization demonstrated that two ERVA genotypes have a limited cross-neutralization. The lack of an in vitro broad cross-protection between both genotypes supported the increased recent diarrhoea outbreaks due to equine G14P[12] in foals born to dams immunized with the inactivated monovalent equine G3P[12] vaccine. Finally, using the structural modelling approach, we provided the genetic basis of the antigenic divergence between ERVA G3P[12] and G14P[12] strains. The results of this study will provide a framework for further investigation of infection biology, pathogenesis and cross-protection of equine rotaviruses.
Collapse
Affiliation(s)
- Tirth Uprety
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shalini Soni
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Chithra Sreenivasan
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Ben M. Hause
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Ahsan Naveed
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shuisong Ni
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA
| | - Amy J. Graves
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Jennifer K. Morrow
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Nathan Meade
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Kenneth H. Mellits
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Emma Adam
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Michael A. Kennedy
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Dan Wang
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Feng Li
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| |
Collapse
|
7
|
Hendricks GG, Grigoryan L, Navarro MJ, Catanzaro NJ, Hubbard ML, Powers JM, Mattocks M, Treichel C, Walls AC, Lee J, Ellis D, Wang JY(J, Cheng S, Miranda MC, Valdez A, Chao CW, Chan S, Men C, Johnson MR, Hui H, Wu SY, Lujan V, Muramatsu H, Lin PJ, Sung MM, Tam YK, Leaf EM, Pardi N, Baric RS, Pulendran B, Veesler D, Schäfer A, King NP. Computationally designed mRNA-launched protein nanoparticle vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604655. [PMID: 39091730 PMCID: PMC11291046 DOI: 10.1101/2024.07.22.604655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Both protein nanoparticle and mRNA vaccines were clinically de-risked during the COVID-19 pandemic1-6. These vaccine modalities have complementary strengths: antigen display on protein nanoparticles can enhance the magnitude, quality, and durability of antibody responses7-10, while mRNA vaccines can be rapidly manufactured11 and elicit antigen-specific CD4 and CD8 T cells12,13. Here we leverage a computationally designed icosahedral protein nanoparticle that was redesigned for optimal secretion from eukaryotic cells14 to develop an mRNA-launched nanoparticle vaccine for SARS-CoV-2. The nanoparticle, which displays 60 copies of a stabilized variant of the Wuhan-Hu-1 Spike receptor binding domain (RBD)15, formed monodisperse, antigenically intact assemblies upon secretion from transfected cells. An mRNA vaccine encoding the secreted RBD nanoparticle elicited 5- to 28-fold higher levels of neutralizing antibodies than an mRNA vaccine encoding membrane-anchored Spike, induced higher levels of CD8 T cells than the same immunogen when delivered as an adjuvanted protein nanoparticle, and protected mice from vaccine-matched and -mismatched SARS-CoV-2 challenge. Our data establish that delivering protein nanoparticle immunogens via mRNA vaccines can combine the benefits of each modality and, more broadly, highlight the utility of computational protein design in genetic immunization strategies.
Collapse
Affiliation(s)
- Grace G. Hendricks
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Lilit Grigoryan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Mary Jane Navarro
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Nicholas J. Catanzaro
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miranda L. Hubbard
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M. Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melissa Mattocks
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Catherine Treichel
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Jimin Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Daniel Ellis
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Jing Yang (John) Wang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Suna Cheng
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Marcos C. Miranda
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Adian Valdez
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Cara W. Chao
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Christine Men
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Max R. Johnson
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Harold Hui
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Sheng-Yang Wu
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Victor Lujan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Elizabeth M. Leaf
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Lead contact
| |
Collapse
|
8
|
Cárcamo-Calvo R, Boscá-Sánchez I, López-Navarro S, Navarro-Lleó N, Peña-Gil N, Santiso-Bellón C, Buesa J, Gozalbo-Rovira R, Rodríguez-Díaz J. Immunogenicity of a Rotavirus VP8* Multivalent Subunit Vaccine in Mice. Viruses 2024; 16:1135. [PMID: 39066297 PMCID: PMC11281511 DOI: 10.3390/v16071135] [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: 05/15/2024] [Revised: 07/09/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Rotavirus remains a significant public health threat, especially in low-income countries, where it is the leading cause of severe acute childhood gastroenteritis, contributing to over 128,500 deaths annually. Although the introduction of the Rotarix and RotaTeq vaccines in 2006 marked a milestone in reducing mortality rates, approximately 83,158 preventable deaths persisted, showing ongoing challenges in vaccine accessibility and effectiveness. To address these issues, a novel subcutaneous vaccine formulation targeting multiple rotavirus genotypes has been developed. This vaccine consists of nine VP8* proteins from nine distinct rotavirus genotypes and sub-genotypes (P[4], P[6], P[8]LI, P[8]LIII, P[8]LIV, P[9], P[11], P[14], and P[25]) expressed in E. coli. Two groups of mice were immunized either with a single immunogen, the VP8* from the rotavirus Wa strain (P[8]LI), or with the nonavalent formulation. Preliminary results from mouse immunization studies showed promising outcomes, eliciting antibody responses against six of the nine immunogens. Notably, significantly higher antibody titers against VP8* P[8]LI were observed in the group immunized with the nonavalent vaccine compared to mice specifically immunized against this genotype alone. Overall, the development of parenteral vaccines targeting multiple rotavirus genotypes represents a promising strategy in mitigating the global burden of rotavirus-related morbidity and mortality, offering new avenues for disease prevention and control.
Collapse
Affiliation(s)
- Roberto Cárcamo-Calvo
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Irene Boscá-Sánchez
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
| | - Sergi López-Navarro
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Noemi Navarro-Lleó
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
| | - Nazaret Peña-Gil
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Cristina Santiso-Bellón
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Javier Buesa
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Roberto Gozalbo-Rovira
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (R.C.-C.); (I.B.-S.); (S.L.-N.); (N.N.-L.); (N.P.-G.); (C.S.-B.); (J.B.); (R.G.-R.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| |
Collapse
|
9
|
Woodyear S, Chandler TL, Kawagishi T, Lonergan TM, Patel VA, Williams CA, Permar SR, Ding S, Caddy SL. Chimeric Viruses Enable Study of Antibody Responses to Human Rotaviruses in Mice. Viruses 2024; 16:1145. [PMID: 39066309 PMCID: PMC11281508 DOI: 10.3390/v16071145] [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: 05/14/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
The leading cause of gastroenteritis in children under the age of five is rotavirus infection, accounting for 37% of diarrhoeal deaths in infants and young children globally. Oral rotavirus vaccines have been widely incorporated into national immunisation programs, but whilst these vaccines have excellent efficacy in high-income countries, they protect less than 50% of vaccinated individuals in low- and middle-income countries. In order to facilitate the development of improved vaccine strategies, a greater understanding of the immune response to existing vaccines is urgently needed. However, the use of mouse models to study immune responses to human rotavirus strains is currently limited as rotaviruses are highly species-specific and replication of human rotaviruses is minimal in mice. To enable characterisation of immune responses to human rotavirus in mice, we have generated chimeric viruses that combat the issue of rotavirus host range restriction. Using reverse genetics, the rotavirus outer capsid proteins (VP4 and VP7) from either human or murine rotavirus strains were encoded in a murine rotavirus backbone. Neonatal mice were infected with chimeric viruses and monitored daily for development of diarrhoea. Stool samples were collected to quantify viral shedding, and antibody responses were comprehensively evaluated. We demonstrated that chimeric rotaviruses were able to efficiently replicate in mice. Moreover, the chimeric rotavirus containing human rotavirus outer capsid proteins elicited a robust antibody response to human rotavirus antigens, whilst the control chimeric murine rotavirus did not. This chimeric human rotavirus therefore provides a new strategy for studying human-rotavirus-specific immunity to the outer capsid, and could be used to investigate factors causing variability in rotavirus vaccine efficacy. This small animal platform therefore has the potential to test the efficacy of new vaccines and antibody-based therapeutics.
Collapse
Affiliation(s)
- Sarah Woodyear
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | - Tawny L. Chandler
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | - Takahiro Kawagishi
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63101, USA
| | - Tom M. Lonergan
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | - Vanshika A. Patel
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | | | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10001, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63101, USA
| | - Sarah L. Caddy
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| |
Collapse
|
10
|
Hensley C, Roier S, Zhou P, Schnur S, Nyblade C, Parreno V, Frazier A, Frazier M, Kiley K, O’Brien S, Liang Y, Mayer BT, Wu R, Mahoney C, McNeal MM, Petsch B, Rauch S, Yuan L. mRNA-Based Vaccines Are Highly Immunogenic and Confer Protection in the Gnotobiotic Pig Model of Human Rotavirus Diarrhea. Vaccines (Basel) 2024; 12:260. [PMID: 38543894 PMCID: PMC10974625 DOI: 10.3390/vaccines12030260] [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: 01/16/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 04/01/2024] Open
Abstract
Human rotavirus (HRV) is still a leading cause of severe dehydrating gastroenteritis globally, particularly in infants and children. Previously, we demonstrated the immunogenicity of mRNA-based HRV vaccine candidates expressing the viral spike protein VP8* in rodent models. In the present study, we assessed the immunogenicity and protective efficacy of two mRNA-based HRV trivalent vaccine candidates, encoding VP8* of the genotypes P[8], P[6], or P[4], in the gnotobiotic (Gn) pig model of Wa (G1P[8]) HRV infection and diarrhea. Vaccines either encoded VP8* alone fused to the universal T-cell epitope P2 (P2-VP8*) or expressed P2-VP8* as a fusion protein with lumazine synthase (LS-P2-VP8*) to allow the formation and secretion of protein particles that present VP8* on their surface. Gn pigs were randomly assigned into groups and immunized three times with either P2-VP8* (30 µg) or LS-P2-VP8* (30 µg or 12 µg). A trivalent alum-adjuvanted P2-VP8* protein vaccine or an LNP-formulated irrelevant mRNA vaccine served as the positive and negative control, respectively. Upon challenge with virulent Wa HRV, a significantly shortened duration and decreased severity of diarrhea and significant protection from virus shedding was induced by both mRNA vaccine candidates compared to the negative control. Both LS-P2-VP8* doses induced significantly higher VP8*-specific IgG antibody titers in the serum after immunizations than the negative as well as the protein control. The P[8] VP8*-specific IgG antibody-secreting cells in the ileum, spleen, and blood seven days post-challenge, as well as VP8*-specific IFN-γ-producing T-cell numbers increased in all three mRNA-vaccinated pig groups compared to the negative control. Overall, there was a clear tendency towards improved responses in LS-P2-VP8* compared to the P2-VP8*mRNA vaccine. The demonstrated strong humoral immune responses, priming for effector T cells, and the significant reduction of viral shedding and duration of diarrhea in Gn pigs provide a promising proof of concept and may provide guidance for the further development of mRNA-based rotavirus vaccines.
Collapse
Affiliation(s)
- Casey Hensley
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Sandro Roier
- CureVac SE, 72076 Tübingen, Germany; (S.R.); (B.P.); (S.R.)
| | - Peng Zhou
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Sofia Schnur
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Charlotte Nyblade
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Viviana Parreno
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Annie Frazier
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Maggie Frazier
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Kelsey Kiley
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Samantha O’Brien
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Yu Liang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| | - Bryan T. Mayer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (B.T.M.); (R.W.); (C.M.)
| | - Ruizhe Wu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (B.T.M.); (R.W.); (C.M.)
| | - Celia Mahoney
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (B.T.M.); (R.W.); (C.M.)
| | - Monica M. McNeal
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | | | - Susanne Rauch
- CureVac SE, 72076 Tübingen, Germany; (S.R.); (B.P.); (S.R.)
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; (C.H.); (P.Z.); (S.S.); (C.N.); (V.P.); (A.F.); (M.F.); (K.K.); (S.O.); (Y.L.)
| |
Collapse
|
11
|
Lu C, Li Y, Chen R, Hu X, Leng Q, Song X, Lin X, Ye J, Wang J, Li J, Yao L, Tang X, Kuang X, Zhang G, Sun M, Zhou Y, Li H. Safety, Immunogenicity, and Mechanism of a Rotavirus mRNA-LNP Vaccine in Mice. Viruses 2024; 16:211. [PMID: 38399987 PMCID: PMC10892174 DOI: 10.3390/v16020211] [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: 12/16/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Rotaviruses (RVs) are a major cause of diarrhea in young children worldwide. The currently available and licensed vaccines contain live attenuated RVs. Optimization of live attenuated RV vaccines or developing non-replicating RV (e.g., mRNA) vaccines is crucial for reducing the morbidity and mortality from RV infections. Herein, a nucleoside-modified mRNA vaccine encapsulated in lipid nanoparticles (LNP) and encoding the VP7 protein from the G1 type of RV was developed. The 5' untranslated region of an isolated human RV was utilized for the mRNA vaccine. After undergoing quality inspection, the VP7-mRNA vaccine was injected by subcutaneous or intramuscular routes into mice. Mice received three injections in 21 d intervals. IgG antibodies, neutralizing antibodies, cellular immunity, and gene expression from peripheral blood mononuclear cells were evaluated. Significant differences in levels of IgG antibodies were not observed in groups with adjuvant but were observed in groups without adjuvant. The vaccine without adjuvant induced the highest antibody titers after intramuscular injection. The vaccine elicited a potent antiviral immune response characterized by antiviral clusters of differentiation CD8+ T cells. VP7-mRNA induced interferon-γ secretion to mediate cellular immune responses. Chemokine-mediated signaling pathways and immune response were activated by VP7-mRNA vaccine injection. The mRNA LNP vaccine will require testing for protective efficacy, and it is an option for preventing rotavirus infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yan Zhou
- Correspondence: (Y.Z.); (H.L.); Tel.: +86-13888340684 (Y.Z.); +86-13888918945 (H.L.)
| | - Hongjun Li
- Correspondence: (Y.Z.); (H.L.); Tel.: +86-13888340684 (Y.Z.); +86-13888918945 (H.L.)
| |
Collapse
|