1
|
J LAA, Pa P, Seng CY, Rhee JH, Lee SE. Protein nanocages: A new frontier in mucosal vaccine delivery and immune activation. Hum Vaccin Immunother 2025; 21:2492906. [PMID: 40353600 PMCID: PMC12077460 DOI: 10.1080/21645515.2025.2492906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/15/2025] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Mucosal infectious diseases represent a significant global health burden, impacting millions of people worldwide through pathogens that invade the respiratory, gastrointestinal, and urogenital tracts. Mucosal vaccines provide a promising strategy to combat these diseases by preventing pathogens from entering through the portals as well as within the systemic response compartment. However, challenges such as antigen instability, inefficient delivery, suboptimal immune activation, and the complex biology of mucosal barriers hinder their development. These limitations require integrating specialized adjuvants and delivery systems. Protein nanocages, self-assembling nanoscale structures that can be engineered, may provide an innovative solution for co-delivering antigens and adjuvants. With their remarkable stability, biocompatibility, and design versatility, protein nanocages can potentially overcome existing challenges in mucosal vaccine delivery and enhance protective immune responses. This review highlights the potential of protein nanocages to revolutionize mucosal vaccine development by addressing these challenges.
Collapse
Affiliation(s)
- Lavanya Agnes Angalene J
- Department of Biomedical Sciences, Chonnam National University, Hwasun-gun, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Republic of Korea
| | - Paopachapich Pa
- Department of Biomedical Sciences, Chonnam National University, Hwasun-gun, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Republic of Korea
| | - Chheng Y Seng
- Department of Biomedical Sciences, Chonnam National University, Hwasun-gun, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Republic of Korea
| | - Joon Haeng Rhee
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Republic of Korea
| | - Shee Eun Lee
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
2
|
Johnson VJ, Ryan K, Luster MI, Pandiri A, Hobbie K, Cora M, Shockley KR, Burleson GR, Xie G, Germolec DR. Developmental immunotoxicity study of tris(chloropropyl) phosphate in Hsd:Sprague Dawley SD rats exposed through dosed feed. Toxicol Sci 2025; 205:166-179. [PMID: 39908456 PMCID: PMC12038234 DOI: 10.1093/toxsci/kfaf006] [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] [Indexed: 02/07/2025] Open
Abstract
Tris(chloropropyl) phosphate (TCPP) is a member of organophosphate flame retardants used commonly as a replacement for polybrominated diphenyl ethers in consumer and commercial products. Flame retardants have been shown to modulate immune function in vivo and in vitro and there is evidence that at least some related compounds such as organophosphate pesticides can cause developmental immunotoxicity. Developmental immunotoxicology studies were conducted by administering 0, 2500, 5000, or 10,000 ppm TCPP in feed to pregnant Hsd:Sprague Dawley SD rats from gestation day 6 through weaning on postnatal day 28. Feed exposure to TCPP was continued in the F1 offspring until terminal euthanasia at ∼16 to 21 weeks of age when assessments for developmental immunotoxicity were conducted. Innate, humoral, and cell-mediated immune function were assessed in the F1 adults. The antibody-forming cells (AFCs) response to sheep red blood cells was reduced in male and female F1 rats in the 10,000 ppm treatment group but coincided with reduced bodyweights. The AFC response was also significantly reduced in male rats exposed to 5000 ppm where only moderate effects on bodyweights occurred. TCPP exposure affected baseline T-cell proliferation without stimulation; however, the relevance of this change for immunotoxicity risk is unknown. TCPP exposure did not affect cytotoxic T-lymphocyte activity. Only minor and inconsistent treatment-related effects on hematology, innate NK cell function, and immune cell population distributions in the spleen were observed. Taken together, these data indicate that TCPP has the potential to impact humoral immune responses following developmental exposure.
Collapse
Affiliation(s)
- Victor J Johnson
- Burleson Research Technologies, Inc, Morrisville, NC 27560, United States
| | - Kristen Ryan
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| | - Michael I Luster
- Burleson Research Technologies, Inc, Morrisville, NC 27560, United States
| | - Arun Pandiri
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| | | | - Michelle Cora
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| | - Keith R Shockley
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| | - Gary R Burleson
- Burleson Research Technologies, Inc, Morrisville, NC 27560, United States
| | | | - Dori R Germolec
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| |
Collapse
|
3
|
Jha K, Jaishwal P, Yadav TP, Singh SP. Self-assembling of coiled-coil peptides into virus-like particles: Basic principles, properties, design, and applications with special focus on vaccine design and delivery. Biophys Chem 2025; 318:107375. [PMID: 39674128 DOI: 10.1016/j.bpc.2024.107375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Self-assembling peptide nanoparticles (SAPN) based delivery systems, including virus-like particles (VLP), have shown great potential for becoming prominent in next-generation vaccine and drug development. The VLP can mimic properties of natural viral capsid in terms of size (20-200 nm), geometry (i.e., icosahedral structures), and the ability to generate a robust immune response (with multivalent epitopes) through activation of innate and/or adaptive immune signals. In this regard, coiled-coil (CC) domains are suitable building blocks for designing VLP because of their programmable interaction specificity, affinity, and well-established sequence-to-structure relationships. Generally, two CC domains with different oligomeric states (trimer and pentamer) are fused to form a monomeric protein through a short, flexible spacer sequence. By using combinations of symmetry axes (2-, 3- and 5- folds) that are unique to the geometry of the desired protein cage, it is possible, in principle, to assemble well-defined protein cages like VLP. In this review, we have discussed the crystallographic rules and the basic principles involved in the design of CC-based VLP. It also explored the functions of numerous noncovalent interactions in generating stable VLP structures, which play a crucial role in improving the properties of vaccine immunogenicity, drug delivery, and 3D cell culturing.
Collapse
Affiliation(s)
- Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Thakur Prasad Yadav
- Department of Physics, Faculty of Science, University of Allahabad, Prayagraj 211002, India.
| | | |
Collapse
|
4
|
Ma Q, Yang J, Zhang X, Li H, Hao Y, Feng X. Immunogenicity of HIV-1 Env mRNA and Env-Gag VLP mRNA Vaccines in Mice. Vaccines (Basel) 2025; 13:84. [PMID: 39852863 PMCID: PMC11768961 DOI: 10.3390/vaccines13010084] [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: 12/10/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND The development of a protective vaccine is critical for conclusively ending the human immunodeficiency virus (HIV) epidemic. METHODS We constructed nucleotide-modified mRNA vaccines expressing HIV-1 Env and Gag proteins. Env-gag virus-like particles (VLPs) were generated through co-transfection with env and gag mRNA vaccines. BALB/c mice were immunized with env mRNA, env-gag VLP mRNA, env plasmid DNA vaccine, or lipid nanoparticle (LNP) controls. HIV Env-specific binding and neutralizing antibodies in mouse sera were assessed via enzyme-linked immunosorbent assay (ELISA) and pseudovirus-based neutralization assays, respectively. Env-specific cellular immune responses in mouse splenocytes were evaluated using an Enzyme-linked immunosorbent assay (ELISpot) and in vivo cytotoxic T cell-killing assays. RESULTS The Env-specific humoral and cellular immune responses elicited by HIV-1 env mRNA and env-gag VLP mRNA vaccine were stronger than those induced by the DNA vaccine. Specific immune responses induced by the env mRNA vaccine were significantly stronger in the high-dose group than in the low-dose group. Immunization with co-formulated env and gag mRNAs elicited superior cellular immune responses compared to env mRNA alone. CONCLUSIONS These findings suggest that the env-gag VLP mRNA platform holds significant promise for HIV-1 vaccine development.
Collapse
Affiliation(s)
| | | | | | | | - Yanzhe Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (Q.M.); (J.Y.); (X.Z.); (H.L.)
| | - Xia Feng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (Q.M.); (J.Y.); (X.Z.); (H.L.)
| |
Collapse
|
5
|
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
|
6
|
Tang S, Zhao C, Zhu X. Engineering Escherichia coli-Derived Nanoparticles for Vaccine Development. Vaccines (Basel) 2024; 12:1287. [PMID: 39591189 PMCID: PMC11598912 DOI: 10.3390/vaccines12111287] [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: 10/29/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The development of effective vaccines necessitates a delicate balance between maximizing immunogenicity and minimizing safety concerns. Subunit vaccines, while generally considered safe, often fail to elicit robust and durable immune responses. Nanotechnology presents a promising approach to address this dilemma, enabling subunit antigens to mimic critical aspects of native pathogens, such as nanoscale dimensions, geometry, and highly repetitive antigen display. Various expression systems, including Escherichia coli (E. coli), yeast, baculovirus/insect cells, and Chinese hamster ovary (CHO) cells, have been explored for the production of nanoparticle vaccines. Among these, E. coli stands out due to its cost-effectiveness, scalability, rapid production cycle, and high yields. However, the E. coli manufacturing platform faces challenges related to its unfavorable redox environment for disulfide bond formation, lack of post-translational modifications, and difficulties in achieving proper protein folding. This review focuses on molecular and protein engineering strategies to enhance protein solubility in E. coli and facilitate the in vitro reassembly of virus-like particles (VLPs). We also discuss approaches for antigen display on nanocarrier surfaces and methods to stabilize these carriers. These bioengineering approaches, in combination with advanced nanocarrier design, hold significant potential for developing highly effective and affordable E. coli-derived nanovaccines, paving the way for improved protection against a wide range of infectious diseases.
Collapse
Affiliation(s)
- Shubing Tang
- Shanghai Reinovax Biologics Co., Ltd., Pudong New District, Shanghai 200135, China;
| | - Chen Zhao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Xianchao Zhu
- Shanghai Reinovax Biologics Co., Ltd., Pudong New District, Shanghai 200135, China;
| |
Collapse
|
7
|
Schulte MC, Barcellona AT, Wang X, Schrum AG, Ulery BD. M2e-Derived Peptidyl and Peptide Amphiphile Micelles as Novel Influenza Vaccines. Pharmaceuticals (Basel) 2024; 17:1503. [PMID: 39598414 PMCID: PMC11597048 DOI: 10.3390/ph17111503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Background: A significant problem with current influenza vaccines is their reliance on predictions of the most prevalent strains for the upcoming season, with inaccurate forecasts greatly reducing the overall efficacy of the immunization campaign. A universal influenza vaccine, which leverages epitopes conserved across many, if not all, strains of influenza, could reduce the need for extremely accurate forecasting. The highly conserved ectodomain of the influenza M2 protein contains a B cell epitope in the M22-16 region, making it a promising candidate as a universal influenza vaccine. Unfortunately, free peptide antigens alone are limited as vaccines due to their poor stability and weak immunogenicity in vivo. To improve the potential of peptide vaccines, immunostimulatory micellar nanoparticles can be generated from them by lipid conjugation (i.e., peptide amphiphiles-PAs). Methods: M22-16 peptides and Palm2K-M22-16-(KE)4 PAs were synthesized and characterized. BALB/c mice were subcutaneously vaccinated with these formulations, and ELISAs were conducted on serum collected from the vaccinated mice to evaluate induced antibody responses. Results: Unlike other peptide antigens previously studied, the unmodified M22-16 peptide micellized without any peptidyl or lipid modifications. M22-16 peptidyl micelles (PMs) were spherical with largely undefined secondary structure somewhat different from the cylindrical, β-sheet-containing Palm2K-M22-16-(KE)4 peptide amphiphile micelles (PAMs). Differences in physical properties were found to correlate with slightly different immune responses with PAMs eliciting higher antibody titers after the initial immunization, whereas both micelle types elicited strong IgG titers after a prime-boost regimen. Conclusions: These results suggest the viability of PAMs as single-dose vaccines, while both PMs and PAMs show potential using a multi-dose immunization approach.
Collapse
Affiliation(s)
- Megan C. Schulte
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
| | - Agustin T. Barcellona
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
| | - Xiaofei Wang
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
| | - Adam G. Schrum
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA
- Department of Surgery, University of Missouri, Columbia, MO 65211, USA
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
| | - Bret D. Ulery
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (M.C.S.); (A.T.B.); (A.G.S.)
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
- Materials Science & Engineering Institute, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
8
|
Mezgec K, Snoj J, Ulčakar L, Ljubetič A, Tušek Žnidarič M, Škarabot M, Jerala R. Coupling of Spectrin Repeat Modules for the Assembly of Nanorods and Presentation of Protein Domains. ACS NANO 2024; 18:28748-28763. [PMID: 39392430 PMCID: PMC11503911 DOI: 10.1021/acsnano.4c07701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Modular protein engineering is a powerful approach for fabricating high-molecular-weight assemblies and biomaterials with nanoscale precision. Herein, we address the challenge of designing an extended nanoscale filamentous architecture inspired by the central rod domain of human dystrophin, which protects sarcolemma during muscle contraction and consists of spectrin repeats composed of three-helical bundles. A module of three tandem spectrin repeats was used as a rigid building block self-assembling via coiled-coil (CC) dimer-forming peptides. CC peptides were precisely integrated to maintain the spectrin α-helix continuity in an appropriate frame to form extended nanorods. An orthogonal set of customizable CC heterodimers was harnessed for modular rigid domain association, which could be additionally regulated by metal ions and chelators. We achieved a robust assembly of rigid rods several micrometers in length, determined by atomic force microscopy and negative stain transmission electron microscopy. Furthermore, these rigid rods can serve as a scaffold for the decoration of diverse proteins or biologically active peptides along their length with adjustable spacing up to tens of nanometers, as confirmed by the DNA-PAINT super-resolution microscopy. This demonstrates the potential of modular bottom-up protein engineering and tunable CCs for the fabrication of functionalized protein biomaterials.
Collapse
Affiliation(s)
- Klemen Mezgec
- Department
of Synthetic Biology and Immunology, National
Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- Graduate
School of Biomedicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Jaka Snoj
- Department
of Synthetic Biology and Immunology, National
Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- Graduate
School of Biomedicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Liza Ulčakar
- Department
of Synthetic Biology and Immunology, National
Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- Graduate
School of Biomedicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Ajasja Ljubetič
- Department
of Synthetic Biology and Immunology, National
Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- EN-FIST
Centre of Excellence, SI-1000 Ljubljana, Slovenia
| | - Magda Tušek Žnidarič
- Department
of Biotechnology and Systems Biology, National
Institute of Biology, SI-1000 Ljubljana, Slovenia
| | - Miha Škarabot
- Condensed
Matter Department, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | - Roman Jerala
- Department
of Synthetic Biology and Immunology, National
Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- CTGCT, Centre
of Technology of Gene and Cell Therapy, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
9
|
Wholey WY, Meyer AR, Yoda ST, Mueller JL, Mathenge R, Chackerian B, Zikherman J, Cheng W. An Integrated Signaling Threshold Initiates IgG Response toward Virus-like Immunogens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1061-1075. [PMID: 39212443 PMCID: PMC11458362 DOI: 10.4049/jimmunol.2400101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
Class-switched neutralizing Ab (nAb) production is rapidly induced upon many viral infections. However, due to the presence of multiple components in virions, the precise biochemical and biophysical signals from viral infections that initiate nAb responses remain inadequately defined. Using a reductionist system of synthetic virus-like structures, in this study, we show that a foreign protein on a virion-sized liposome can serve as a stand-alone danger signal to initiate class-switched nAb responses without T cell help or TLR but requires CD19. Introduction of internal nucleic acids (iNAs) obviates the need for CD19, lowers the epitope density (ED) required to elicit the Ab response, and transforms these structures into highly potent immunogens that rival conventional virus-like particles in their ability to elicit strong Ag-specific IgG. As early as day 5 after immunization, structures harboring iNAs and decorated with just a few molecules of surface Ag at doses as low as 100 ng induced all IgG subclasses of Ab in mice and reproduced the IgG2a/2c restriction that is long observed in live viral infections. These findings reveal a shared mechanism for the nAb response in mice. High ED is capable but not necessary for driving Ab secretion. Instead, even a few molecules of surface Ag, when combined with nucleic acids within these structures, can trigger strong IgG production. As a result, the signaling threshold for induction of IgG in individual B cells is set by dual signals originating from both ED on the surface and the presence of iNAs within viral particulate immunogens.
Collapse
Affiliation(s)
- Wei-Yun Wholey
- Department of Pharmaceutical Sciences, 428 Church Street, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Alexander R. Meyer
- Department of Pharmaceutical Sciences, 428 Church Street, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Sekou-Tidiane Yoda
- Department of Pharmaceutical Sciences, 428 Church Street, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - James L. Mueller
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, California 94143 USA
| | - Raisa Mathenge
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, California 94143 USA
| | - Bryce Chackerian
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Julie Zikherman
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, California 94143 USA
| | - Wei Cheng
- Department of Pharmaceutical Sciences, 428 Church Street, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Biological Chemistry, 1150 W. Medical Center Dr., University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
10
|
Li H, Liu P, Dong H, Dekker A, Harmsen MM, Guo H, Wang X, Sun S. Foot-and-mouth disease virus antigenic landscape and reduced immunogenicity elucidated in atomic detail. Nat Commun 2024; 15:8774. [PMID: 39389971 PMCID: PMC11467346 DOI: 10.1038/s41467-024-53027-5] [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: 02/21/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
Unlike most other picornaviruses, foot-and-mouth disease (FMD) intact virions (146S) dissociate easily into small pentameric subunits (12S). This causes a dramatically decreased immunogenicity by a mechanism that remains elusive. Here, we present the high-resolution structures of 12S (3.2 Å) and its immune complex of a single-domain antibody (VHH) targeting the particle interior (3.2 Å), as well as two 146S-specific VHHs complexed to distinct sites on the 146S capsid surface (3.6 Å and 2.9 Å). The antigenic landscape of 146S is depicted using 13 known FMD virus-antibody complexes. Comparison of the immunogenicity of 146S and 12S in pigs, focusing on the resulting antigenic sites and incorporating structural analysis, reveals that dissociation of 146S leads to structural alteration and destruction of multiple epitopes, resulting in significant differences in antibody profiles/lineages induced by 12S and 146S. Furthermore, 146S generates higher synergistic neutralizing antibody titers compared to 12S, whereas both particles induce similar total FMD virus specific antibody titers. This study can guide the structure-based rational design of novel multivalent and broad-spectrum recombinant vaccines for protection against FMD.
Collapse
Grants
- 22JR5RA032, 23JRRA551 Natural Science Foundation of Gansu Province
- 22JR5RA032, 23JRRA551 Natural Science Foundation of Gansu Province
- 32072847,32072859, 32301127 National Natural Science Foundation of China (National Science Foundation of China)
- the National Key Research and Development Program of China (2021YFD1800303), Postdoctoral Science Foundation Funded Project (2023M733819, 23JRRA554), Lanzhou Talent Innovation and Entrepreneurship Project(2023-RC-3)
- the National Key Research and Development Program of China (2018YFA0900801), CAS (YSBR-010), the National Science Foundation Grants (12034006, 32325004 and T2394482), National Science Fund for Distinguished Young Scholar (No. 32325004), the NSFS Innovative Research Group (No. 81921005)
- the Ministry of Agriculture, Nature and Food Quality, the Netherlands (project WOT-01-002-034)
Collapse
Affiliation(s)
- Haozhou Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pan Liu
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hu Dong
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Aldo Dekker
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Michiel M Harmsen
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
| | - Xiangxi Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
| |
Collapse
|
11
|
Myers ML, Gallagher JR, Woolfork DD, Khorrami ND, Park WB, Maldonado-Puga S, Bohrnsen E, Schwarz BH, Alves DA, Bock KW, Dearborn AD, Harris AK. Structure-guided assembly of an influenza spike nanobicelle vaccine provides pan H1 intranasal protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613335. [PMID: 39372767 PMCID: PMC11451756 DOI: 10.1101/2024.09.16.613335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Development of intranasal vaccines for respiratory viruses has gained popularity. However, currently only a live-attenuated influenza vaccine is FDA-approved for intranasal administration. Here, we focused on influenza virus as it circulates seasonally, has pandemic potential, and has vaccine formulations that present hemagglutinin (HA) in different structural arrangements. These display differences have not been correlated with induction of pan-H1 antibodies or shown to provide intranasal protection. Using electron microscopy, biochemistry and animal studies, we identified HA complexes arranged as lipid discs with multiple trimeric HAs displayed along the perimeter, termed spike nanobicelles (SNB). We utilized a structure-guided approach to synthesize in vitro assembled spiked nanobicelles (IA-SNB) from a classical 1934 H1N1 influenza virus. IA-SNBs elicited pan-H1 antibodies and provided protection against antigenically divergent H1N1 viruses via intranasal immunizations. Viral glycoprotein spikes displayed as SNBs could aid in combating antigenic variation and provide innovative intranasal vaccines to aid universal influenza vaccine development.
Collapse
Affiliation(s)
- Mallory L. Myers
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - John R. Gallagher
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - De’Marcus D. Woolfork
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Noah D. Khorrami
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - William B. Park
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Samantha Maldonado-Puga
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Eric Bohrnsen
- Protein Chemistry Section, Research and Technologies Branch, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, USA 59840
| | - Benjamin H. Schwarz
- Protein Chemistry Section, Research and Technologies Branch, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, USA 59840
| | - Derron A. Alves
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, USA 20892
| | - Kevin W. Bock
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, USA 20892
| | - Altaira D. Dearborn
- Structural Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6531, Bethesda, MD, USA 20892
| | - Audray K. Harris
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| |
Collapse
|
12
|
Chiba S, Maemura T, Loeffler K, Frey SJ, Gu C, Biswas A, Hatta M, Kawaoka Y, Kane RS. Single immunization with an influenza hemagglutinin nanoparticle-based vaccine elicits durable protective immunity. Bioeng Transl Med 2024; 9:e10689. [PMID: 39553436 PMCID: PMC11561850 DOI: 10.1002/btm2.10689] [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: 03/20/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 11/19/2024] Open
Abstract
Vaccination is the most effective strategy to combat influenza. Ideally, potent and persistent vaccine effects would be induced with a single vaccine dose. Here, we designed a virus-like particle (VLP)-based vaccine presenting multiple copies of the influenza hemagglutinin (HA) from A/Puerto Rico/8/1934 (PR8HA-VLP) and examined its immunogenicity and protective efficacy in ferrets. Serum-neutralizing antibodies were effectively induced against the homologous virus at 3-week post-vaccination with a single dose of PR8HA-VLP with or without adjuvants. When the single-immunized ferrets were challenged with the homologous virus, virus replication in the nasal mucosa was significantly reduced. Long-term monitoring of serum titers revealed that after adjuvanted vaccination with PR8HA-VLP, neutralizing antibodies were retained at similar levels 20- to 183-week post-vaccination, although a 4- to 8-fold titer decline was observed from 3- to 20-week post-vaccination. Boost immunization at 183 weeks after the first immunization elicited higher neutralizing antibody titers than those at 3 weeks after the initial immunization in most of the animals. These results confirm that nanoparticle-based vaccines are a promising approach to effectively elicit durable multiyear neutralizing antibody responses against influenza viruses.
Collapse
Affiliation(s)
- Shiho Chiba
- Department of Pathobiological Sciences, School of Veterinary MedicineInfluenza Research Institute, University of Wisconsin‐MadisonMadisonWisconsinUSA
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA)The University of TokyoTokyoJapan
| | - Tadashi Maemura
- Department of Pathobiological Sciences, School of Veterinary MedicineInfluenza Research Institute, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kathryn Loeffler
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Steven J. Frey
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Chunyang Gu
- Department of Pathobiological Sciences, School of Veterinary MedicineInfluenza Research Institute, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Asim Biswas
- Department of Pathobiological Sciences, School of Veterinary MedicineInfluenza Research Institute, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Masato Hatta
- Department of Pathobiological Sciences, School of Veterinary MedicineInfluenza Research Institute, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary MedicineInfluenza Research Institute, University of Wisconsin‐MadisonMadisonWisconsinUSA
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA)The University of TokyoTokyoJapan
- Division of Virology, Department of Microbiology and ImmunologyInstitute of Medical Science, University of TokyoTokyoJapan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research InstituteTokyoJapan
| | - Ravi S. Kane
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| |
Collapse
|
13
|
Hong Y, Kwak K. Both sides now: evolutionary traits of antigens and B cells in tolerance and activation. Front Immunol 2024; 15:1456220. [PMID: 39185403 PMCID: PMC11341355 DOI: 10.3389/fimmu.2024.1456220] [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: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
B cells are the cornerstone of our body's defense system, producing precise antibodies and safeguarding immunological memory for future protection against pathogens. While we have a thorough understanding of how naïve B cells differentiate into plasma or memory B cells, the early B cell response to various antigens-whether self or foreign-remains a thrilling and evolving area of study. Advances in imaging have illuminated the molecular intricacies of B cell receptor (BCR) signaling, yet the dynamic nature of B cell activation continues to reveal new insights based on the nature of antigen exposure. This review explores the evolutionary journey of B cells as they adapt to the unique challenges presented by pathogens. We begin by examining the specific traits of antigens that influence their pathogenic potential, then shift our focus to the distinct characteristics of B cells that counteract these threats. From foundational discoveries to the latest cutting-edge research, we investigate how B cells are effectively activated and distinguish between self and non-self antigens, ensuring a balanced immune response that defends against pathogenic diseases but not self-antigens.
Collapse
Affiliation(s)
- Youngjae Hong
- Department of Microbiology and Immunology, College of Medicine, Yonsei University, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Kihyuck Kwak
- Department of Microbiology and Immunology, College of Medicine, Yonsei University, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Riedmiller I, Fougeroux C, Jensen RW, Kana IH, Sander AF, Theander TG, Lavstsen T, Turner L. Mosaic and cocktail capsid-virus-like particle vaccines for induction of antibodies against the EPCR-binding CIDRα1 domain of PfEMP1. PLoS One 2024; 19:e0302243. [PMID: 39046960 PMCID: PMC11268589 DOI: 10.1371/journal.pone.0302243] [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: 03/30/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
The sequestration of Plasmodium falciparum-infected erythrocytes to the host endothelium is central to the pathogenesis of malaria. The sequestration is mediated by the parasite´s diverse Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) variants, which bind select human receptors on the endothelium. Severe malaria is associated with PfEMP1 binding human endothelial protein C receptor (EPCR) via their CIDRα1 domains. Antibodies binding and inhibiting across the sequence diverse CIDRα1 domains are likely important in acquired immunity against severe malaria. In this study, we explored if immunization with AP205 bacteriophage capsid-virus-like particles (cVLPs) presenting a mosaic of diverse CIDRα1 protein variants would stimulate broadly reactive and inhibitory antibody responses in mice. Three different mosaic cVLP vaccines each composed of five CIDRα1 protein variants with varying degrees of sequence conservation of residues at and near the EPCR binding site, were tested. All mosaic cVLP vaccines induced functional antibodies comparable to those induced by matched cocktails of cVLPs decorated with the single CIDRα1 variant. No broadly reactive responses were observed. However, the vaccines did induce some cross-reactivity and inhibition within the CIDRα1 subclasses included in the vaccines, demonstrating potential use of the cVLP vaccine platform for the design of multivalent vaccines.
Collapse
Affiliation(s)
- Ilary Riedmiller
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Rasmus W. Jensen
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ikhlaq H. Kana
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Thor G. Theander
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Lavstsen
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Louise Turner
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Prosper P, Rodríguez Puertas R, Guérin DMA, Branda MM. Computational method for designing vaccines applied to virus-like particles (VLPs) as epitope carriers. Vaccine 2024; 42:3916-3929. [PMID: 38782665 DOI: 10.1016/j.vaccine.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 04/06/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
Nonenveloped virus-like particles (VLPs) are self-assembled oligomeric structures composed of one or more proteins that originate from diverse viruses. Because these VLPs have similar antigenicity to the parental virus, they are successfully used as vaccines against cognate virus infection. Furthermore, after foreign antigenic sequences are inserted in their protein components (chimVLPs), some VLPs are also amenable to producing vaccines against pathogens other than the virus it originates from (these VLPs are named platform or epitope carrier). Designing chimVLP vaccines is challenging because the immunogenic response must be oriented against a given antigen without altering stimulant properties inherent to the VLP. An important step in this process is choosing the location of the sequence modifications because this must be performed without compromising the assembly and stability of the original VLP. Currently, many immunogenic data and computational tools can help guide the design of chimVLPs, thus reducing experimental costs and work. In this study, we analyze the structure of a novel VLP that originate from an insect virus and describe the putative regions of its three structural proteins amenable to insertion. For this purpose, we employed molecular dynamics (MD) simulations to assess chimVLP stability by comparing mutated and wild-type (WT) VLP protein trajectories. We applied this procedure to design a chimVLP that can serve as a prophylactic vaccine against the SARS-CoV-2 virus. The methodology described in this work is generally applicable for VLP-based vaccine development.
Collapse
Affiliation(s)
- Pascalita Prosper
- Instituto de Física Aplicada - INFAP, Universidad Nacional de San Luis/CONICET, Argentina, Av. Ejército de los Andes 950, 5700 San Luis, San Luis, Argentina
| | - Rafael Rodríguez Puertas
- Universidad del País Vasco (UPV/EHU), Dept. Farmacología, Facultad de Medicina, B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain; Neurodegenerative Diseases, BioCruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Diego M A Guérin
- Universidad del País Vasco (UPV/EHU) and Instituto Biofisika (CSIC, UPV/EHU), B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain
| | - María Marta Branda
- Instituto de Física Aplicada - INFAP, Universidad Nacional de San Luis/CONICET, Argentina, Av. Ejército de los Andes 950, 5700 San Luis, San Luis, Argentina.
| |
Collapse
|
16
|
Cao LM, Yu YF, Li ZZ, Zhong NN, Wang GR, Xiao Y, Liu B, Wu QJ, Feng C, Bu LL. Adjuvants for cancer mRNA vaccines in the era of nanotechnology: strategies, applications, and future directions. J Nanobiotechnology 2024; 22:308. [PMID: 38825711 PMCID: PMC11145938 DOI: 10.1186/s12951-024-02590-6] [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: 04/13/2024] [Accepted: 05/28/2024] [Indexed: 06/04/2024] Open
Abstract
Research into mRNA vaccines is advancing rapidly, with proven efficacy against coronavirus disease 2019 and promising therapeutic potential against a variety of solid tumors. Adjuvants, critical components of mRNA vaccines, significantly enhance vaccine effectiveness and are integral to numerous mRNA vaccine formulations. However, the development and selection of adjuvant platforms are still in their nascent stages, and the mechanisms of many adjuvants remain poorly understood. Additionally, the immunostimulatory capabilities of certain novel drug delivery systems (DDS) challenge the traditional definition of adjuvants, suggesting that a revision of this concept is necessary. This review offers a comprehensive exploration of the mechanisms and applications of adjuvants and self-adjuvant DDS. It thoroughly addresses existing issues mentioned above and details three main challenges of immune-related adverse event, unclear mechanisms, and unsatisfactory outcomes in old age group in the design and practical application of cancer mRNA vaccine adjuvants. Ultimately, this review proposes three optimization strategies which consists of exploring the mechanisms of adjuvant, optimizing DDS, and improving route of administration to improve effectiveness and application of adjuvants and self-adjuvant DDS.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yi-Fu Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiu-Ji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| | - Chun Feng
- Department of Gynecology, Maternal and Child Health Hospital of Hubei Province, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
- Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
17
|
Yang RS, Traver M, Barefoot N, Stephens T, Alabanza C, Manzella-Lapeira J, Zou G, Wolff J, Li Y, Resto M, Shadrick W, Yang Y, Ivleva VB, Tsybovsky Y, Carlton K, Brzostowski J, Gall JG, Lei QP. Mosaic quadrivalent influenza vaccine single nanoparticle characterization. Sci Rep 2024; 14:4534. [PMID: 38402303 PMCID: PMC10894272 DOI: 10.1038/s41598-024-54876-2] [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: 12/29/2023] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
Recent work by our laboratory and others indicates that co-display of multiple antigens on protein-based nanoparticles may be key to induce cross-reactive antibodies that provide broad protection against disease. To reach the ultimate goal of a universal vaccine for seasonal influenza, a mosaic influenza nanoparticle vaccine (FluMos-v1) was developed for clinical trial (NCT04896086). FluMos-v1 is unique in that it is designed to co-display four recently circulating haemagglutinin (HA) strains; however, current vaccine analysis techniques are limited to nanoparticle population analysis, thus, are unable to determine the valency of an individual nanoparticle. For the first time, we demonstrate by total internal reflection fluorescence microscopy and supportive physical-chemical methods that the co-display of four antigens is indeed achieved in single nanoparticles. Additionally, we have determined percentages of multivalent (mosaic) nanoparticles with four, three, or two HA proteins. The integrated imaging and physicochemical methods we have developed for single nanoparticle multivalency will serve to further understand immunogenicity data from our current FluMos-v1 clinical trial.
Collapse
Affiliation(s)
- Rong Sylvie Yang
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Maria Traver
- Twinbrook Imaging Facility, LIG, NIAID, NIH, Gaithersburg, MD, USA
| | - Nathan Barefoot
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Tyler Stephens
- Vaccine Research Center Electron Microscopy Unit, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Casper Alabanza
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | | | - Guozhang Zou
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Jeremy Wolff
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Yile Li
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Melissa Resto
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - William Shadrick
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Yanhong Yang
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Vera B Ivleva
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Yaroslav Tsybovsky
- Vaccine Research Center Electron Microscopy Unit, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kevin Carlton
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | | | - Jason G Gall
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Q Paula Lei
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA.
| |
Collapse
|
18
|
Wholey WY, Meyer AR, Yoda ST, Chackerian B, Zikherman J, Cheng W. Minimal determinants for lifelong antiviral antibody responses in BALB/c mice from a single exposure to virus-like immunogens at low doses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.20.529089. [PMID: 36865112 PMCID: PMC9979986 DOI: 10.1101/2023.02.20.529089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
However, due to the complex compositions of natural virions, the molecular determinants of Ab durability from viral infection or inactivated viral vaccines have been incompletely understood. Here we used a reductionist system of liposome-based virus-like structures to examine the durability of Abs in primary immune responses in mice. This system allowed us to independently vary fundamental viral attributes and to do so without additional adjuvants to model natural viruses. We show that a single injection of antigens (Ags) orderly displayed on a virion-sized liposome is sufficient to induce a long-lived neutralizing Ab (nAb) response. Introduction of internal nucleic acids dramatically modulates the magnitude of long-term Ab responses without alteration of the long-term kinetic trends. These Abs are characterized by exceptionally slow off-rates of ~0.0005 s-1, which emerged as early as day 5 after injection and these off-rates are comparable to that of affinity-matured monoclonal Abs. A single injection of these structures at doses as low as 100 ng led to lifelong nAb production in BALB/c mice. Thus, a minimal virus-like immunogen can give rise to potent and long-lasting antiviral Abs in a primary response in mice without live infection. This has important implications for understanding both live viral infection and for optimized vaccine design.
Collapse
|
19
|
Wholey WY, Meyer AR, Yoda ST, Mueller JL, Mathenge R, Chackerian B, Zikherman J, Cheng W. An integrated signaling threshold initiates IgG response towards virus-like immunogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.28.577643. [PMID: 38469153 PMCID: PMC10926662 DOI: 10.1101/2024.01.28.577643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Class-switched neutralizing antibody (nAb) production is rapidly induced upon many viral infections. However, due to the presence of multiple components in typical virions, the precise biochemical and biophysical signals from viral infections that initiate nAb responses remain inadequately defined. Using a reductionist system of synthetic virus-like structures (SVLS) containing minimal, highly purified biochemical components commonly found in enveloped viruses, here we show that a foreign protein on a virion-sized liposome can serve as a stand-alone danger signal to initiate class-switched nAb responses in the absence of cognate T cell help or Toll-like receptor signaling but requires CD19, the antigen (Ag) coreceptor on B cells. Introduction of internal nucleic acids (iNAs) obviates the need for CD19, lowers the epitope density (ED) required to elicit the Ab response and transforms these structures into highly potent immunogens that rival conventional virus-like particles in their ability to elicit strong Ag-specific IgG. As early as day 5 after immunization, structures harbouring iNAs and decorated with just a few molecules of surface Ag at doses as low as 100 ng induced all IgG subclasses of Ab known in mice and reproduced the IgG2a/2c restriction that has been long observed in live viral infections. These findings reveal a shared mechanism for nAb response upon viral infection. High ED is capable but not necessary for driving Ab secretion in vivo . Instead, even a few molecules of surface Ag, when combined with nucleic acids within these structures, can trigger strong antiviral IgG production. As a result, the signaling threshold for the induction of neutralizing IgG is set by dual signals originating from both ED on the surface and the presence of iNAs within viral particulate immunogens. One-sentence summary Reconstitution of minimal viral signals necessary to initiate antiviral IgG.
Collapse
|
20
|
Newbrook K, Khan N, Fisher A, Chong K, Gubbins S, Davies WC, Sanders C, Busquets MG, Cooke L, Corla A, Ashby M, Flannery J, Batten C, Stokes JE, Sanz-Bernardo B, Carpenter S, Moffat K, Darpel KE. Specific T-cell subsets have a role in anti-viral immunity and pathogenesis but not viral dynamics or onwards vector transmission of an important livestock arbovirus. Front Immunol 2024; 15:1328820. [PMID: 38357545 PMCID: PMC10864546 DOI: 10.3389/fimmu.2024.1328820] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Bluetongue virus (BTV) is an arthropod-borne Orbivirus that is almost solely transmitted by Culicoides biting midges and causes a globally important haemorrhagic disease, bluetongue (BT), in susceptible ruminants. Infection with BTV is characterised by immunosuppression and substantial lymphopenia at peak viraemia in the host. Methods In this study, the role of cell-mediated immunity and specific T-cell subsets in BTV pathogenesis, clinical outcome, viral dynamics, immune protection, and onwards transmission to a susceptible Culicoides vector is defined in unprecedented detail for the first time, using an in vivo arboviral infection model system that closely mirrors natural infection and transmission of BTV. Individual circulating CD4+, CD8+, or WC1+ γδ T-cell subsets in sheep were depleted through the administration of specific monoclonal antibodies. Results The absence of cytotoxic CD8+ T cells was consistently associated with less severe clinical signs of BT, whilst the absence of CD4+ and WC1+ γδ T cells both resulted in an increased clinical severity. The absence of CD4+ T cells also impaired both a timely protective neutralising antibody response and the production of IgG antibodies targeting BTV non-structural protein, NS2, highlighting that the CD4+ T-cell subset is important for a timely protective immune response. T cells did not influence viral replication characteristics, including onset/dynamics of viraemia, shedding, or onwards transmission of BTV to Culicoides. We also highlight differences in T-cell dependency for the generation of immunoglobulin subclasses targeting BTV NS2 and the structural protein, VP7. Discussion This study identifies a diverse repertoire of T-cell functions during BTV infection in sheep, particularly in inducing specific anti-viral immune responses and disease manifestation, and will support more effective vaccination strategies.
Collapse
Affiliation(s)
- Kerry Newbrook
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
| | - Nakibul Khan
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- Department of Biology, University of York, York, United Kingdom
| | - Aimee Fisher
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- School of Biosciences AND School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Karen Chong
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- School of Biosciences AND School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Simon Gubbins
- Transmission Biology, The Pirbright Institute, Woking, United Kingdom
| | - William C. Davies
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| | | | | | - Lyndsay Cooke
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
| | - Amanda Corla
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | - Martin Ashby
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | - John Flannery
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | - Carrie Batten
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | | | - Beatriz Sanz-Bernardo
- Large Deoxyribonucleic Acid (DNA), Viruses, The Pirbright Institute, Woking, United Kingdom
| | | | - Katy Moffat
- Flow Cytometry, The Pirbright Institute, Woking, United Kingdom
| | - Karin E. Darpel
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- Department of Diagnostics and Development, Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
21
|
Krenger PS, Josi R, Sobczak J, Velazquez TLC, Balke I, Skinner MA, Kramer MF, Scott CJW, Hewings S, Heath MD, Zeltins A, Bachmann MF. Influence of antigen density and TLR ligands on preclinical efficacy of a VLP-based vaccine against peanut allergy. Allergy 2024; 79:184-199. [PMID: 37815010 DOI: 10.1111/all.15897] [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: 03/27/2023] [Revised: 08/17/2023] [Accepted: 09/04/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Virus-like particle (VLP) Peanut is a novel immunotherapeutic vaccine candidate for the treatment of peanut allergy. The active pharmaceutical ingredient represents cucumber mosaic VLPs (CuMVTT -VLPs) that are genetically fused with one of the major peanut allergens, Ara h 2 (CuMVTT -Ara h 2). We previously demonstrated the immunogenicity and the protective capacity of VLP Peanut-based immunization in a murine model for peanut allergy. Moreover, a Phase I clinical trial has been initiated using VLP Peanut material manufactured following a GMP-compliant manufacturing process. Key product characterization studies were undertaken here to understand the role and contribution of critical quality attributes that translate as predictive markers of immunogenicity and protective efficacy for clinical vaccine development. METHOD The role of prokaryotic RNA encapsulated within VLP Peanut on vaccine immunogenicity was assessed by producing a VLP Peanut batch with a reduced RNA content (VLP Peanut low RNA). Immunogenicity and peanut allergen challenge studies were conducted with VLP Peanut low RNA, as well as with VLP Peanut in WT and TLR 7 KO mice. Furthermore, mass spectrometry and SDS-PAGE based methods were used to determine Ara h 2 antigen density on the surface of VLP Peanut particles. This methodology was subsequently applied to investigate the relationship between Ara h 2 antigen density and immunogenicity of VLP Peanut. RESULTS A TLR 7 dependent formation of Ara h 2 specific high-avidity IgG antibodies, as well as a TLR 7 dependent change in the dominant IgG subclass, was observed following VLP Peanut vaccination, while total allergen-specific IgG remained relatively unaffected. Consistently, a missing TLR 7 signal caused only a weak decrease in allergen tolerability after vaccination. In contrast, a reduced RNA content for VLP Peanut resulted in diminished total Ara h 2 specific IgG responses, followed by a significant impairment in peanut allergen tolerability. The discrepant effect on allergen tolerance caused by an absent TLR 7 signal versus a reduced RNA content is explained by the observation that VLP Peanut-derived RNA not only stimulates TLR 7 but also TLR 3. Additionally, a strong correlation was observed between the number of Ara h 2 antigens displayed on the surface of VLP Peanut particles and the vaccine's immunogenicity and protective capacity. CONCLUSIONS Our findings demonstrate that prokaryotic RNA encapsulated within VLP Peanut, including antigen density of Ara h 2 on viral particles, are key contributors to the immunogenicity and protective capacity of the vaccine. Thus, antigenicity and RNA content are two critical quality attributes that need to be determined at the stage of manufacturing, providing robust information regarding the immunogenicity and protective capacity of VLP Peanut in the mouse which has translational relevance to the human setting.
Collapse
Affiliation(s)
- Pascal S Krenger
- Department of Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Romano Josi
- Department of Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Jan Sobczak
- Department of Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Ina Balke
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | - Matthias F Kramer
- Allergy Therapeutics (UK) Ltd, Worthing, UK
- Bencard Allergie GmbH, Munich, Germany
| | | | | | | | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Saiba AG, Zurich, Switzerland
| | - Martin F Bachmann
- Department of Rheumatology and Immunology, University Hospital of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, Centre for Cellular and Molecular Physiology (CCMP), The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Sekikawa K, Bidadi H, Maeda M, Fux L, Nataf Y, Komatsubara H, Komiyama Y, Kobayashi O. A Novel Cross-Immune Antigen Vaccine Platform for Influenza Virus Type A. Biol Pharm Bull 2024; 47:2028-2031. [PMID: 39647906 DOI: 10.1248/bpb.b24-00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
We have developed a novel cross-immune antigen vaccine platform for influenza A virus. The vaccine antigen is a fusion protein of headless hemagglutinin (HA) and matrix protein 1 (M1), which possess B-cell and T-cell epitopes, respectively, that are conserved among subgroup A viruses. The single molecule of headless HA and M1 fusion protein forms an oligomer by self-assembly of M1. T-Helper 1 (Th1) and Th2 cells were activated in an antigen-specific manner in lymphocyte cultures of antigen-immunized mice. The antigen-immunized antiserum neutralized influenza virus A subtypes H1N1 and H3N2, and intranasal administration of the antigen reduced mortality to less than 30% in a protection assay.
Collapse
Affiliation(s)
| | | | - Miki Maeda
- Research Center of Genetic Resources, National Agriculture and Food Research Organization
| | | | | | | | - Yutaka Komiyama
- Kirin Central Research Institute, Research & Development Division, Kirin Holdings Company, Limited
| | - Osamu Kobayashi
- Kirin Central Research Institute, Research & Development Division, Kirin Holdings Company, Limited
| |
Collapse
|
23
|
Gao X, Wang X, Li S, Saif Ur Rahman M, Xu S, Liu Y. Nanovaccines for Advancing Long-Lasting Immunity against Infectious Diseases. ACS NANO 2023; 17:24514-24538. [PMID: 38055649 DOI: 10.1021/acsnano.3c07741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Infectious diseases, particularly life-threatening pathogens such as small pox and influenza, have substantial implications on public health and global economies. Vaccination is a key approach to combat existing and emerging pathogens. Immunological memory is an essential characteristic used to evaluate vaccine efficacy and durability and the basis for the long-term effects of vaccines in protecting against future infections; however, optimizing the potency, improving the quality, and enhancing the durability of immune responses remains challenging and a focus for research involving investigation of nanovaccine technologies. In this review, we describe how nanovaccines can address the challenges for conventional vaccines in stimulating adaptive immune memory responses to protect against reinfection. We discuss protein and nonprotein nanoparticles as useful antigen platforms, including those with highly ordered and repetitive antigen array presentation to enhance immunogenicity through cross-linking with multiple B cell receptors, and with a focus on antigen properties. In addition, we describe how nanoadjuvants can improve immune responses by providing enhanced access to lymph nodes, lymphnode targeting, germinal center retention, and long-lasting immune response generation. Nanotechnology has the advantage to facilitate vaccine induction of long-lasting immunity against infectious diseases, now and in the future.
Collapse
Affiliation(s)
- Xinglong Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | | | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P.R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
24
|
Aves KL, Guerra PR, Fresno AH, Saraiva MMS, Cox E, Bækbo PJ, Nielsen MA, Sander AF, Olsen JE. A Virus-like Particle-Based F4 Enterotoxigenic Escherichia coli Vaccine Is Inhibited by Maternally Derived Antibodies in Piglets but Generates Robust Responses in Sows. Pathogens 2023; 12:1388. [PMID: 38133272 PMCID: PMC10745950 DOI: 10.3390/pathogens12121388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
F4-positive enterotoxigenic Escherichia coli is associated with diarrhea and poor growth outcomes in neonatal and newly weaned piglets and is thus a major economic and welfare burden in the swine industry. Vaccination of sows with F4 fimbriae protects against the neonatal disease via passive transfer of maternal immunity. However, this strategy does not protect against infection post-weaning. Consequently, prevention and treatment methods in weaner pigs heavily rely on the use of antimicrobials. Therefore, in order to reduce antimicrobial consumption, more effective prophylactic alternatives are needed. In this study, we describe the development of a capsid virus-like particle (cVLP)-based vaccine targeting the major F4 fimbriae subunit and adhesion molecule, FaeG, and evaluate its immunogenicity in mice, piglets, and sows. cVLP-display significantly increased systemic and mucosal antibody responses towards the recombinant FaeG antigen in mice models. However, in piglets, the presence of anti-F4 maternally derived antibodies severely inhibited the induction of active humoral responses towards the FaeG antigen. This inhibition could not be overcome, even with the enhanced immunogenicity achieved via cVLP display. However, in sows, intramuscular vaccination with the FaeG.cVLP vaccine was able to generate robust IgG and IgA responses that were comparable with a commercial fimbriae-based vaccine, and which were effectively transferred to piglets via colostrum intake. These results demonstrate that cVLP display has the potential to improve the systemic humoral responses elicited against low-immunogenic antigens in pigs; however, this effect is dependent on the use of antigens, which are not the targets of pre-existing maternal immunity.
Collapse
Affiliation(s)
- Kara-Lee Aves
- Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Priscila R. Guerra
- Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Ana H. Fresno
- Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Mauro M. S. Saraiva
- Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium
| | - Poul J. Bækbo
- SEGES Innovation, Danish Pig Research Centre, Agro Food Park 15, DK-8200 Aarhus, Denmark
| | - Morten A. Nielsen
- Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Adam F. Sander
- Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark
- AdaptVac, Ole Maaløes Vej 3, DK-2200 Copenhagen, Denmark
| | - John E. Olsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| |
Collapse
|
25
|
Brooks JF, Riggs J, Mueller JL, Mathenge R, Wholey WY, Meyer AR, Yoda ST, Vykunta VS, Nielsen HV, Cheng W, Zikherman J. Molecular basis for potent B cell responses to antigen displayed on particles of viral size. Nat Immunol 2023; 24:1762-1777. [PMID: 37653247 PMCID: PMC10950062 DOI: 10.1038/s41590-023-01597-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
Multivalent viral epitopes induce rapid, robust and T cell-independent humoral immune responses, but the biochemical basis for such potency remains incompletely understood. We take advantage of a set of liposomes of viral size engineered to display affinity mutants of the model antigen (Ag) hen egg lysozyme. Particulate Ag induces potent 'all-or-none' B cell responses that are density dependent but affinity independent. Unlike soluble Ag, particulate Ag induces signal amplification downstream of the B cell receptor by selectively evading LYN-dependent inhibitory pathways and maximally activates NF-κB in a manner that mimics T cell help. Such signaling induces MYC expression and enables even low doses of particulate Ag to trigger robust B cell proliferation in vivo in the absence of adjuvant. We uncover a molecular basis for highly sensitive B cell responses to viral Ag display that is independent of encapsulated nucleic acids and is not merely accounted for by avidity and B cell receptor cross-linking.
Collapse
Affiliation(s)
- Jeremy F Brooks
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Julianne Riggs
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - James L Mueller
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Raisa Mathenge
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Wei-Yun Wholey
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Alexander R Meyer
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Sekou-Tidiane Yoda
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Vivasvan S Vykunta
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Hailyn V Nielsen
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Wei Cheng
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Julie Zikherman
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
26
|
Hou Y, Chen M, Bian Y, Zheng X, Tong R, Sun X. Advanced subunit vaccine delivery technologies: From vaccine cascade obstacles to design strategies. Acta Pharm Sin B 2023; 13:3321-3338. [PMID: 37655334 PMCID: PMC10465871 DOI: 10.1016/j.apsb.2023.01.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Designing and manufacturing safe and effective vaccines is a crucial challenge for human health worldwide. Research on adjuvant-based subunit vaccines is increasingly being explored to meet clinical needs. Nevertheless, the adaptive immune responses of subunit vaccines are still unfavorable, which may partially be attributed to the immune cascade obstacles and unsatisfactory vaccine design. An extended understanding of the crosstalk between vaccine delivery strategies and immunological mechanisms could provide scientific insight to optimize antigen delivery and improve vaccination efficacy. In this review, we summarized the advanced subunit vaccine delivery technologies from the perspective of vaccine cascade obstacles after administration. The engineered subunit vaccines with lymph node and specific cell targeting ability, antigen cross-presentation, T cell activation properties, and tailorable antigen release patterns may achieve effective immune protection with high precision, efficiency, and stability. We hope this review can provide rational design principles and inspire the exploitation of future subunit vaccines.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xi Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
27
|
Zhao T, Cai Y, Jiang Y, He X, Wei Y, Yu Y, Tian X. Vaccine adjuvants: mechanisms and platforms. Signal Transduct Target Ther 2023; 8:283. [PMID: 37468460 PMCID: PMC10356842 DOI: 10.1038/s41392-023-01557-7] [Citation(s) in RCA: 283] [Impact Index Per Article: 141.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
Adjuvants are indispensable components of vaccines. Despite being widely used in vaccines, their action mechanisms are not yet clear. With a greater understanding of the mechanisms by which the innate immune response controls the antigen-specific response, the adjuvants' action mechanisms are beginning to be elucidated. Adjuvants can be categorized as immunostimulants and delivery systems. Immunostimulants are danger signal molecules that lead to the maturation and activation of antigen-presenting cells (APCs) by targeting Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs) to promote the production of antigen signals and co-stimulatory signals, which in turn enhance the adaptive immune responses. On the other hand, delivery systems are carrier materials that facilitate antigen presentation by prolonging the bioavailability of the loaded antigens, as well as targeting antigens to lymph nodes or APCs. The adjuvants' action mechanisms are systematically summarized at the beginning of this review. This is followed by an introduction of the mechanisms, properties, and progress of classical vaccine adjuvants. Furthermore, since some of the adjuvants under investigation exhibit greater immune activation potency than classical adjuvants, which could compensate for the deficiencies of classical adjuvants, a summary of the adjuvant platforms under investigation is subsequently presented. Notably, we highlight the different action mechanisms and immunological properties of these adjuvant platforms, which will provide a wide range of options for the rational design of different vaccines. On this basis, this review points out the development prospects of vaccine adjuvants and the problems that should be paid attention to in the future.
Collapse
Affiliation(s)
- Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yulong Cai
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yujie Jiang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yifan Yu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
28
|
Xue VW, Wong SCC, Li B, Cho WCS. The discovery and development of mRNA vaccines for the prevention of SARS-CoV-2 infection. Expert Opin Drug Discov 2023; 18:769-780. [PMID: 37237360 DOI: 10.1080/17460441.2023.2218083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
INTRODUCTION COVID-19 pandemic is one of the most serious public health events of this century. There have been more than 670 million confirmed cases and more than 6 million deaths worldwide. From the emergence of the Alpha variant to the later rampant Omicron variant, the high transmissibility and pathogenicity of SARS-CoV-2 accelerate the research and development of effective vaccines. Against this background, mRNA vaccines stepped onto the historical stage and became an important tool for COVID-19 prevention. AREAS COVERED This article introduces the characteristics of different mRNA vaccines in the prevention of COVID-19, including antigen selection, therapeutic mRNA design and modification, and different delivery systems of mRNA molecules. It also summarizes and discusses the mechanisms, safety, effectiveness, side effects, and limitations of current COVID-19 mRNA vaccines. EXPERT OPINION Therapeutic mRNA molecules have plenty of advantages, including flexible design, rapid production, sufficient immune activation, safety without the risk of genome insertion in the host cells, and no viral vectors or particles involved, making them an important tool to fight diseases in the future. However, the application of COVID-19 mRNA vaccines also faces many challenges, such as storage and transportation, mass production, and nonspecific immunity.
Collapse
Affiliation(s)
- Vivian Weiwen Xue
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, China
| | - Sze Chuen Cesar Wong
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Bo Li
- School of Textile Science and Engineering, Xi'an Polytechnic University, Xi'an, China
| | | |
Collapse
|
29
|
Wang D, Yuan Y, Liu B, Epstein ND, Yang Y. Protein-based nano-vaccines against SARS-CoV-2: Current design strategies and advances of candidate vaccines. Int J Biol Macromol 2023; 236:123979. [PMID: 36907305 PMCID: PMC9998285 DOI: 10.1016/j.ijbiomac.2023.123979] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023]
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shaken the global health system. Various nanotechnology-based strategies for vaccine development have played pivotal roles in fighting against SARS-CoV-2. Among them, the safe and effective protein-based nanoparticle (NP) platforms display a highly repetitive array of foreign antigens on their surface, which is urgent for improving the immunogenicity of vaccines. These platforms greatly improved antigen uptake by antigen presenting cells (APCs), lymph node trafficking, and B cell activation, due to the optimal size, multivalence, and versatility of NPs. In this review, we summarize the advances of protein-based NP platforms, strategies of antigen attachment, and the current progress of clinical and preclinical trials in the development of SARS-CoV-2 vaccines based on protein-based NP platforms. Importantly, the lessons learnt and design approaches developed for these NP platforms against SARS-CoV-2 also provide insights into the development of protein-based NP strategies for preventing other epidemic diseases.
Collapse
Affiliation(s)
- Dongliang Wang
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; College of Biology, Hunan University, Changsha 410082, China
| | - Youqing Yuan
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha 410082, China
| | - Neal D Epstein
- Cell and Developmental Biology Center, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Yi Yang
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
30
|
Sylvie Yang R, Yang Y, Gowetski DB, Tsybovsky Y, Gall JGD, Paula Lei Q. Characterization of Flu MOSAIC nanoparticle vaccine candidate using high performance size-exclusion chromatography to support vaccine process development. Vaccine 2023; 41:2534-2545. [PMID: 36906406 DOI: 10.1016/j.vaccine.2023.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023]
Abstract
This report describes an application of analytical high performance size exclusion chromatography with UV and Fluorescent detection (HPSEC-UV/FLR) method that enabled a bridging from research vaccine candidate discovery (His-tagged model) to clinical product development (Non-His-tagged molecules). HPSEC measurement can accurately determine the total trimer-to-pentamer molar ratio by either titration evaluation during the nanoparticle being assembled or dissociation during a well-formed nanoparticle being dis-assembled. Through experimental design with small sample consumptions, HPSEC can provide a quick determination on the nanoparticle assembling efficiency which can therefore guide the buffer optimization for an assembly, from His-tagged model nanoparticle, to non-His-tagged clinical development product. HPSEC has also discovered a difference in assembling efficiencies for various strains of HAx-dn5B with Pentamer-dn5A components, and different efficiencies for monovalent assembly vs. multivalent assembly. The present study demonstrates HPSEC as a pivotal tool to support the Flu Mosaic nanoparticle vaccine development from research to clinical production.
Collapse
Affiliation(s)
- Rong Sylvie Yang
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Gaithersburg, MD, USA
| | - Yanhong Yang
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Gaithersburg, MD, USA
| | - Daniel B Gowetski
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Gaithersburg, MD, USA
| | - Yaroslav Tsybovsky
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Gaithersburg, MD, USA
| | - Jason G D Gall
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Gaithersburg, MD, USA
| | - Q Paula Lei
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Gaithersburg, MD, USA.
| |
Collapse
|
31
|
Brooks JF, Riggs J, Mueller JL, Mathenge R, Wholey WY, Yoda ST, Vykunta VS, Cheng W, Zikherman J. Molecular basis for potent B cell responses to antigen displayed on particles of viral size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528761. [PMID: 36824873 PMCID: PMC9949087 DOI: 10.1101/2023.02.15.528761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Although it has long been appreciated that multivalent antigens - and particularly viral epitope display - produce extremely rapid, robust, and T-independent humoral immune responses, the biochemical basis for such potency has been incompletely understood. Here we take advantage of a set of neutral liposomes of viral size that are engineered to display affinity mutants of the model antigen (Ag) hen egg lysozyme at precisely varied density. We show that particulate Ag display by liposomes induces highly potent B cell responses that are dose-and density-dependent but affinity-independent. Titrating dose of particulate, but not soluble, Ag reveals bimodal Erk phosphorylation and cytosolic calcium increases. Particulate Ag induces signal amplification downstream of the B cell receptor (BCR) by selectively evading LYN-dependent inhibitory pathways, but in vitro potency is independent of CD19. Importantly, Ag display on viral-sized particles signals independently of MYD88 and IRAK1/4, but activates NF- κ B robustly in a manner that mimics T cell help. Together, such biased signaling by particulate Ag promotes MYC expression and reduces the threshold required for B cell proliferation relative to soluble Ag. These findings uncover a molecular basis for highly sensitive B cell response to viral Ag display and remarkable potency of virus-like particle vaccines that is not merely accounted for by avidity and BCR cross-linking, and is independent of the contribution of B cell nucleic acid-sensing machinery.
Collapse
|
32
|
Characterization of Systemic and Mucosal Humoral Immune Responses to an Adjuvanted Intranasal SARS-CoV-2 Protein Subunit Vaccine Candidate in Mice. Vaccines (Basel) 2022; 11:vaccines11010030. [PMID: 36679875 PMCID: PMC9865305 DOI: 10.3390/vaccines11010030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Continuous viral evolution of SARS-CoV-2 has resulted in variants capable of immune evasion, vaccine breakthrough infections and increased transmissibility. New vaccines that invoke mucosal immunity may provide a solution to reducing virus transmission. Here, we evaluated the immunogenicity of intranasally administered subunit protein vaccines composed of a stabilized SARS-CoV-2 spike trimer or the receptor binding domain (RBD) adjuvanted with either cholera toxin (CT) or an archaeal lipid mucosal adjuvant (AMVAD). We show robust induction of immunoglobulin (Ig) G and IgA responses in plasma, nasal wash and bronchoalveolar lavage in mice only when adjuvant is used in the vaccine formulation. While the AMVAD adjuvant was more effective at inducing systemic antibodies against the RBD antigen than CT, CT was generally more effective at inducing overall higher IgA and IgG titers against the spike antigen in both systemic and mucosal compartments. Furthermore, vaccination with adjuvanted spike led to superior mucosal IgA responses than with the RBD antigen and produced broadly targeting neutralizing plasma antibodies against ancestral, Delta and Omicron variants in vitro; whereas adjuvanted RBD elicited a narrower antibody response with neutralizing activity only against ancestral and Delta variants. Our study demonstrates that intranasal administration of an adjuvanted protein subunit vaccine in immunologically naïve mice induced both systemic and mucosal neutralizing antibody responses that were most effective at neutralizing SARS-CoV-2 variants when the trimeric spike was used as an antigen compared to RBD.
Collapse
|
33
|
Halfmann PJ, Frey SJ, Loeffler K, Kuroda M, Maemura T, Armbrust T, Yang JE, Hou YJ, Baric R, Wright ER, Kawaoka Y, Kane RS. Multivalent S2-based vaccines provide broad protection against SARS-CoV-2 variants of concern and pangolin coronaviruses. EBioMedicine 2022; 86:104341. [PMID: 36375316 PMCID: PMC9651965 DOI: 10.1016/j.ebiom.2022.104341] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The COVID-19 pandemic continues to cause morbidity and mortality worldwide. Most approved COVID-19 vaccines generate a neutralizing antibody response that primarily targets the highly variable receptor-binding domain (RBD) of the SARS-CoV-2 spike (S) protein. SARS-CoV-2 "variants of concern" have acquired mutations in this domain allowing them to evade vaccine-induced humoral immunity. Recent approaches to improve the breadth of protection beyond SARS-CoV-2 have required the use of mixtures of RBD antigens from different sarbecoviruses. It may therefore be beneficial to develop a vaccine in which the protective immune response targets a more conserved region of the S protein. METHODS Here we have developed a vaccine based on the conserved S2 subunit of the S protein and optimized the adjuvant and immunization regimen in Syrian hamsters and BALB/c mice. We have characterized the efficacy of the vaccine against SARS-CoV-2 variants and other coronaviruses. FINDINGS Immunization with S2-based constructs elicited a broadly cross-reactive IgG antibody response that recognized the spike proteins of not only SARS-CoV-2 variants, but also SARS-CoV-1, and the four endemic human coronaviruses. Importantly, immunization reduced virus titers in respiratory tissues in vaccinated animals challenged with SARS-CoV-2 variants B.1.351 (beta), B.1.617.2 (delta), and BA.1 (omicron) as well as a pangolin coronavirus. INTERPRETATION These results suggest that S2-based constructs can elicit a broadly cross-reactive antibody response resulting in limited virus replication, thus providing a framework for designing vaccines that elicit broad protection against coronaviruses. FUNDING NIH, Japan Agency for Medical Research and Development, Garry Betty/ V Foundation Chair Fund, and NSF.
Collapse
Affiliation(s)
- Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Steven J Frey
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Kathryn Loeffler
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Makoto Kuroda
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Tadashi Maemura
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Tammy Armbrust
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Jie E Yang
- Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA; Cryo-EM Research Center, Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA; Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA
| | - Yixuan J Hou
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ralph Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Elizabeth R Wright
- Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA; Cryo-EM Research Center, Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA; Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA; Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan.
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
34
|
Nelson SA, Richards KA, Glover MA, Chaves FA, Crank MC, Graham BS, Kanekiyo M, Sant AJ. CD4 T cell epitope abundance in ferritin core potentiates responses to hemagglutinin nanoparticle vaccines. NPJ Vaccines 2022; 7:124. [PMID: 36289232 PMCID: PMC9605951 DOI: 10.1038/s41541-022-00547-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/06/2022] [Indexed: 11/20/2022] Open
Abstract
Nanoparticle vaccines based on H. pylori ferritin are increasingly used as a vaccine platform for many pathogens, including RSV, influenza, and SARS-CoV-2. They have been found to elicit enhanced, long-lived B cell responses. The basis for improved efficacy of ferritin nanoparticle vaccines remains unresolved, including whether recruitment of CD4 T cells specific for the ferritin component of these vaccines contributes to cognate help in the B cell response. Using influenza HA-ferritin nanoparticles as a prototype, we have performed an unbiased assessment of the CD4 T cell epitope composition of the ferritin particles relative to that contributed by influenza HA using mouse models that express distinct constellations of MHC class II molecules. The role that these CD4 T cells play in the B cell responses was assessed by quantifying follicular helper cells (TFH), germinal center (GC) B cells, and antibody secreting cells. When mice were immunized with equimolar quantities of soluble HA-trimers and HA-Fe nanoparticles, HA-nanoparticle immunized mice had an increased overall abundance of TFH that were found to be largely ferritin-specific. HA-nanoparticle immunized mice had an increased abundance of HA-specific isotype-switched GC B cells and HA-specific antibody secreting cells (ASCs) relative to mice immunized with soluble HA-trimers. Further, there was a strong, positive correlation between CD4 TFH abundance and GC B cell abundance. Thus, availability of helper CD4 T cell epitopes may be a key additional mechanism that underlies the enhanced immunogenicity of ferritin-based HA-Fe-nanoparticle vaccines.
Collapse
Affiliation(s)
- Sean A Nelson
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Maryah A Glover
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Francisco A Chaves
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Michelle C Crank
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Institute for Asthma & Allergy, Chevy Chase, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
35
|
Chen WH, Strych U, Bottazzi ME, Lin YP. Past, present, and future of Lyme disease vaccines: antigen engineering approaches and mechanistic insights. Expert Rev Vaccines 2022; 21:1405-1417. [PMID: 35836340 PMCID: PMC9529901 DOI: 10.1080/14760584.2022.2102484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/13/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Transmitted by ticks, Lyme disease is the most common vector-borne disease in the Northern hemisphere. Despite the geographical expansion of human Lyme disease cases, no effective preventive strategies are currently available. Developing an efficacious and safe vaccine is therefore urgently needed. Efforts have previously been taken to identify vaccine targets in the causative pathogen (Borrelia burgdorferi sensu lato) and arthropod vector (Ixodes spp.). However, progress was impeded due to a lack of consumer confidence caused by the myth of undesired off-target responses, low immune responses, a limited breadth of immune reactivity, as well as by the complexities of the vaccine process development. AREA COVERED In this review, we summarize the antigen engineering approaches that have been applied to overcome those challenges and the underlying mechanisms that can be exploited to improve both safety and efficacy of future Lyme disease vaccines. EXPERT OPINION Over the past two decades, several new genetically redesigned Lyme disease vaccine candidates have shown success in both preclinical and clinical settings and built a solid foundation for further development. These studies have greatly informed the protective mechanisms of reducing Lyme disease burdens and ending the endemic of this disease.
Collapse
Affiliation(s)
- Wen-Hsiang Chen
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Ulrich Strych
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
- Department of Biology, Baylor University, Waco, TX, United States
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA
| |
Collapse
|
36
|
Expression and Evaluation of a Novel PPRV Nanoparticle Antigen Based on Ferritin Self-Assembling Technology. Pharmaceutics 2022; 14:pharmaceutics14091902. [PMID: 36145650 PMCID: PMC9500948 DOI: 10.3390/pharmaceutics14091902] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Peste des Petits Ruminants (PPR) is a highly pathogenic disease that is classified as a World Organization for Animal Health (OIE)-listed disease. PPRV mainly infects small ruminants such as goats and sheep. In view of the global and high pathogenicity of PPRV, in this study, we proposed a novel nanoparticle vaccine strategy based on ferritin (Fe) self-assembly technology. Using Helicobacter pylori (H. pylori) ferritin as an antigen delivery vector, a PPRV hemagglutinin (H) protein was fused with ferritin and then expressed and purified in both Escherichia coli (E. coli) and silkworm baculovirus expression systems. Subsequently, the nanoparticle antigens’ expression level, immunogenicity and protective immune response were evaluated. Our results showed that the PPRV hemagglutinin–ferritin (H-Fe) protein was self-assembled in silkworms, while it was difficult to observe the correctly folded nanoparticle in E. coli. Meanwhile, the expression level of the H-Fe protein was higher than that of the H protein alone. Furthermore, the immunogenicity and protective immune response of H-Fe nanoparticle antigens expressed by silkworms were improved compared with the H antigen alone. Particularly, the protective immune response of H-Fe antigens expressed in E. coli did not change, as opposed to the H antigen, which was probably due to the incomplete nanoparticle structure in E. coli. This study indicated that the use of ferritin nanoparticles as antigen delivery carriers could increase the expression of antigen proteins and improve the immunogenicity and immune effect of antigens.
Collapse
|
37
|
Nazarian S, Olad G, Abdolhamidi R, Motamedi MJ, Kazemi R, Kordbacheh E, Felagari A, Olad H, Ahmadi A, Bahiraee A, Farahani P, Haghighi L, Hassani F, Hajhassan V, Nadi M, Sheikhi A, Salimian J, Amani J. Preclinical study of formulated recombinant nucleocapsid protein, the receptor binding domain of the spike protein, and truncated spike (S1) protein as vaccine candidates against COVID-19 in animal models. Mol Immunol 2022; 149:107-118. [PMID: 35802999 PMCID: PMC9222294 DOI: 10.1016/j.molimm.2022.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/13/2022] [Accepted: 06/19/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND In this pre-clinical study, we designed a candidate vaccine based on severe acute respiratory syndrome-related -coronavirus 2 (SARS-CoV-2) antigens and evaluated its safety and immunogenicity. METHODS SARS-CoV-2 recombinant protein antigens, including truncated spike protein (SS1, lacking the N-terminal domain of S1), receptor-binding domain (RBD), and nucleoprotein (N) were used. Immunization program was performed via injection of RBD, SS1 +RBD, and SS1 +N along with different adjuvants, Alum, AS03, and Montanide at doses of 0, 40, 80, and 120 μg at three-time points in mice, rabbits, and primates. The humoral and cellular immunity were analyzed by ELISA, VNT, splenocyte cytokine assay, and flow cytometry. RESULTS The candidate vaccine produced strong IgG antibody titers at doses of 80 and 120 μg on days 35 and 42. Even though AS03 and Montanide produced high-titer antibodies compared to Alum adjuvant, these sera did not neutralize the virus. Strong virus neutralization was recorded during immunization with SS1 +RBD and RBD with Alum. AS03 and Montanide showed a strong humoral and cellular immunity; however, Alum showed mild to moderate cellular responses. Ultimately, no cytotoxicity and pathologic change were observed. CONCLUSION These findings strongly suggest that RBD with Alum adjuvant is highly immunogenic as a potential vaccine.
Collapse
Affiliation(s)
- Shahram Nazarian
- Department of Biology, Faculty of Science, Imam Hossein University, Tehran, Iran
| | - Gholamreza Olad
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raziyeh Abdolhamidi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Emad Kordbacheh
- Department of Biology, Faculty of Science, Imam Hossein University, Tehran, Iran
| | - Alireza Felagari
- Department of Biology, Faculty of Science, Imam Hossein University, Tehran, Iran
| | - Hanieh Olad
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Bahiraee
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Parisa Farahani
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Leila Haghighi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Faezeh Hassani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Mona Nadi
- Molecular Biology Department, Green Gene Company, Tehran, Iran
| | - Abdolkarim Sheikhi
- Department of Immunology and Microbiology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Jafar Salimian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran; Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
38
|
Schneider CG, Fey J, Zou X, Gerbasi V, Savransky T, Batt C, Bergmann-Leitner E, Angov E. Norovirus-VLPs expressing pre-erythrocytic malaria antigens induce functional immunity against sporozoite infection. Vaccine 2022; 40:4270-4280. [PMID: 35697572 DOI: 10.1016/j.vaccine.2022.05.076] [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: 03/22/2022] [Revised: 04/27/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
Despite the development of prophylactic anti-malarial drugs and practices to prevent infection, malaria remains a health concern. Preclinical testing of novel malaria vaccine strategies achieved through rational antigen selection and novel particle-based delivery platforms is yielding encouraging results. One such platform, self-assembling virus-like particles (VLP) is safer than attenuated live viruses, and has been approved as a vaccination tool by the FDA. We explore the use of Norovirus sub-viral particles lacking the natural shell (S) domain forming the interior shell but that retain the protruding (P) structures of the native virus as a vaccine vector. Epitope selection and their surface display has the potential to focus antigen specific immune responses to crucial epitopes. Recombinant P-particles displaying epitopes from two malaria antigens, Plasmodium falciparum (Pf) CelTOS and Plasmodium falciparum (Pf) CSP, were evaluated for immunogenicity and their ability to confer protection in a murine challenge model. Immune responses induced in mice resulted either in sterile protection (displaying PfCelTOS epitopes) or in antibodies with functional activity against sporozoites (displaying PfCSP epitopes) in an in vitro liver-stage development assay (ILSDA). These results are encouraging and support further evaluation of this platform as a vaccine delivery system.
Collapse
Affiliation(s)
- Cosette G Schneider
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831, USA.
| | - Julien Fey
- Agave BioSystems, Ithaca, NY 14850, USA.
| | - Xiaoyan Zou
- Naval Medical Research Center, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Vince Gerbasi
- Naval Medical Research Center, Silver Spring, MD 20910, USA.
| | - Tatyana Savransky
- Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; General Dynamics Information Technology, Falls Church, VA 22042, USA.
| | - Carl Batt
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA.
| | - Elke Bergmann-Leitner
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| | - Evelina Angov
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
39
|
Li Y, Bao Q, Yang S, Yang M, Mao C. Bionanoparticles in cancer imaging, diagnosis, and treatment. VIEW 2022. [DOI: 10.1002/viw.20200027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yan Li
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Qing Bao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
| | - Shuxu Yang
- Department of Neurosurgery Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Mingying Yang
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Chuanbin Mao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
- Department of Chemistry and Biochemistry Stephenson Life Science Research Center University of Oklahoma Norman Oklahoma USA
| |
Collapse
|
40
|
Rashidijahanabad Z, Kelly M, Kamruzzaman M, Qadri F, Bhuiyan TR, McFall-Boegeman H, Wu D, Piszczek G, Xu P, Ryan ET, Huang X. Virus-like Particle Display of Vibrio choleraeO-Specific Polysaccharide as a Potential Vaccine against Cholera. ACS Infect Dis 2022; 8:574-583. [PMID: 35170309 PMCID: PMC9119010 DOI: 10.1021/acsinfecdis.1c00585] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vibrio cholerae, a noninvasive mucosal pathogen, is endemic in more than 50 countries. Oral cholera vaccines, based on killed whole-cell strains of Vibrio cholerae, can provide significant protection in adults and children for 2-5 years. However, they have relatively limited direct protection in young children. To overcome current challenges, in this study, a potential conjugate vaccine was developed by linking O-specific polysaccharide (OSP) antigen purified from V. cholerae O1 El Tor Inaba strain PIC018 with Qβ virus-like particles efficiently via squarate chemistry. The Qβ-OSP conjugate was characterized with mass photometry (MP) on the whole particle level. Pertinent immunologic display of OSP was confirmed by immunoreactivity of the conjugate with convalescent phase samples from humans with cholera. Mouse immunization with the Qβ-OSP conjugate showed that the construct generated prominent and long-lasting IgG antibody responses against OSP, and the resulting antibodies could recognize the native lipopolysaccharide from Vibrio cholerae O1 Inaba. This was the first time that Qβ was conjugated with a bacterial polysaccharide for vaccine development, broadening the scope of this powerful carrier.
Collapse
Affiliation(s)
- Zahra Rashidijahanabad
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Meagan Kelly
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Mohammad Kamruzzaman
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Firdausi Qadri
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Taufiqur R Bhuiyan
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Hunter McFall-Boegeman
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Di Wu
- Biophysics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Grzegorz Piszczek
- Biophysics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peng Xu
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Edward T Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States.,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
41
|
Nanoparticle and virus-like particle vaccine approaches against SARS-CoV-2. J Microbiol 2022; 60:335-346. [PMID: 35089583 PMCID: PMC8795728 DOI: 10.1007/s12275-022-1608-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
The global spread of coronavirus disease 2019 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has provoked an urgent need for prophylactic measures. Several innovative vaccine platforms have been introduced and billions of vaccine doses have been administered worldwide. To enable the creation of safer and more effective vaccines, additional platforms are under development. These include the use of nanoparticle (NP) and virus-like particle (VLP) technology. NP vaccines utilize self-assembling scaffold structures designed to load the entire spike protein or receptor-binding domain of SARS-CoV-2 in a trimeric configuration. In contrast, VLP vaccines are genetically modified recombinant viruses that are considered safe, as they are generally replication-defective. Furthermore, VLPs have indigenous immunogenic potential due to their microbial origin. Importantly, NP and VLP vaccines have shown stronger immunogenicity with greater protection by mimicking the physicochemical characteristics of SARS-CoV-2. The study of NP- and VLP-based coronavirus vaccines will help ensure the development of rapid-response technology against SARS-CoV-2 variants and future coronavirus pandemics.
Collapse
|
42
|
Chevillard C, Amen A, Besson S, Hannani D, Bally I, Dettling V, Gout E, Moreau CJ, Buisson M, Gallet S, Fenel D, Vassal-Stermann E, Schoehn G, Poignard P, Dagher MC, Fender P. Elicitation of potent SARS-CoV-2 neutralizing antibody responses through immunization with a versatile adenovirus-inspired multimerization platform. Mol Ther 2022; 30:1913-1925. [PMID: 35151843 PMCID: PMC8828441 DOI: 10.1016/j.ymthe.2022.02.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/19/2022] [Accepted: 02/07/2022] [Indexed: 11/24/2022] Open
Abstract
Virus-like particles (VLPs) are highly suited platforms for protein-based vaccines. In the present work, we adapted a previously designed non-infectious adenovirus-inspired 60-mer dodecahedric VLP (ADDomer) to display a multimeric array of large antigens through a SpyTag/SpyCatcher system. To validate the platform as a potential COVID-19 vaccine approach, we decorated the newly designed VLP with the glycosylated receptor binding domain (RBD) of SARS-CoV-2. Cryoelectron microscopy structure revealed that up to 60 copies of this antigenic domain could be bound on a single ADDomer particle, with the symmetrical arrangements of a dodecahedron. Mouse immunization with the RBD decorated VLPs already showed a significant specific humoral response following prime vaccination, greatly reinforced by a single boost. Neutralization assays with SARS-CoV-2 spike pseudo-typed virus demonstrated the elicitation of strong neutralization titers, superior to those of COVID-19 convalescent patients. Notably, the presence of pre-existing immunity against the adenoviral-derived particles did not hamper the immune response against the antigen displayed on its surface. This plug and play vaccine platform represents a promising new highly versatile tool to combat emergent pathogens.
Collapse
Affiliation(s)
- Christopher Chevillard
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Axelle Amen
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France; CHU Grenoble Alpes, 38000 Grenoble, France
| | - Solène Besson
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Dalil Hannani
- University Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000 Grenoble, France
| | - Isabelle Bally
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Valentin Dettling
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Evelyne Gout
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Christophe J Moreau
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Marlyse Buisson
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Salomé Gallet
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Daphna Fenel
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Emilie Vassal-Stermann
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Guy Schoehn
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Pascal Poignard
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France; CHU Grenoble Alpes, 38000 Grenoble, France; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Marie-Claire Dagher
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Pascal Fender
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France.
| |
Collapse
|
43
|
Firdaus FZ, Skwarczynski M, Toth I. Developments in Vaccine Adjuvants. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2412:145-178. [PMID: 34918245 DOI: 10.1007/978-1-0716-1892-9_8] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vaccines, including subunit, recombinant, and conjugate vaccines, require the use of an immunostimulator/adjuvant for maximum efficacy. Adjuvants not only enhance the strength and longevity of immune responses but may also influence the type of response. In this chapter, we review the adjuvants that are available for use in human vaccines, such as alum, MF59, AS03, and AS01. We extensively discuss their composition, characteristics, mechanism of action, and effects on the immune system. Additionally, we summarize recent trends in adjuvant discovery, providing a brief overview of saponins, TLRs agonists, polysaccharides, nanoparticles, cytokines, and mucosal adjuvants.
Collapse
Affiliation(s)
- Farrhana Ziana Firdaus
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,Institute of Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
44
|
Islam KU, A-Elgadir TME, Afaq S, Ahmad T, Iqbal J. Molecular and Clinical Aspects of COVID-19 Vaccines and Other Therapeutic Interventions Apropos Emerging Variants of Concern. Front Pharmacol 2021; 12:778219. [PMID: 35002711 PMCID: PMC8734653 DOI: 10.3389/fphar.2021.778219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has overwhelmed the healthcare and economy of the world, with emerging new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posing an everlasting threat to humanity. While most COVID-19 vaccines provide adequate protective immunological response against the original SARS-CoV-2 variant, there is a pressing need to understand their biological and clinical responses. Recent evidence suggests that some of the new variants of SARS-CoV-2 evade the protection conferred by the existing vaccines, which may impede the ongoing efforts to expedite the vaccination programs worldwide. These concerns have also highlighted the importance of a pan-COVID-19 vaccine, which is currently in the making. Thus, it is imperative to have a better molecular and clinical understanding of the various COVID-19 vaccines and their immunological trajectory against any emerging variant of concerns (VOCs) in particular to break this vicious cycle. Furthermore, other treatment regimens based on cellular therapies and monoclonal antibodies should be explored systematically as an alternative and readily available option considering the possibility of the emergence of more virulent SARS-CoV-2 mutants. In this review, we shed light on the various molecular mechanisms and clinical responses of COVID-19 vaccines. Importantly, we review the recent findings of their long-term immune protection and efficacy against emerging VOCs. Considering that other targeted and effective treatments will complement vaccine therapy, we provide a comprehensive understanding of the role of cell-based therapies, monoclonal antibodies, and immunomodulatory agents as alternative and readily available treatment modalities against any emerging SARS-CoV-2 variant.
Collapse
Affiliation(s)
- Khursheed Ul Islam
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | | | - Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Jawed Iqbal
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
45
|
Minimal Dosage of Porcine Circovirus Type 2d Based Virus-like Particles to Induce Stable Protective Immunity against Infection. Pathogens 2021; 10:pathogens10121644. [PMID: 34959599 PMCID: PMC8706284 DOI: 10.3390/pathogens10121644] [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: 11/19/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 11/16/2022] Open
Abstract
In recent years, porcine circovirus type 2d (PCV2d) has achieved a dominant position worldwide. Various PCV2d capsid-based vaccines have been used to alleviate concerns regarding the emergence of the variant. This study aimed to determine the dosage of recombinant PCV2d capsid protein to induce protective efficacy against experimental challenge with a virulent PCV2d strain. Conventional 3-week-old pigs were intramuscularly inoculated with different doses of the protein (60, 20, 10 and 2 µg). Four weeks after vaccination, all pigs were challenged with pathogenic PCV2d (SNU140003), which was isolated from a farm severely experiencing PCV2-associated disease in Korea. Vaccination with greater than 10 µg of the capsid protein caused a significant (p < 0.05) reduction in PCV2d viremia, lymphoid lesions and lymphoid PCV2 antigen levels in vaccinated challenged pigs compared to unvaccinated challenged pigs. The vaccination also resulted in significantly higher (p < 0.05) titers of neutralizing antibodies against PCV2d. However, the pigs vaccinated with 2 µg had significantly lower neutralizing antibody titers than the other vaccinated groups. They showed a similar level of challenged PCV2d in serum and lymphoid lesion score compared to unvaccinated challenged pigs. The difference in efficacy among the vaccinated groups indicates that there may be a baseline dosage to induce sufficient neutralizing antibodies to prevent viral replication in pigs. In conclusion, at least 10 µg dosage of capsid protein is essential for stable protective efficacy against PCV2d in a pig model.
Collapse
|
46
|
Caradonna TM, Schmidt AG. Protein engineering strategies for rational immunogen design. NPJ Vaccines 2021; 6:154. [PMID: 34921149 PMCID: PMC8683408 DOI: 10.1038/s41541-021-00417-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Antibody immunodominance refers to the preferential and asymmetric elicitation of antibodies against specific epitopes on a complex protein antigen. Traditional vaccination approaches for rapidly evolving pathogens have had limited success in part because of this phenomenon, as elicited antibodies preferentially target highly variable regions of antigens, and thus do not confer long lasting protection. While antibodies targeting functionally conserved epitopes have the potential to be broadly protective, they often make up a minority of the overall repertoire. Here, we discuss recent protein engineering strategies used to favorably alter patterns of immunodominance, and selectively focus antibody responses toward broadly protective epitopes in the pursuit of next-generation vaccines for rapidly evolving pathogens.
Collapse
Affiliation(s)
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
47
|
Fayd'herbe De Maudave A, Leconet W, Toupet K, Constantinides M, Bossis G, de Toledo M, Vialaret J, Hirtz C, Lopez-Noriega A, Jorgensen C, Noël D, Louis-Plence P, Grizot S, Villalba M. Intra-articular delivery of full-length antibodies through the use of an in situ forming depot. J Control Release 2021; 341:578-590. [PMID: 34915070 DOI: 10.1016/j.jconrel.2021.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022]
Abstract
Monoclonal antibodies (mAbs) are large size molecules that have demonstrated high therapeutic potential for the treatment of cancer or autoimmune diseases. Despite some excellent results, their intravenous administration results in high plasma concentration. This triggers off-target effects and sometimes poor targeted tissue distribution. To circumvent this issue, we investigated a local controlled-delivery approach using an in situ forming depot technology. Two clinically relevant mAbs, rituximab (RTX) and daratumumab (DARA), were formulated using an injectable technology based on biodegradable PEG-PLA copolymers. The stability and controlled release features of the formulations were investigated. HPLC and mass spectrometry revealed the preservation of the protein structure. In vitro binding of formulated antibodies to their target antigens and to their cellular FcγRIIIa natural killer cell receptor was fully maintained. Furthermore, encapsulated RTX was as efficient as classical intravenous RTX treatment to inhibit the in vivo tumor growth of malignant human B cells in immunodeficient NSG mice. Finally, the intra-articular administration of the formulated mAbs yielded a sustained local release associated with a lower plasma concentration compared to the intra-articular delivery of non-encapsulated mAbs. Our results demonstrate that the utilization of this polymeric technology is a reliable alternative for the local delivery of fully functional clinically relevant mAbs.
Collapse
Affiliation(s)
| | | | | | - Michael Constantinides
- IRMB, Univ Montpellier, INSERM, Montpellier, France; IRMB, CHU Montpellier, Montpellier, France
| | | | | | | | | | | | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, Montpellier, France; IRMB, CHU Montpellier, Montpellier, France
| | - Daniele Noël
- IRMB, Univ Montpellier, INSERM, Montpellier, France; IRMB, CHU Montpellier, Montpellier, France
| | | | | | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, Montpellier, France; IRMB, CHU Montpellier, Montpellier, France; IRMB, Univ Montpellier, INSERM, CNRS, CHU Montpellier, Montpellier, France; Institut Sainte-Catherine, Avignon, France.
| |
Collapse
|
48
|
A triple-sugar regulated Salmonella vaccine protects against Clostridium perfringens-induced necrotic enteritis in broiler chickens. Poult Sci 2021; 101:101592. [PMID: 34922043 PMCID: PMC8686071 DOI: 10.1016/j.psj.2021.101592] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023] Open
Abstract
Gram-positive Clostridium perfringens type G, the causative agent of necrotic enteritis (NE), has gained more attention in the poultry industry due to governmental restrictions on the use of growth-promoting antibiotics in poultry feed. Our previous work has proved that regulated delayed lysis Salmonella vaccines delivering a plasmid encoding an operon fusion of the nontoxic C-terminal adhesive part of alpha toxin and a GST-NetB toxin fusion were able to elicit significant protective immunity in broilers against C. perfringens challenge. We recently improved our S. Typhimurium antigen delivery vaccine strain by integrating a rhamnose-regulated O-antigen synthesis gene enabling a triple-sugar regulation system to control virulence, antigen-synthesis and lysis in vivo traits. The strain also includes a ΔsifA mutation that was previously shown to increase the immunogenicity of and level of protective immunity induced by Salmonella vectored influenza and Eimeria antigens. The new antigen-delivery vaccine vector system confers on the vaccine strain a safe profile and improved protection against C. perfringens challenge. The strain with the triple-sugar regulation system delivering a regulated lysis plasmid pG8R220 encoding the PlcC and GST-NetB antigens protected chickens at a similar level observed in antibiotic-treated chickens. Feed conversion and growth performance were also similar to antibiotic-treated chickens. These studies made use of a severe C. perfringens challenge with lesion formation and mortality enhanced by pre-exposure to Eimeria maxima oocysts. The vaccine achieved effectiveness through three different immunization routes, oral, spray and in drinking water. The vaccine has a potential for application in commercial hatcher and broiler-rearing conditions.
Collapse
|
49
|
Cattel L, Giordano S, Traina S, Lupia T, Corcione S, Angelone L, La Valle G, De Rosa FG, Cattel F. Vaccine development and technology for SARS-CoV-2: Current insight. J Med Virol 2021; 94:878-896. [PMID: 34713912 PMCID: PMC8662109 DOI: 10.1002/jmv.27425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 02/23/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 is associated with a severe respiratory disease in China, that rapidly spread across continents. Since the beginning of the pandemic, available data suggested the asymptomatic transmission and patients were treated with specific drugs with efficacy and safety data not always satisfactory. The aim of this review is to describe the vaccines developed by three companies, Pfizer‐BioNTech, Moderna, and University of Oxford/AstraZeneca, in terms of both technological and pharmaceutical formulation, safety, efficacy, and immunogenicity. A critical analysis of Phases 1, 2, and 3 clinical trial results available was conducted, comparing the three vaccine candidates, underlining their similarities and differences. All candidates showed consistent efficacy and tolerability; although some differences can be noted, such as their technological formulation, temperature storage, which will be related to logistics and costs. Further studies will be necessary to evaluate long‐term effects and to assess the vaccine safety and efficacy in the general population. The rapid development of SARS‐CoV‐2 vaccines has been possible because in the last 20 years there have been rapid improvements in vaccine development in different scientific areas, such as molecular biology, genetic engineering, nano‐materials and lipid nanotechnology. The first three SARS‐CoV‐2 vaccines developed by Pfizer–BioNTech, Moderna and the University of Oxford/AstraZeneca are described, in terms of technology, pharmaceutical formulation, safety, efficacy and immunogenicity. The main detectable difference concerns the technological formulation, influencing logistics: two of these are mRNA‐based vaccines encapsulated in LNPs, whereas one is a DNA‐based vaccine encapsulated in viral vector. mRNA‐based vaccines have an advantage over DNA ones, because mRNA is directly translated in the cytoplasm form ribosomes once inside the cell. Technological and scientific advances allowed the encapsulation of mRNA into custom‐designed LNPs that mimic the structural features of a viral vector. Efficacy and safety studies of Pfizer–BioNTech, Moderna and the University of Oxford/AstraZeneca vaccines are reported, to report the massive pharmaceutical innovation brought.
Collapse
Affiliation(s)
- Luigi Cattel
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Susanna Giordano
- Department of Hospital Pharmacy, Città della Salute e della Scienza, Turin, Italy
| | - Sara Traina
- Department of Hospital Pharmacy, Città della Salute e della Scienza, Turin, Italy
| | - Tommaso Lupia
- Infectious Diseases Unit, Cardinal Massaia Hospital, Asti, Italy
| | - Silvia Corcione
- Department of Medical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA.,Department of Medical Sciences, Infectious Diseases, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Lorenzo Angelone
- General Management, Città della Salute e della Scienza, Turin, Italy
| | - Giovanni La Valle
- General Management, Città della Salute e della Scienza, Turin, Italy
| | - Francesco G De Rosa
- Infectious Diseases Unit, Cardinal Massaia Hospital, Asti, Italy.,Department of Medical Sciences, Infectious Diseases, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Francesco Cattel
- Department of Hospital Pharmacy, Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
50
|
Polla Ravi S, Shamiya Y, Chakraborty A, Elias C, Paul A. Biomaterials, biological molecules, and polymers in developing vaccines. Trends Pharmacol Sci 2021; 42:813-828. [PMID: 34454774 DOI: 10.1016/j.tips.2021.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
Vaccines have been used to train the immune system to recognize pathogens, and prevent and treat diseases, such as cancer, for decades. However, there are continuing challenges in their manufacturing, large-scale production, and storage. Some of them also show suboptimal immunogenicity, requiring additional adjuvants and booster doses. As an alternate vaccination strategy, a new class of biomimetic materials with unique functionalities has emerged in recent years. Here, we explore the current bioengineering techniques that make use of hydrogels, modified polymers, cell membranes, self-assembled proteins, virus-like particles (VLPs), and nucleic acids to deliver and develop biomaterial-based vaccines. We also review design principles and key regulatory issues associated with their development. Finally, we critically assess their limitations, explore approaches to overcome these limitations, and discuss potential future applications for clinical translation.
Collapse
Affiliation(s)
- Shruthi Polla Ravi
- School of Biomedical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Yasmeen Shamiya
- Department of Chemistry, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Cynthia Elias
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada; Biologics Manufacturing Centre, The National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Arghya Paul
- School of Biomedical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada; Department of Chemistry, The University of Western Ontario, London, ON, N6A 5B9, Canada; Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada.
| |
Collapse
|