1
|
Wu H, Weng R, Li J, Huang Z, Tie X, Li J, Chen K. Self-Assembling protein nanoparticle platform for multivalent antigen delivery in vaccine development. Int J Pharm 2025; 676:125597. [PMID: 40233885 DOI: 10.1016/j.ijpharm.2025.125597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025]
Abstract
Nanoparticle vaccines can efficiently and repeatedly display multivalent antigens, thereby improving the targeted delivery of antigens and inducing more durable immune responses, making them an important representative of novel vaccines. The global COVID-19 pandemic has accelerated the development of nanoparticle vaccines, offering a promising solution for the prevention and control of infectious diseases. Currently, the development of nanoparticle vaccines involves the use of various types of nanoparticles, including liposomes, polymers, inorganic materials, and emulsions. Protein nanoparticles candidate vaccines are attracting increasing attention because of their unique antigen presentation methods and self-assembly characteristics during their development, leading to a broad consensus on their promising future. Naturally self-assembling protein nanoparticles, such as ferritin, enhance antigen presentation, which aids in the activation of both humoral and cellular immune responses. This has led to significant advancements in the study of hepatitis B virus. Meanwhile, some synthetically engineered protein nanoparticles, such as mi3, and I53-50, can induce higher antibody titers through chemical conjugation with the SpyTag-SpyCatcher system, thereby providing better immunoprotection and showing promising prospects in the prevention of H1N1 and H3N2 influenza virus infections. This article reviews the unique advantages of protein nanoparticles as antigen delivery platforms, progress made in immunological design mechanisms, advances in the application of related adjuvants in preclinical and clinical trials, and the performance of commonly used computationally designed protein nanoparticles in preclinical trials, with a particular emphasis on the progress in the application of cationic nanoparticle vaccines. The aim is to provide future researchers with effective adjuvant strategies and high-quality selections for computationally designed protein nanoparticles, thereby promoting the clinical trial process of protein nanoparticles vaccines.
Collapse
Affiliation(s)
- Hao Wu
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Ruiqi Weng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jiaxuan Li
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zhiwei Huang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Xiaotian Tie
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jianhua Li
- Zhejiang Key Laboratory of Public Health Detection and Pathogenesis Research, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, PR China.
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China.
| |
Collapse
|
2
|
Chiba S, Halfmann PJ, Iida S, Hirata Y, Sato Y, Kuroda M, Armbrust T, Spyra S, Suzuki T, Kawaoka Y. Correction to "Recombinant Spike protein vaccines coupled with adjuvants that have different modes of action induce protective immunity against SARS-CoV-2" [Vaccine 22 (41) (2023) 6025-6035]. Vaccine 2025; 52:126880. [PMID: 39985967 DOI: 10.1016/j.vaccine.2025.126880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
In the previously published version of the paper, the term "AS03" was used to describe the AddaS03 adjuvant used in animal experiments. This could lead to confusion among the trade and public as to a connection between the AddaS03 adjuvant and GSK's AS03. Upon request by GSK, the authors clarify that no AS03 from GSK was used in this study, and the results obtained with AddaS03 are not transposable to the GSK's AS03 adjuvant. The article has now been corrected, and the conclusions of this paper remain unchanged. Corrections highlighted in bold. The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a glycoprotein, expressed on the virion surface, that mediates infection of host cells by directly interacting with host receptors. As such, it is a reasonable target to neutralize the infectivity of the virus. Here we found that a recombinant S protein vaccine adjuvanted with Alhydrogel or the QS-21-like adjuvant Quil-A effectively induced anti-S receptor binding domain (RBD) serum IgG and neutralizing antibody titers in the Syrian hamster model, resulting in significantly low SARS-CoV-2 replication in respiratory organs and reduced body weight loss upon virus challenge. Severe lung inflammation upon virus challenge was also strongly suppressed by vaccination. We also found that the S protein vaccine adjuvanted with Alhydrogel, Quil-A, or AddaS03 elicited significantly higher neutralizing antibody titers in mice than did unadjuvanted vaccine. Although the neutralizing antibody titers against the variant viruses B.1.351 and B.1.617.2 declined markedly in mice immunized with wild-type S protein, the binding antibody levels against the variant S proteins were equivalent to those against wild-type S. When splenocytes from the immunized mice were re-stimulated with the S protein in vitro, the induced Th1 or Th2 cytokine levels were not significantly different upon re-stimulation with wild-type S or variant S, suggesting that the T-cell responses against the variants were the same as those against the wild-type virus. Upon Omicron XBB-challenge in hamsters, wild-type S-vaccination with Alhydrogel or AddaS03 reduced lung virus titers on Day 3, and the Quil-A adjuvanted group showed less body weight loss, although serum neutralizing antibody titers against XBB were barely detected in vitro. Collectively, recombinant vaccines coupled with different adjuvants may be promising modalities to combat new variant viruses by inducing various arms of the immune response.
Collapse
Affiliation(s)
- Shiho Chiba
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuichiro Hirata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Kuroda
- 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
| | - Samuel Spyra
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - 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; The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan.; The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center, Tokyo 162-8655, Japan.
| |
Collapse
|
3
|
Lang Q, Huang N, Li L, Liu K, Chen H, Liu X, Ge L, Yang X. Novel and efficient yeast-based strategies for subunit vaccine delivery against COVID-19. Int J Biol Macromol 2025; 294:139254. [PMID: 39743073 DOI: 10.1016/j.ijbiomac.2024.139254] [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: 07/15/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025]
Abstract
Yeast shows promise as a delivery system for drugs and vaccines due to its specific targeting and immunogenic properties. The objective of this research is to create novel and effective yeast-based methods for delivering subunit vaccines. Through the modification of yeast expression plasmids and optimization of expression techniques, a new dual-expression system has been developed. We have successfully generated a S. cerevisiae vaccine strain exhibiting stable dual expression of RBD, as well as an inducible S. cerevisiae vaccine strain with dual expression of RBD. The vaccine efficacy assay in mice indicated that the dual-RBD S. cerevisiae vaccine elicited a significantly more robust humoral and mucosal immune response in comparison to the conventional S. cerevisiae vaccine expressing RBD solely on Aga2p. This study demonstrated a cost-effective dual-expression S. cerevisiae system that not only exhibits potential in combating COVID-19, but also harbors the capacity to foster vaccine development against other infectious diseases.
Collapse
Affiliation(s)
- Qiaoli Lang
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Nan Huang
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Liping Li
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Kun Liu
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Hongyu Chen
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Xueqin Liu
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Liangpeng Ge
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China.
| | - Xi Yang
- Institute of Bioengineering, Chongqing Academy of Animal Sciences, Chongqing 402460, China.
| |
Collapse
|
4
|
Huang K, Li N, Li Y, Zhu J, Fan Q, Yang J, Gao Y, Liu Y, Gao S, Zhao P, Wei K, Deng C, Zuo C, Sun Z. Circular mRNA Vaccine against SARS-COV-2 Variants Enabled by Degradable Lipid Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4699-4710. [PMID: 39789795 DOI: 10.1021/acsami.4c20770] [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: 01/12/2025]
Abstract
The emergence of mRNA vaccines offers great promise and a potent platform in combating various diseases, notably COVID-19. Nevertheless, challenges such as inherent instability and potential side effects of current delivery systems underscore the critical need for the advancement of stable, safe, and efficacious mRNA vaccines. In this study, a robust mRNA vaccine (cmRNA-1130) eliciting potent immune activation has been developed from a biodegradable lipid with eight ester bonds in the branched tail (AX4) and synthetic circular mRNA (cmRNA) encoding the trimeric Delta receptor binding domain of the SARS-CoV-2 spike protein. Notably, the cmRNA-1130 vaccine exhibits outstanding stability, remaining effective after six months of storage at 4 °C and multiple freeze-thaw cycles. In comparison with the commercial MC3 lipid, the nanoparticles formed from the degradable AX4 lipid revealed a much faster metabolic rate from the liver and spleen, affording negligible impairment to the hepatorenal function. Following intramuscular administration, cmRNA-1130 generates robust and sustained neutralizing antibodies and induces the activation of Delta RBD-specific CD4+ and CD8+ T effector memory cells (TEM) and Th1-biased T cells in mice. Featured with potent immune activation, high stability, and decent safety, vaccines formed from cmRNA and AX4 hold a huge clinical potential for the prophylaxis and treatment of different diseases.
Collapse
Affiliation(s)
- Ke Huang
- Department of Chemical and Biological Engineering, Monash University, Clayton 3800, Victoria, Australia
| | - Na Li
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Yingwen Li
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Jiafeng Zhu
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Qianyi Fan
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Jiali Yang
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Yinjia Gao
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Yuping Liu
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Shufeng Gao
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Peng Zhao
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Ke Wei
- Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Materials, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Chijian Zuo
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Zhenhua Sun
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| |
Collapse
|
5
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringquist R, Smith C, Ochoa MA, Roy K. A Dual-Adjuvanted Parenteral-Intranasal Subunit Nanovaccine generates Robust Systemic and Mucosal Immunity Against SARS-CoV-2 in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402792. [PMID: 39352717 PMCID: PMC11615772 DOI: 10.1002/advs.202402792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 09/09/2024] [Indexed: 12/06/2024]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters can overcome the shortcomings of parenteral vaccines and enhance pre-existing systemic immunity. Here, a new protein subunit nanovaccine is developed by utilizing dual-adjuvanted (RIG-I: PUUC RNA and TLR-9: CpG DNA) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) along with SARS-CoV-2 S1 trimer protein, that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL subunit nanovaccine, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lungs and showed robust systemic humoral immunity. Interestingly, as a purely intranasal subunit vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. The data suggest that PUUC+CpG PAL subunit nanovaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
MESH Headings
- Animals
- Mice
- Immunity, Mucosal/immunology
- Immunity, Mucosal/drug effects
- SARS-CoV-2/immunology
- Administration, Intranasal/methods
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19/immunology
- COVID-19/prevention & control
- Nanoparticles/administration & dosage
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Antibodies, Viral/immunology
- Female
- Adjuvants, Vaccine/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Neutralizing/immunology
- Mice, Inbred BALB C
- Nanovaccines
Collapse
Affiliation(s)
- Bhawana Pandey
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Zhengying Wang
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Angela Jimenez
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Eshant Bhatia
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Ritika Jain
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Alexander Beach
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Drishti Maniar
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Justin Hosten
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Laura O'Farrell
- Physiological Research LaboratoryGeorgia Institute of TechnologyAtlantaGAUSA
| | - Casey Vantucci
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - David Hur
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Richard Noel
- Physiological Research LaboratoryGeorgia Institute of TechnologyAtlantaGAUSA
| | - Rachel Ringquist
- The Parker H. Petit Institute for Bioengineering and BiosciencesSchool of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Clinton Smith
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Miguel A. Ochoa
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical EngineeringThe Parker H. Petit Institute for Bioengineering and BiosciencesMarcus Center for Therapeutic Cell Characterization and ManufacturingGeorgia Institute of TechnologyAtlantaGAUSA
- Department of Biomedical EngineeringDepartment of Chemical and Biomolecular EngineeringSchool of EngineeringDepartment of Pathology, Microbiology and ImmunologySchool of MedicineVanderbilt UniversityNashvilleTNUSA
| |
Collapse
|
6
|
Azevedo-Silva J, Amorim M, Tavares-Valente D, Sousa P, Mohamath R, Voigt EA, Guderian JA, Kinsey R, Viana S, Reis F, Pintado ME, Paddon CJ, Fox CB, Fernandes JC. Exploring yeast glucans for vaccine enhancement: Sustainable strategies for overcoming adjuvant challenges in a SARS-CoV-2 model. Eur J Pharm Biopharm 2024:114538. [PMID: 39461571 DOI: 10.1016/j.ejpb.2024.114538] [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: 08/02/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Vaccine adjuvants are important for enhancing vaccine efficacy, and although aluminium salts (Alum) are the most used, their limited ability to induce specific immune responses has spurred the search for new adjuvants. However, many adjuvants fail during product development due to manufacturability, supply, stability, or safety concerns. This work hypothesizes that protein-free yeast glucans can be used as vaccine adjuvants due to their known immunostimulatory activity and high abundancy. Thus, high molecular weight glucans with over 99% purity, comprising 64-70% β-glucans and 29-35% α-glucans, were extracted from a wild-type yeast and an engineered yeast to produce a steviol glycoside. These glucans underwent carboxymethylation to enhance solubility. Both water-dispersible and particulate glucans were evaluated as adjuvants, either alone or in combination with Alum or squalene stable emulsion (SE), for a SARS-CoV-2 vaccine. The study demonstrated that glucans triggered a robust immune response and enhanced the effects of Alum and SE when used in combination, both in vitro and in vivo. Water-dispersible glucans combined with Alum, and particulate glucans combined with SE, increased the production of specific antibodies against SARS-CoV-2 spike protein and enhanced serum neutralization titers against SARS-CoV-2 pseudovirus. Furthermore, the results indicated that larger molecular weight glucans from engineered yeast exhibited stronger immunogenic activity in comparison to wild-type yeast glucans. In conclusion, appropriately formulated glucans have the potential to be scalable, low-cost vaccine adjuvants, potentially overcoming the limitations of current adjuvants.
Collapse
Affiliation(s)
- João Azevedo-Silva
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005, Porto, Portugal
| | - Manuela Amorim
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005, Porto, Portugal
| | - Diana Tavares-Valente
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005, Porto, Portugal
| | - Pedro Sousa
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005, Porto, Portugal
| | - Randolph Mohamath
- Access to Advanced Health Institute, Formerly Infectious Disease Research Institute, Seattle, WA, USA
| | - Emily A Voigt
- Access to Advanced Health Institute, Formerly Infectious Disease Research Institute, Seattle, WA, USA
| | - Jeffrey A Guderian
- Access to Advanced Health Institute, Formerly Infectious Disease Research Institute, Seattle, WA, USA
| | - Robert Kinsey
- Access to Advanced Health Institute, Formerly Infectious Disease Research Institute, Seattle, WA, USA
| | - Sofia Viana
- University of Coimbra, Faculty of Medicine, Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), 3000-548, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), 3004-504, Coimbra, Portugal; Pharmacy, Coimbra Health School, Polytechnic Institute of Coimbra, Rua 5 de Outubro-SM Bispo, Apartado 7006 3046-854, Coimbra, Portugal
| | - Flávio Reis
- University of Coimbra, Faculty of Medicine, Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), 3000-548, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), 3004-504, Coimbra, Portugal
| | - Manuela E Pintado
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005, Porto, Portugal
| | | | - Christopher B Fox
- Access to Advanced Health Institute, Formerly Infectious Disease Research Institute, Seattle, WA, USA
| | - João C Fernandes
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005, Porto, Portugal.
| |
Collapse
|
7
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 PMCID: PMC11736809 DOI: 10.1016/j.cell.2024.07.021] [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: 01/24/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.
Collapse
Affiliation(s)
| | - Galit Alter
- Moderna Therapeutics, Cambridge, MA 02139, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Arunachalam PS, Ha N, Dennison SM, Spreng RL, Seaton KE, Xiao P, Feng Y, Zarnitsyna VI, Kazmin D, Hu M, Santagata JM, Xie X, Rogers K, Shirreff LM, Chottin C, Spencer AJ, Dutta S, Prieto K, Julien JP, Tomai M, Fox CB, Villinger F, Hill AVS, Tomaras GD, Pulendran B. A comparative immunological assessment of multiple clinical-stage adjuvants for the R21 malaria vaccine in nonhuman primates. Sci Transl Med 2024; 16:eadn6605. [PMID: 39083589 DOI: 10.1126/scitranslmed.adn6605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Authorization of the Matrix-M (MM)-adjuvanted R21 vaccine by three countries and its subsequent endorsement by the World Health Organization for malaria prevention in children are a milestone in the fight against malaria. Yet, our understanding of the innate and adaptive immune responses elicited by this vaccine remains limited. Here, we compared three clinically relevant adjuvants [3M-052 + aluminum hydroxide (Alum) (3M), a TLR7/8 agonist formulated in Alum; GLA-LSQ, a TLR4 agonist formulated in liposomes with QS-21; and MM, the now-approved adjuvant for R21] for their capacity to induce durable immune responses to R21 in macaques. R21 adjuvanted with 3M on a 0, 8, and 23-week schedule elicited anti-circumsporozoite antibody responses comparable in magnitude to the R21/MM vaccine administered using a 0-4-8-week regimen and persisted up to 72 weeks with a half-life of 337 days. A booster dose at 72 weeks induced a recall response similar to the R21/MM vaccination. In contrast, R21/GLA-LSQ immunization induced a lower, short-lived response at the dose used. Consistent with the durable serum antibody responses, MM and 3M induced long-lived plasma cells in the bone marrow and other tissues, including the spleen. Furthermore, whereas 3M stimulated potent and persistent antiviral transcriptional and cytokine signatures after primary and booster immunizations, MM induced enhanced expression of interferon- and TH2-related signatures more highly after the booster vaccination. Collectively, these findings provide a resource on the immune responses of three clinically relevant adjuvants with R21 and highlight the promise of 3M as another adjuvant for malarial vaccines.
Collapse
Affiliation(s)
- Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - NaYoung Ha
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - S Moses Dennison
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
| | - Rachel L Spreng
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
- Duke Human Vaccine Institute, Duke University, Durham, NC 27703, USA
| | - Kelly E Seaton
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
| | - Peng Xiao
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | | - Dmitri Kazmin
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jordan M Santagata
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Xia Xie
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kenneth Rogers
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Lisa M Shirreff
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Claire Chottin
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | | | - Sheetij Dutta
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Katherine Prieto
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Departments of Biochemistry and Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | | | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
- Duke Human Vaccine Institute, Duke University, Durham, NC 27703, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
9
|
de Lima VA, Nunes JPS, Rosa DS, Ferreira R, Oliva MLV, Andreata‐Santos R, Duarte‐Barbosa M, Janini LMR, Maricato JT, Akamatsu MA, Ho PL, Schenkman S. Development and characterization of a multimeric recombinant protein using the spike protein receptor binding domain as an antigen to induce SARS-CoV-2 neutralization. Immun Inflamm Dis 2024; 12:e1353. [PMID: 39056544 PMCID: PMC11273545 DOI: 10.1002/iid3.1353] [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: 03/11/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND SARS-CoV2 virus, responsible for the COVID-19 pandemic, has four structural proteins and 16 nonstructural proteins. S-protein is one of the structural proteins exposed on the virus surface and is the main target for producing neutralizing antibodies and vaccines. The S-protein forms a trimer that can bind the angiotensin-converting enzyme 2 (ACE2) through its receptor binding domain (RBD) for cell entry. AIMS The goal of this study was to express in HEK293 cells a new RBD recombinant protein in a constitutive and stable manner in order to use it as an alternative immunogen and diagnostic tool for COVID-19. MATERIALS & METHODS The protein was designed to contain an immunoglobulin signal sequence, an explanded C-terminal section of the RBD, a region responsible for the bacteriophage T4 trimerization inducer, and six histidines in the pCDNA-3.1 plasmid. Following transformation, the cells were selected with geneticin-G418 and purified from serum-fre culture supernatants using Ni2+-agarand size exclusion chromatography. The protein was structurally identified by cross-linking and circular dichroism experiments, and utilized to immunize mice in conjuction with AS03 or alum adjuvants. The mice sera were examined for antibody recognition, receptor-binding inhibition, and virus neutralization, while spleens were evaluated for γ-interferon production in the presence of RBD. RESULTS The protein released in the culture supernatant of cells, and exhibited a molecular mass of 135 kDa with a secondary structure like the monomeric and trimeric RBD. After purification, it formed a multimeric structure comprising trimers and hexamers, which were able to bind the ACE2 receptor. It generated high antibody titers in mice when combined with AS03 adjuvant (up to 1:50,000). The sera were capable of inhibiting binding of biotin-labeled ACE2 to the virus S1 subunit and could neutralize the entry of the Wuhan virus strain into cells at dilutions up to 1:2000. It produced specific IFN-γ producing cells in immunized mouse splenocytes. DISCUSSION Our data describe a new RBD containing protein, forming trimers and hexamers, which are able to induce a protective humoral and cellular response against SARS-CoV2. CONCLUSION These results add a new arsenal to combat COVID-19, as an alternative immunogen or antigen for diagnosis.
Collapse
Affiliation(s)
- Veronica A. de Lima
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - João P. S. Nunes
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Daniela S. Rosa
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Rodrigo Ferreira
- Department of Biochemistry, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Maria L. V. Oliva
- Department of Biochemistry, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Robert Andreata‐Santos
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Marcia Duarte‐Barbosa
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Luiz M. R. Janini
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Juliana T. Maricato
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Milena A. Akamatsu
- Núcleo de Produção de Vacinas Bacterianas, Centro BioIndustrial, Instituto ButantanSão PauloSão PauloBrazil
| | - Paulo L. Ho
- Núcleo de Produção de Vacinas Bacterianas, Centro BioIndustrial, Instituto ButantanSão PauloSão PauloBrazil
| | - Sergio Schenkman
- Department of Microbiology, Immunology and ParasitologyUniversidade Federal de São PauloSão PauloSão PauloBrazil
| |
Collapse
|
10
|
Kawai T, Ikegawa M, Ori D, Akira S. Decoding Toll-like receptors: Recent insights and perspectives in innate immunity. Immunity 2024; 57:649-673. [PMID: 38599164 DOI: 10.1016/j.immuni.2024.03.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024]
Abstract
Toll-like receptors (TLRs) are an evolutionarily conserved family in the innate immune system and are the first line of host defense against microbial pathogens by recognizing pathogen-associated molecular patterns (PAMPs). TLRs, categorized into cell surface and endosomal subfamilies, recognize diverse PAMPs, and structural elucidation of TLRs and PAMP complexes has revealed their intricate mechanisms. TLRs activate common and specific signaling pathways to shape immune responses. Recent studies have shown the importance of post-transcriptional regulation in TLR-mediated inflammatory responses. Despite their protective functions, aberrant responses of TLRs contribute to inflammatory and autoimmune disorders. Understanding the delicate balance between TLR activation and regulatory mechanisms is crucial for deciphering their dual role in immune defense and disease pathogenesis. This review provides an overview of recent insights into the history of TLR discovery, elucidation of TLR ligands and signaling pathways, and their relevance to various diseases.
Collapse
Affiliation(s)
- Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan; Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan.
| | - Moe Ikegawa
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Daisuke Ori
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Shizuo Akira
- Center for Advanced Modalities and DSS (CAMaD), Osaka University, Osaka 565-0871, Japan; Laboratory of Host Defense, Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan; Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
11
|
Liu R, Natekar JP, Kim KH, Pathak H, Bhatnagar N, Raha JR, Park BR, Guglani A, Shin CH, Kumar M, Kang SM. Multivalent and Sequential Heterologous Spike Protein Vaccinations Effectively Induce Protective Humoral Immunity against SARS-CoV-2 Variants. Vaccines (Basel) 2024; 12:362. [PMID: 38675744 PMCID: PMC11053539 DOI: 10.3390/vaccines12040362] [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: 03/06/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of new SARS-CoV-2 variants continues to cause challenging problems for the effective control of COVID-19. In this study, we tested the hypothesis of whether a strategy of multivalent and sequential heterologous spike protein vaccinations would induce a broader range and higher levels of neutralizing antibodies against SARS-CoV-2 variants and more effective protection than homologous spike protein vaccination in a mouse model. We determined spike-specific IgG, receptor-binding inhibition titers, and protective efficacy in the groups of mice that were vaccinated with multivalent recombinant spike proteins (Wuhan, Delta, Omicron), sequentially with heterologous spike protein variants, or with homologous spike proteins. Trivalent (Wuhan + Delta + Omicron) and sequential heterologous spike protein vaccinations were more effective in inducing serum inhibition activities of receptor binding to spike variants and virus neutralizing antibody titers than homologous spike protein vaccination. The higher efficacy of protection was observed in mice with trivalent and sequential heterologous spike protein vaccination after a challenge with a mouse-adapted SARS-CoV-2 MA10 strain compared to homologous spike protein vaccination. This study provides evidence that a strategy of multivalent and sequential heterologous variant spike vaccination might provide more effective protection against emerging SARS-CoV-2 variants than homologous spike vaccination and significantly alleviate severe inflammation due to COVID-19.
Collapse
Affiliation(s)
- Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Janhavi P. Natekar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Heather Pathak
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Anchala Guglani
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| |
Collapse
|
12
|
Silva MDO, Castro-Amarante MF, Venceslau-Carvalho AA, Almeida BDS, Daher IP, de Souza-Silva GA, Yamamoto MM, Koike G, de Souza EE, Wrenger C, Ferreira LCDS, Boscardin SB. Enhanced Immunogenicity and Protective Effects against SARS-CoV-2 Following Immunization with a Recombinant RBD-IgG Chimeric Protein. Vaccines (Basel) 2024; 12:356. [PMID: 38675739 PMCID: PMC11054318 DOI: 10.3390/vaccines12040356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 04/28/2024] Open
Abstract
The unprecedented global impact caused by SARS-CoV-2 imposed huge health and economic challenges, highlighting the urgent need for safe and effective vaccines. The receptor-binding domain (RBD) of SARS-CoV-2 is the major target for neutralizing antibodies and for vaccine formulations. Nonetheless, the low immunogenicity of the RBD requires the use of alternative strategies to enhance its immunological properties. Here, we evaluated the use of a subunit vaccine antigen generated after the genetic fusing of the RBD with a mouse IgG antibody. Subcutaneous administration of RBD-IgG led to the extended presence of the protein in the blood of immunized animals and enhanced RBD-specific IgG titers. Furthermore, RBD-IgG immunized mice elicited increased virus neutralizing antibody titers, measured both with pseudoviruses and with live original (Wuhan) SARS-CoV-2. Immunized K18-hACE2 mice were fully resistant to the lethal challenge of the Wuhan SARS-CoV-2, demonstrated by the control of body-weight loss and virus loads in their lungs and brains. Thus, we conclude that the genetic fusion of the RBD with an IgG molecule enhanced the immunogenicity of the antigen and the generation of virus-neutralizing antibodies, supporting the use of IgG chimeric antigens as an approach to improve the performance of SARS-CoV-2 subunit vaccines.
Collapse
Affiliation(s)
- Mariângela de Oliveira Silva
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Maria Fernanda Castro-Amarante
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Alexia Adrianne Venceslau-Carvalho
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Bianca da Silva Almeida
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Isabela Pazotti Daher
- Laboratory of Immunology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo 05403-000, Brazil
| | - Guilherme Antonio de Souza-Silva
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Marcio Massao Yamamoto
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Gabriela Koike
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| | - Edmarcia Elisa de Souza
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Luís Carlos de Souza Ferreira
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.d.O.S.)
| |
Collapse
|
13
|
D'Oro U, O'Hagan DT. The scientific journey of a novel adjuvant (AS37) from bench to bedside. NPJ Vaccines 2024; 9:26. [PMID: 38332005 PMCID: PMC10853242 DOI: 10.1038/s41541-024-00810-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
A decade ago, we described a new approach to discover next generation adjuvants, identifying small-molecule immune potentiators (SMIPs) as Toll-like receptor (TLR)7 agonists. We also optimally formulated these drugs through adsorption to aluminum salts (alum), allowing them to be evaluated with a range of established and early-stage vaccines. Early proof-of-concept studies showed that a TLR7 agonist (TLR7a)-based SMIP, when adsorbed to alum, could perform as an effective adjuvant for a variety of different antigens, in both small and large animals. Studies in rodents demonstrated that the adjuvant enhanced immunogenicity of a recombinant protein-based vaccine against Staphylococcus aureus, and also showed potential to improve existing vaccines against pertussis or meningococcal infection. Extensive evaluations showed that the adjuvant was effective in non-human primates (NHPs), exploiting a mechanism of action that was consistent across the different animal models. The adjuvant formulation (named AS37) has now been advanced into clinical evaluation. A systems biology-based evaluation of the phase I clinical data with a meningococcal C conjugate vaccine showed that the AS37-adjuvanted formulation had an acceptable safety profile, was potent, and activated the expected immune pathways in humans, which was consistent with observations from the NHP studies. In the intervening decade, several alternative TLR7 agonists have also emerged and advanced into clinical development, such as the alum adsorbed TLR7/8 SMIP present in a widely distributed COVID-19 vaccine. This review summarizes the research and early development of the new adjuvant AS37, with an emphasis on the steps taken to allow its progression into clinical evaluations.
Collapse
|
14
|
Sui Y, Andersen H, Li J, Hoang T, Minai M, Nagata BM, Bock KW, Alves DA, Lewis MG, Berzofsky JA. SARS-CoV-2 mucosal vaccine protects against clinical disease with sex bias in efficacy. Vaccine 2024; 42:339-351. [PMID: 38071106 PMCID: PMC10843685 DOI: 10.1016/j.vaccine.2023.11.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/14/2023] [Accepted: 11/28/2023] [Indexed: 01/01/2024]
Abstract
Intranasal mucosal vaccines can more effectively induce mucosal immune responses against SARS-CoV-2. Here, we show in hamsters that an intranasal subunit mucosal vaccine boost with the beta variant S1 can prevent weight loss, in addition to reducing viral load, which cannot be studied in macaques that don't develop COVID-like disease. Protective efficacy against both viral load and weight loss correlated with serum antibody titers. A sex bias was detected in that immune responses and protection against viral load were greater in females than males. We also found that priming with S1 from the Wuhan strain elicited lower humoral immune responses against beta variant and led to less protection against beta viral challenge, suggesting the importance of matched antigens. The greater efficacy of mucosal vaccines in the upper respiratory tract and the need to consider sex differences in vaccine protection are important in the development of future improved COVID-19 vaccines.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, Center of for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | | | - Jianping Li
- Vaccine Branch, Center of for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Tanya Hoang
- Vaccine Branch, Center of for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Derron A Alves
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | | | - Jay A Berzofsky
- Vaccine Branch, Center of for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Zhang T, Zheng N, Wang Z, Xu X. Structure-based design of oligomeric receptor-binding domain (RBD) recombinant proteins as potent vaccine candidates against SARS-CoV-2. Hum Vaccin Immunother 2023; 19:2174755. [PMID: 36846890 PMCID: PMC10026890 DOI: 10.1080/21645515.2023.2174755] [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: 03/01/2023] Open
Abstract
The receptor-binding domain (RBD) of SARS-CoV-2 S protein is proved to be the major target of neutralizing antibodies. However, on the S protein, only a portion of epitopes in RBD can be effectively displayed with dynamic changes in spatial conformations. Using RBD fragment as antigen can better expose the neutralizing epitopes, but the immunogenicity of RBD monomer is suboptimal. Multimeric display of RBD molecules is a feasible strategy to optimize RBD-based vaccines. In this study, RBD single-chain dimer derived from Wuhan-Hu-1 was fused with a trimerization motif, and a cysteine was also introduced at the C-terminus. The resultant recombinant protein 2RBDpLC was expressed in Sf9 cells using a baculovirus expression system. Reducing/non-reducing PAGE, size-exclusion chromatography and in silico structure prediction indicated that 2RBDpLC polymerized and possibly formed RBD dodecamers through trimerization motif and intermolecular disulfide bonds. In mice, 2RBDpLC induced higher levels of RBD-specific and neutralizing antibody responses than RBD dimer, RBD trimer and prefusion-stabilized S protein (S2P). In addition, cross-neutralizing antibodies against Delta and Omicron VOC were also detected in the immune sera. Our results demonstrate that 2RBDpLC is a promising vaccine candidate, and the method of constructing dodecamers may be an effective strategy for designing RBD-based vaccines.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Ningchen Zheng
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zhirong Wang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xuemei Xu
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Barbey C, Su J, Billmeier M, Stefan N, Bester R, Carnell G, Temperton N, Heeney J, Protzer U, Breunig M, Wagner R, Peterhoff D. Immunogenicity of a silica nanoparticle-based SARS-CoV-2 vaccine in mice. Eur J Pharm Biopharm 2023; 192:41-55. [PMID: 37774890 DOI: 10.1016/j.ejpb.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Safe and effective vaccines have been regarded early on as critical in combating the COVID-19 pandemic. Among the deployed vaccine platforms, subunit vaccines have a particularly good safety profile but may suffer from a lower immunogenicity compared to mRNA based or viral vector vaccines. In fact, this phenomenon has also been observed for SARS-CoV-2 subunit vaccines comprising the receptor-binding domain (RBD) of the spike (S) protein. Therefore, RBD-based vaccines have to rely on additional measures to enhance the immune response. It is well accepted that displaying antigens on nanoparticles can improve the quantity and quality of vaccine-mediated both humoral and cell-mediated immune responses. Based on this, we hypothesized that SARS-CoV-2 RBD as immunogen would benefit from being presented to the immune system via silica nanoparticles (SiNPs). Herein we describe the preparation, in vitro characterization, antigenicity and in vivo immunogenicity of SiNPs decorated with properly oriented RBD in mice. We found our RBD-SiNP conjugates show narrow, homogeneous particle distribution with optimal size of about 100 nm for efficient transport to and into the lymph node. The colloidal stability and binding of the antigen was stable for at least 4 months at storage- and in vivo-temperatures. The antigenicity of the RBD was maintained upon binding to the SiNP surface, and the receptor-binding motif was readily accessible due to the spatial orientation of the RBD. The particles were efficiently taken up in vitro by antigen-presenting cells. In a mouse immunization study using an mRNA vaccine and spike protein as benchmarks, we found that the SiNP formulation was able to elicit a stronger RBD-specific humoral response compared to the soluble protein. For the adjuvanted RBD-SiNP we found strong S-specific multifunctional CD4+ T cell responses, a balanced T helper response, improved auto- and heterologous virus neutralization capacity, and increased serum avidity, suggesting increased affinity maturation. In summary, our results provide further evidence for the possibility of optimizing the cellular and humoral immune response through antigen presentation on SiNP.
Collapse
Affiliation(s)
- Clara Barbey
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Jinpeng Su
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany
| | - Martina Billmeier
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Nadine Stefan
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Romina Bester
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany
| | - George Carnell
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham ME4 4BF, United Kingdom
| | - Jonathan Heeney
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
17
|
Ou BS, Saouaf OM, Yan J, Bruun TUJ, Baillet J, Zhou X, King NP, Appel EA. Broad and Durable Humoral Responses Following Single Hydrogel Immunization of SARS-CoV-2 Subunit Vaccine. Adv Healthc Mater 2023; 12:e2301495. [PMID: 37278391 DOI: 10.1002/adhm.202301495] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 06/07/2023]
Abstract
Most vaccines require several immunizations to induce robust immunity, and indeed, most SARS-CoV-2 vaccines require an initial two-shot regimen followed by several boosters to maintain efficacy. Such a complex series of immunizations unfortunately increases the cost and complexity of populations-scale vaccination and reduces overall compliance and vaccination rate. In a rapidly evolving pandemic affected by the spread of immune-escaping variants, there is an urgent need to develop vaccines capable of providing robust and durable immunity. In this work, a single immunization SARS-CoV-2 subunit vaccine is developed that can rapidly generate potent, broad, and durable humoral immunity. Injectable polymer-nanoparticle (PNP) hydrogels are leveraged as a depot technology for the sustained delivery of a nanoparticle antigen (RND-NP) displaying multiple copies of the SARS-CoV-2 receptor-binding domain (RBD) and potent adjuvants including CpG and 3M-052. Compared to a clinically relevant prime-boost regimen with soluble vaccines formulated with CpG/alum or 3M-052/alum adjuvants, PNP hydrogel vaccines more rapidly generated higher, broader, and more durable antibody responses. Additionally, these single-immunization hydrogel-based vaccines elicit potent and consistent neutralizing responses. Overall, it is shown that PNP hydrogels elicit improved anti-COVID immune responses with only a single administration, demonstrating their potential as critical technologies to enhance overall pandemic readiness.
Collapse
Affiliation(s)
- Ben S Ou
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Olivia M Saouaf
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Jerry Yan
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Theodora U J Bruun
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Julie Baillet
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
- CNRS, Bordeaux INP, LCPO, University of Bordeaux, Pessac, 33600, France
| | - Xueting Zhou
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, 98109, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pediatrics-Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Woods Institute for the Environment, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
18
|
Routhu NK, Stampfer SD, Lai L, Akhtar A, Tong X, Yuan D, Chicz TM, McNamara RP, Jakkala K, Davis-Gardner ME, St Pierre EL, Smith B, Green KM, Golden N, Picou B, Jean SM, Wood J, Cohen J, Moore IN, Patel N, Guebre-Xabier M, Smith G, Glenn G, Kozlowski PA, Alter G, Ahmed R, Suthar MS, Amara RR. Efficacy of mRNA-1273 and Novavax ancestral or BA.1 spike booster vaccines against SARS-CoV-2 BA.5 infection in nonhuman primates. Sci Immunol 2023; 8:eadg7015. [PMID: 37191508 PMCID: PMC10451060 DOI: 10.1126/sciimmunol.adg7015] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Omicron SARS-CoV-2 variants escape vaccine-induced neutralizing antibodies and cause nearly all current COVID-19 cases. Here, we compared the efficacy of three booster vaccines against Omicron BA.5 challenge in rhesus macaques: mRNA-1273, the Novavax ancestral spike protein vaccine (NVX-CoV2373), or Omicron BA.1 spike protein version (NVX-CoV2515). All three booster vaccines induced a strong BA.1 cross-reactive binding antibody and changed immunoglobulin G (Ig) dominance from IgG1 to IgG4 in the serum. All three booster vaccines also induced strong and comparable neutralizing antibody responses against multiple variants of concern, including BA.5 and BQ.1.1, along with long-lived plasma cells in the bone marrow. The ratio of BA.1 to WA-1 spike-specific antibody-secreting cells in the blood was higher in NVX-CoV2515 animals compared with NVX-CoV2373 animals, suggesting a better recall of BA.1-specific memory B cells by the BA.1 spike-specific vaccine compared with the ancestral spike-specific vaccine. Further, all three booster vaccines induced low levels of spike-specific CD4 but not CD8 T cell responses in the blood. After challenge with SARS-CoV-2 BA.5 variant, all three vaccines showed strong protection in the lungs and controlled virus replication in the nasopharynx. In addition, both Novavax vaccines blunted viral replication in nasopharynx at day 2. The protection against SARS-CoV-2 BA.5 infection in the upper respiratory airways correlated with binding, neutralizing, and ADNP activities of the serum antibody. These data have important implications for COVID-19 vaccine development, because vaccines that lower nasopharyngeal virus may help to reduce transmission.
Collapse
Affiliation(s)
- Nanda Kishore Routhu
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Samuel David Stampfer
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lilin Lai
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Akil Akhtar
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - Xin Tong
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Dansu Yuan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Taras M. Chicz
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ryan P. McNamara
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Kishor Jakkala
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Meredith E. Davis-Gardner
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Brandon Smith
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Breanna Picou
- Tulane National Primate Research Center, Covington, LA, USA
| | - Sherrie M. Jean
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joyce Cohen
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ian N. Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Nita Patel
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, MD 20878, USA
| | | | - Gale Smith
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, MD 20878, USA
| | - Greg Glenn
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, MD 20878, USA
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - Mehul S. Suthar
- Department of Pediatrics, Division of Infectious Diseases Vaccine Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329
| | - Rama Rao Amara
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
19
|
Chiba S, Halfmann PJ, Iida S, Hirata Y, Sato Y, Kuroda M, Armbrust T, Spyra S, Suzuki T, Kawaoka Y. Recombinant spike protein vaccines coupled with adjuvants that have different modes of action induce protective immunity against SARS-CoV-2. Vaccine 2023; 41:6025-6035. [PMID: 37635002 DOI: 10.1016/j.vaccine.2023.08.054] [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: 01/06/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 08/29/2023]
Abstract
The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a glycoprotein, expressed on the virion surface, that mediates infection of host cells by directly interacting with host receptors. As such, it is a reasonable target to neutralize the infectivity of the virus. Here we found that a recombinant S protein vaccine adjuvanted with Alhydrogel or the QS-21-like adjuvant Quil-A effectively induced anti-S receptor binding domain (RBD) serum IgG and neutralizing antibody titers in the Syrian hamster model, resulting in significantly low SARS-CoV-2 replication in respiratory organs and reduced body weight loss upon virus challenge. Severe lung inflammation upon virus challenge was also strongly suppressed by vaccination. We also found that the S protein vaccine adjuvanted with Alhydrogel, Quil-A, or an AS03-like adjuvant elicited significantly higher neutralizing antibody titers in mice than did unadjuvanted vaccine. Although the neutralizing antibody titers against the variant viruses B.1.351 and B.1.617.2 declined markedly in mice immunized with wild-type S protein, the binding antibody levels against the variant S proteins were equivalent to those against wild-type S. When splenocytes from the immunized mice were re-stimulated with the S protein in vitro, the induced Th1 or Th2 cytokine levels were not significantly different upon re-stimulation with wild-type S or variant S, suggesting that the T-cell responses against the variants were the same as those against the wild-type virus. Upon Omicron XBB-challenge in hamsters, wild-type S-vaccination with Alhydrogel or AS03 reduced lung virus titers on Day 3, and the Quil-A adjuvanted group showed less body weight loss, although serum neutralizing antibody titers against XBB were barely detected in vitro. Collectively, recombinant vaccines coupled with different adjuvants may be promising modalities to combat new variant viruses by inducing various arms of the immune response.
Collapse
Affiliation(s)
- Shiho Chiba
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuichiro Hirata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Kuroda
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Tammy Armbrust
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Samuel Spyra
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA; Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan; Pandemic Preparedness, Infection and Advanced Research Center, The University of Tokyo, Tokyo 162-8655, Japan.
| |
Collapse
|
20
|
Zhang Y, Kang X, Liu S, Han P, Lei W, Xu K, Xu Z, Gao Z, Zhou X, An Y, Han Y, Liu K, Zhao X, Dai L, Wang P, Wu G, Qi J, Xu K, Gao GF. Broad protective RBD heterotrimer vaccines neutralize SARS-CoV-2 including Omicron sub-variants XBB/BQ.1.1/BF.7. PLoS Pathog 2023; 19:e1011659. [PMID: 37721934 PMCID: PMC10538664 DOI: 10.1371/journal.ppat.1011659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 09/28/2023] [Accepted: 09/04/2023] [Indexed: 09/20/2023] Open
Abstract
SARS-CoV-2 variants with severe immune evasion are a major challenge for COVID-19 prevention, especially the circulating Omicron XBB/BQ.1.1/BF.7 strains. Thus, the next-generation of broad-spectrum vaccines are urgently needed. Previously, we developed a COVID-19 protein subunit vaccine, ZF2001, based on the RBD-homodimer as the immunogen. To adapt SARS-CoV-2 variants, we developed chimeric RBD-heterodimers to induce broad immune responses. In this study, we further explored the concept of tandem RBD homotrimer and heterotrimer. Prototype SARS-CoV-2 RBD-homotrimer, prototype-Delta-BA.1 (PDO) RBD-heterotrimer and Delta-BA.2-BA.5 (DBA2BA5) RBD-heterotrimer were designed. Biochemical and cryo-EM structural characterization demonstrated total epitope exposure of the RBD-trimers. In mouse experiments, PDO and DBA2BA5 elicited broad SARS-CoV-2 neutralization. Potent protection against SARS-CoV-2 variants was observed in challenge assays and was correlated with neutralizing antibody titer. This study validated the design strategy of tandem RBD-heterotrimers as multivalent immunogens and presented a promising vaccine candidate, DBA2BA5, eliciting broad-spectrum immune responses, including against the circulating XBB/BF.7/BQ.1.1.
Collapse
Affiliation(s)
- Yanfang Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xinrui Kang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Sheng Liu
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, China
| | - Pu Han
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wenwen Lei
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ke Xu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zepeng Xu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Zhengrong Gao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Children’s Hospital, Shenzhen, China
| | - Xuemei Zhou
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Hebei University, Baoding, China
| | - Yaling An
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuxuan Han
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Kefang Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Peiyi Wang
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, China
| | - Guizhen Wu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - George F. Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
21
|
Khalid K, Poh CL. The development of DNA vaccines against SARS-CoV-2. Adv Med Sci 2023; 68:213-226. [PMID: 37364379 PMCID: PMC10290423 DOI: 10.1016/j.advms.2023.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/07/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND The COVID-19 pandemic exerted significant impacts on public health and global economy. Research efforts to develop vaccines at warp speed against SARS-CoV-2 led to novel mRNA, viral vectored, and inactivated vaccines being administered. The current COVID-19 vaccines incorporate the full S protein of the SARS-CoV-2 Wuhan strain but rapidly emerging variants of concern (VOCs) have led to significant reductions in protective efficacies. There is an urgent need to develop next-generation vaccines which could effectively prevent COVID-19. METHODS PubMed and Google Scholar were systematically reviewed for peer-reviewed papers up to January 2023. RESULTS A promising solution to the problem of emerging variants is a DNA vaccine platform since it can be easily modified. Besides expressing whole protein antigens, DNA vaccines can also be constructed to include specific nucleotide genes encoding highly conserved and immunogenic epitopes from the S protein as well as from other structural/non-structural proteins to develop effective vaccines against VOCs. DNA vaccines are associated with low transfection efficiencies which could be enhanced by chemical, genetic, and molecular adjuvants as well as delivery systems. CONCLUSIONS The DNA vaccine platform offers a promising solution to the design of effective vaccines. The challenge of limited immunogenicity in humans might be solved through the use of genetic modifications such as the addition of nuclear localization signal (NLS) peptide gene, strong promoters, MARs, introns, TLR agonists, CD40L, and the development of appropriate delivery systems utilizing nanoparticles to increase uptake by APCs in enhancing the induction of potent immune responses.
Collapse
Affiliation(s)
- Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia.
| |
Collapse
|
22
|
Guo M, Xiong M, Peng J, Guan T, Su H, Huang Y, Yang CG, Li Y, Boraschi D, Pillaiyar T, Wang G, Yi C, Xu Y, Chen C. Multi-omics for COVID-19: driving development of therapeutics and vaccines. Natl Sci Rev 2023; 10:nwad161. [PMID: 37936830 PMCID: PMC10627145 DOI: 10.1093/nsr/nwad161] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 11/09/2023] Open
Abstract
The ongoing COVID-19 pandemic caused by SARS-CoV-2 has raised global concern for public health and economy. The development of therapeutics and vaccines to combat this virus is continuously progressing. Multi-omics approaches, including genomics, transcriptomics, proteomics, metabolomics, epigenomics and metallomics, have helped understand the structural and molecular features of the virus, thereby assisting in the design of potential therapeutics and accelerating vaccine development for COVID-19. Here, we provide an up-to-date overview of the latest applications of multi-omics technologies in strategies addressing COVID-19, in order to provide suggestions towards the development of highly effective knowledge-based therapeutics and vaccines.
Collapse
Affiliation(s)
- Mengyu Guo
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Muya Xiong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinying Peng
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Tong Guan
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haixia Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyi Huang
- Biomedical Pioneering Innovation Centre, Peking University, Beijing 100871, China
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 528107, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - Cai-Guang Yang
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Li
- Laboratory of Immunology and Nanomedicine, and China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Diana Boraschi
- Laboratory of Immunology and Nanomedicine, and China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Institute of Biochemistry and Cell Biology, National Research Council, Napoli 80131, Italy
| | - Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Guanbo Wang
- Biomedical Pioneering Innovation Centre, Peking University, Beijing 100871, China
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 528107, China
| | - Chengqi Yi
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
- Department of Chemical Biology and Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yechun Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunying Chen
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- GBA National Institute for Nanotechnology Innovation, Guangzhou 510700, China
| |
Collapse
|
23
|
Laotee S, Duangkaew M, Jivapetthai A, Tharakhet K, Kaewpang P, Prompetchara E, Phumiamorn S, Sapsutthipas S, Trisiriwanich S, Somsaard T, Roytrakul S, Duangkhae P, Ongpipattanakul B, Limpikirati P, Pornputtapong N, Arunmanee W. CHO-produced RBD-Fc subunit vaccines with alternative adjuvants generate immune responses against SARS-CoV-2. PLoS One 2023; 18:e0288486. [PMID: 37450510 PMCID: PMC10348575 DOI: 10.1371/journal.pone.0288486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023] Open
Abstract
Subunit vaccines feature critical advantages over other vaccine platforms such as stability, price, and minimal adverse effects. To maximize immunological protection of subunit vaccines, adjuvants are considered as main components that are formulated within the subunit vaccine. They can modulate adverse effects and enhance immune outcomes. However, the most suitable formulation providing the best immunological outcomes and safety are still under investigation. In this report, we combined recombinant RBD with human IgG1 Fc to create an RBD dimer. This fusion protein was expressed in CHO and formulated with alternative adjuvants with different immune activation including Montanide ISA51, Poly (I:C), and MPLA/Quil-A® as potential vaccine candidate formulations. Using the murine model, a potent induction of anti-RBD IgG antibodies in immunized mice sera were observed. IgG subclass analyses (IgG1/IgG2a) illustrated that all adjuvanted formulations could stimulate both Th1 and Th2-type immune responses in particular Poly (I:C) and MPLA/Quil-A®, eliciting greater balance. In addition, Montanide ISA51-formulated RBD-Fc vaccination provided a promising level of neutralizing antibodies against live wild-type SARS-CoV-2 in vitro followed by Poly (I:C) and MPLA/Quil-A®, respectively. Also, mice sera from adjuvanted formulations could strongly inhibit RBD:ACE2 interaction. This study offers immunogenicity profiles, forecasted safety based on Vaccine-associated enhanced disease (VAED) caused by Th1-skewed immunity, and neutralizing antibody analysis of candidates of RBD-Fc-based subunit vaccine formulations to obtain an alternative subunit vaccine formulation against SARS-CoV-2.
Collapse
Affiliation(s)
- Sedthawut Laotee
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Methawee Duangkaew
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Araya Jivapetthai
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Kittipan Tharakhet
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Papatsara Kaewpang
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Eakachai Prompetchara
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Integrated Frontier Biotechnology for Emerging Disease, Chulalongkorn University, Bangkok, Thailand
| | - Supaporn Phumiamorn
- Institute of Biological Products, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Sompong Sapsutthipas
- Institute of Biological Products, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Sakalin Trisiriwanich
- Institute of Biological Products, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Thitiporn Somsaard
- Institute of Biological Products, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology for Development Agency, Pathumthani, Thailand
| | - Parichat Duangkhae
- Viral Vaccine Unit, Biologics Research Group, Research and Development Institute, The Government Pharmaceutical Organization, Bangkok, Thailand
| | - Boonsri Ongpipattanakul
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Patanachai Limpikirati
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Natapol Pornputtapong
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Wanatchaporn Arunmanee
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
24
|
Brzuska G, Zimna M, Baranska K, Szewczyk B, Strakova P, Ruzek D, Krol E. The Influence of Adjuvant Type on the Immunogenicity of RBD/N Cocktail Antigens as a Vaccine Candidate against SARS-CoV-2 Virus. Microbiol Spectr 2023; 11:e0256422. [PMID: 37199661 PMCID: PMC10269882 DOI: 10.1128/spectrum.02564-22] [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: 07/06/2022] [Accepted: 04/27/2023] [Indexed: 05/19/2023] Open
Abstract
The emerging virus SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2 virus), agent of COVID-19, appeared in December 2019 in Wuhan, China, and became a serious threat to global health and public safety. Many COVID-19 vaccines have been approved and licensed around the world. Most of the developed vaccines include S protein and induce an antibody-based immune response. Additionally, T-cell response to the SARS-CoV-2 antigens could be beneficial for combating the infection. The type of immune response is greatly dependent not only on the antigen, but also on adjuvants used in vaccine formulation. Here, we compared the effect of four different adjuvants (AddaS03, Alhydrogel/MPLA, Alhydrogel/ODN2395, Quil A) on the immunogenicity of a mixture of recombinant RBD and N SARS-CoV-2 proteins. We have analyzed the antibody and T-cell response specific to RBD and N proteins and assessed the impact of adjuvants on virus neutralization. Our results clearly indicated that Alhydrogel/MPLA and Alhydrogel/ODN2395 adjuvants elicited the higher titers of specific and cross-reactive antibodies to S protein variants from various SARS-CoV-2 and SARS-CoV-1 strains. Moreover, Alhydrogel/ODN2395 stimulated high cellular response to both antigens, as assessed by IFN-γ production. Importantly, sera collected from mice immunized with RBD/N cocktail in combination with these adjuvants exhibited neutralizing activity against the authentic SARS-CoV-2 virus as well as particles pseudotyped with S protein from various virus variants. The results from our study demonstrate the immunogenic potential of RBD and N antigens and point out the importance of adjuvants selection in vaccine formulation in order to enhance the immunological response. IMPORTANCE Although several COVID-19 vaccines have been approved worldwide, continuous emergence of new SARS-CoV-2 variants calls for new efficient vaccines against them, providing long-lasting immunity. As the immune response after vaccination is dependent not only on antigen used, but also on other vaccine components, e.g., adjuvants, the purpose of this work was to study the effect of different adjuvants on the immunogenicity of RBD/N SARS-CoV-2 cocktail proteins. In this work, it has been shown that immunization with both antigens plus the different adjuvants studied elicited higher Th1 and Th2 responses against RBD and N, which contributed to higher neutralization of the virus. The obtained results can be used for design of new vaccines, not only against SARS-CoV-2, but also against other important viral pathogens.
Collapse
Affiliation(s)
- Gabriela Brzuska
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Marta Zimna
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Klaudia Baranska
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Boguslaw Szewczyk
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Petra Strakova
- Laboratory of Emerging Viral Infections, Veterinary Research Institute, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Daniel Ruzek
- Laboratory of Emerging Viral Infections, Veterinary Research Institute, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Ewelina Krol
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
25
|
Gonzalez-Hernandez M, Kaiser FK, Steffen I, Ciurkiewicz M, van Amerongen G, Tchelet R, Emalfarb M, Saloheimo M, Wiebe MG, Vitikainen M, Albulescu IC, Bosch BJ, Baumgärtner W, Haagmans BL, Osterhaus ADME. Preclinical immunogenicity and protective efficacy of a SARS-CoV-2 RBD-based vaccine produced with the thermophilic filamentous fungal expression system Thermothelomyces heterothallica C1. Front Immunol 2023; 14:1204834. [PMID: 37359531 PMCID: PMC10289020 DOI: 10.3389/fimmu.2023.1204834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction The emergency use of vaccines has been the most efficient way to control the coronavirus disease 19 (COVID-19) pandemic. However, the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern has reduced the efficacy of currently used vaccines. The receptor-binding domain (RBD) of the SARS-CoV-2 spike (S) protein is the main target for virus neutralizing (VN) antibodies. Methods A SARS-CoV-2 RBD vaccine candidate was produced in the Thermothelomyces heterothallica (formerly, Myceliophthora thermophila) C1 protein expression system and coupled to a nanoparticle. Immunogenicity and efficacy of this vaccine candidate was tested using the Syrian golden hamster (Mesocricetus auratus) infection model. Results One dose of 10-μg RBD vaccine based on SARS-CoV-2 Wuhan strain, coupled to a nanoparticle in combination with aluminum hydroxide as adjuvant, efficiently induced VN antibodies and reduced viral load and lung damage upon SARS-CoV-2 challenge infection. The VN antibodies neutralized SARS-CoV-2 variants of concern: D614G, Alpha, Beta, Gamma, and Delta. Discussion Our results support the use of the Thermothelomyces heterothallica C1 protein expression system to produce recombinant vaccines against SARS-CoV-2 and other virus infections to help overcome limitations associated with the use of mammalian expression system.
Collapse
Affiliation(s)
- Mariana Gonzalez-Hernandez
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Franziska Karola Kaiser
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Imke Steffen
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Institute for Biochemistry, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | | | - Ronen Tchelet
- Dyadic International, Inc., Jupiter, FL, United States
| | - Mark Emalfarb
- Dyadic International, Inc., Jupiter, FL, United States
| | | | | | | | - Irina C. Albulescu
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Berend-Jan Bosch
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Bart L. Haagmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Albert D. M. E. Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
26
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringuist R, Smith C, Ochoa MA, Roy K. A multiadjuvant polysaccharide-amino acid-lipid (PAL) subunit nanovaccine generates robust systemic and lung-specific mucosal immune responses against SARS-CoV-2 in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539395. [PMID: 37215018 PMCID: PMC10197586 DOI: 10.1101/2023.05.05.539395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, which are essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters could overcome the shortcomings of parenteral vaccines and enhance pre- existing systemic immunity. Here we present a new protein subunit nanovaccine using multiadjuvanted (e.g. RIG-I: PUUC, TLR9: CpG) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL- NPs, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lung, and showed robust systemic humoral immunity. Interestingly, as a purely intranasal vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. Our data suggest that PUUC+CpG PAL-NP subunit vaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
|
27
|
Love J, Rodriguez-Aponte S, Tostanoski L, Dalvie N, Johnston R, Jacob-Dolan C, Powers O, Hachmann N, Miller J, Hall K, Siamatu M, Mazurek C, Surve N, Barouch D. SARS-CoV-2 RBD dimers elicit response comparable to VLPs in mice. RESEARCH SQUARE 2023:rs.3.rs-2692315. [PMID: 37163131 PMCID: PMC10168475 DOI: 10.21203/rs.3.rs-2692315/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
We report the direct comparison of monomeric, dimeric and trimeric RBD protein subunit vaccines to a virus-like particle (VLP) displaying RBD. After two and three doses, a RBD dimer and trimer elicited antibody levels in mice comparable to an RBD-VLP. Furthermore, an Omicron (BA.1) RBD hetero-dimer induced neutralizing activity similar to the RBD-VLP. A RBD hetero-dimer and RBD-VLP also shows comparable breadth to other SARS-CoV-2 variants-of-concern (VOCs).
Collapse
|
28
|
Volosnikova EA, Merkuleva IA, Esina TI, Shcherbakov DN, Borgoyakova MB, Isaeva AA, Nesmeyanova VS, Volkova NV, Belenkaya SV, Zaykovskaya AV, Pyankov OV, Starostina EV, Zadorozhny AM, Zaitsev BN, Karpenko LI, Ilyichev AA, Danilenko ED. SARS-CoV-2 RBD Conjugated to Polyglucin, Spermidine, and dsRNA Elicits a Strong Immune Response in Mice. Vaccines (Basel) 2023; 11:vaccines11040808. [PMID: 37112720 PMCID: PMC10146165 DOI: 10.3390/vaccines11040808] [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: 03/05/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the rapid development and approval of several COVID vaccines based on the full-length spike protein, there is a need for safe, potent, and high-volume vaccines. Considering the predominance of the production of neutralizing antibodies targeting the receptor-binding domain (RBD) of S-protein after natural infection or vaccination, it makes sense to choose RBD as a vaccine immunogen. However, due to its small size, RBD exhibits relatively poor immunogenicity. Searching for novel adjuvants for RBD-based vaccine formulations is considered a good strategy for enhancing its immunogenicity. Herein, we assess the immunogenicity of severe acute respiratory syndrome coronavirus 2 RBD conjugated to a polyglucin:spermidine complex (PGS) and dsRNA (RBD-PGS + dsRNA) in a mouse model. BALB/c mice were immunized intramuscularly twice, with a 2-week interval, with 50 µg of RBD, RBD with Al(OH)3, or conjugated RBD. A comparative analysis of serum RBD-specific IgG and neutralizing antibody titers showed that PGS, PGS + dsRNA, and Al(OH)3 enhanced the specific humoral response in animals. There was no significant difference between the groups immunized with RBD-PGS + dsRNA and RBD with Al(OH)3. Additionally, the study of the T-cell response in animals showed that, unlike adjuvants, the RBD-PGS + dsRNA conjugate stimulates the production of specific CD4+ and CD8+ T cells in animals.
Collapse
Affiliation(s)
- Ekaterina A Volosnikova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Iuliia A Merkuleva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Tatiana I Esina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Dmitry N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Mariya B Borgoyakova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anastasiya A Isaeva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Valentina S Nesmeyanova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Natalia V Volkova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Svetlana V Belenkaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anna V Zaykovskaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Oleg V Pyankov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Ekaterina V Starostina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexey M Zadorozhny
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Boris N Zaitsev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Larisa I Karpenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexander A Ilyichev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Elena D Danilenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| |
Collapse
|
29
|
Oktay E, Alem F, Hernandez K, Girgis M, Green C, Mathur D, Medintz IL, Narayanan A, Veneziano R. DNA origami presenting the receptor binding domain of SARS-CoV-2 elicit robust protective immune response. Commun Biol 2023; 6:308. [PMID: 36959304 PMCID: PMC10034259 DOI: 10.1038/s42003-023-04689-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/10/2023] [Indexed: 03/25/2023] Open
Abstract
Effective and safe vaccines are invaluable tools in the arsenal to fight infectious diseases. The rapid spreading of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) responsible for the coronavirus disease 2019 pandemic has highlighted the need to develop methods for rapid and efficient vaccine development. DNA origami nanoparticles (DNA-NPs) presenting multiple antigens in prescribed nanoscale patterns have recently emerged as a safe, efficient, and easily scalable alternative for rational design of vaccines. Here, we are leveraging the unique properties of these DNA-NPs and demonstrate that precisely patterning ten copies of a reconstituted trimer of the receptor binding domain (RBD) of SARS-CoV-2 along with CpG adjuvants on the DNA-NPs is able to elicit a robust protective immunity against SARS-CoV-2 in a mouse model. Our results demonstrate the potential of our DNA-NP-based approach for developing safe and effective nanovaccines against infectious diseases with prolonged antibody response and effective protection in the context of a viral challenge.
Collapse
Affiliation(s)
- Esra Oktay
- Department of Bioengineering, George Mason University, Fairfax, VA, 22030, USA
| | - Farhang Alem
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA, 20110, USA
| | - Keziah Hernandez
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA, 20110, USA
| | - Michael Girgis
- Department of Bioengineering, George Mason University, Fairfax, VA, 22030, USA
| | - Christopher Green
- Center for Bio/Molecular Science and Engineering Code 6900, U.S. Naval Research Laboratory, Washington, DC, USA
| | - Divita Mathur
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Igor L Medintz
- Center for Bio/Molecular Science and Engineering Code 6900, U.S. Naval Research Laboratory, Washington, DC, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA, 20110, USA.
| | - Remi Veneziano
- Department of Bioengineering, George Mason University, Fairfax, VA, 22030, USA.
| |
Collapse
|
30
|
Phoolcharoen W, Shanmugaraj B, Khorattanakulchai N, Sunyakumthorn P, Pichyangkul S, Taepavarapruk P, Praserthsee W, Malaivijitnond S, Manopwisedjaroen S, Thitithanyanont A, Srisutthisamphan K, Jongkaewwattana A, Tomai M, Fox CB, Taychakhoonavudh S. Preclinical evaluation of immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052-Alum adjuvant. Vaccine 2023; 41:2781-2792. [PMID: 36963999 PMCID: PMC10027959 DOI: 10.1016/j.vaccine.2023.03.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023]
Abstract
Cost-effective, and accessible vaccines are needed for mass immunization to control the ongoing coronavirus disease 2019 (COVID-19), especially in low- and middle-income countries (LMIC).A plant-based vaccine is an attractive technology platform since the recombinant proteins can be easily produced at large scale and low cost. For the recombinant subunit-based vaccines, effective adjuvants are crucial to enhance the magnitude and breadth of immune responses elicited by the vaccine. In this study, we report a preclinical evaluation of the immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052 (TLR7/8 agonist)-Alum adjuvant. This vaccine formulation, named Baiya SARS-CoV-2 Vax 2, induced significant levels of RBD-specific IgG and neutralizing antibody responses in mice. A viral challenge study using humanized K18-hACE2 mice has shown that animals vaccinated with two doses of Baiya SARS-CoV-2 Vax 2 established immune protection against SARS-CoV-2. A study in nonhuman primates (cynomolgus monkeys) indicated that immunization with two doses of Baiya SARS-CoV-2 Vax 2 was safe, well tolerated, and induced neutralizing antibodies against the prototype virus and other viral variants (Alpha, Beta, Gamma, Delta, and Omicron subvariants). The toxicity of Baiya SARS-CoV-2 Vax 2 was further investigated in Jcl:SD rats, which demonstrated that a single dose and repeated doses of Baiya SARS-CoV-2 Vax 2 were well tolerated and no mortality or unanticipated findings were observed. Overall, these preclinical findings support further clinical development of Baiya SARS-CoV-2 Vax 2.
Collapse
Affiliation(s)
- Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | | | - Narach Khorattanakulchai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Sathit Pichyangkul
- US Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Pornnarin Taepavarapruk
- Center for Animal Research and Department of Physiology, Faculty of Medical Science, Naresuan University, Pitsanulok 65000, Thailand
| | | | - Suchinda Malaivijitnond
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | | | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Mark Tomai
- 3M Healthcare, 3M Center, Bldg 270-4N-04, St. Paul, MN 55144-1000, USA
| | - Christopher B Fox
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Ste 400, Seattle, WA 98102, USA
| | - Suthira Taychakhoonavudh
- Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
31
|
Milligan EC, Olstad K, Williams CA, Mallory M, Cano P, Cross KA, Munt JE, Garrido C, Lindesmith L, Watanabe J, Usachenko JL, Hopkins L, Immareddy R, Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Sammak RL, Pollard RE, Yee JL, Herbek S, Scobey T, Miehlke D, Fouda G, Ferrari G, Gao H, Shen X, Kozlowski PA, Montefiori D, Hudgens MG, Edwards DK, Carfi A, Corbett KS, Graham BS, Fox CB, Tomai M, Iyer SS, Baric R, Reader R, Dittmer DP, Van Rompay KKA, Permar SR, De Paris K. Infant rhesus macaques immunized against SARS-CoV-2 are protected against heterologous virus challenge 1 year later. Sci Transl Med 2023; 15:eadd6383. [PMID: 36454813 PMCID: PMC9765459 DOI: 10.1126/scitranslmed.add6383] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The U.S. Food and Drug Administration only gave emergency use authorization of the BNT162b2 and mRNA-1273 SARS-CoV-2 vaccines for infants 6 months and older in June 2022. Yet questions regarding the durability of vaccine efficacy, especially against emerging variants, in this age group remain. We demonstrated previously that a two-dose regimen of stabilized prefusion Washington SARS-CoV-2 S-2P spike (S) protein encoded by mRNA encapsulated in lipid nanoparticles (mRNA-LNP) or purified S-2P mixed with 3M-052, a synthetic Toll-like receptor (TLR) 7/8 agonist, in a squalene emulsion (Protein+3M-052-SE) was safe and immunogenic in infant rhesus macaques. Here, we demonstrate that broadly neutralizing and spike-binding antibodies against variants of concern (VOCs), as well as T cell responses, persisted for 12 months. At 1 year, corresponding to human toddler age, we challenged vaccinated rhesus macaques and age-matched nonvaccinated controls intranasally and intratracheally with a high dose of heterologous SARS-CoV-2 B.1.617.2 (Delta). Seven of eight control rhesus macaques exhibited severe interstitial pneumonia and high virus replication in the upper and lower respiratory tract. In contrast, vaccinated rhesus macaques had faster viral clearance with mild to no pneumonia. Neutralizing and binding antibody responses to the B.1.617.2 variant at the day of challenge correlated with lung pathology and reduced virus replication. Overall, the Protein+3M-052-SE vaccine provided superior protection to the mRNA-LNP vaccine, emphasizing opportunities for optimization of current vaccine platforms. The observed efficacy of both vaccines 1 year after vaccination supports the implementation of an early-life SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Emma C Milligan
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine Olstad
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael Mallory
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patricio Cano
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaitlyn A Cross
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer E Munt
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carolina Garrido
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Lisa Lindesmith
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Jodie L Usachenko
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Lincoln Hopkins
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Ramya Immareddy
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | | | - Sonny R Elizaldi
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Rebecca L Sammak
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Rachel E Pollard
- School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, USA
| | - JoAnn L Yee
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Trevor Scobey
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dieter Miehlke
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Genevieve Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Hongmei Gao
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael G Hudgens
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | - Kizzmekia S Corbett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Christopher B Fox
- Access to Advanced Health Institute, Seattle, WA 98102, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Mark Tomai
- 3M Corporate Research Materials Laboratory, Saint Paul, MN 55144, USA
| | - Smita S Iyer
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Ralph Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel Reader
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Department of Pathology, Microbiology and Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
32
|
Du J, Wu G, Chen Q, Yu C, Xu G, Liu A, Wang L. Fingerprinting trimeric SARS-CoV-2 RBD by capillary isoelectric focusing with whole-column imaging detection. Anal Biochem 2023; 663:115034. [PMID: 36586502 PMCID: PMC9794521 DOI: 10.1016/j.ab.2022.115034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
Because the spike (S) protein of the severe acute respiratory syndrome coronavirus (SARS-CoV) is the immunodominant antigen, the S protein and its receptor-binding domain (RBD) are both targets currently to be genetically engineered for designing the broad-spectrum vaccine. In theory, the expressed protein exists as a set of variants that are roughly the same but slightly different, which depends on the protein expression system. The variants can be phenotypically manifested as charge heterogeneity. Here, we attempted to depict the charge heterogeneity of the trimeric SARS-CoV-2 RBD by using capillary isoelectric focusing with whole-column imaging detection (cIEF-WCID). In its nature form, the electropherogram fingerprints of the trimeric RBD were presented under optimized experimental conditions. The peaks of matrix buffers can be fully distinguishable from peaks of trimeric RBD. The isoelectric point (pI) was determined to be within a range of 6.67-9.54 covering the theoretical pI of 9.02. The fingerprints of three batches of trimeric RBDs are completely the same, with the intra-batch and batch-to-batch relative standard deviations (RSDs) of both pI values and area percentage of each peak no more than 1.0%, indicating that the production process is stable and this method can be used to surveillance the batch-to-batch consistency. The fingerprint remained unchanged after incubating at 37 °C for 7 d and oxidizing by 0.015% H2O2. In addition, the fingerprint was destroyed when adjusting the pH value to higher than 10.0 but still stable when the pH was lower than 4.0. In summary, the cIEF-WCID fingerprint can be used for the identification, batch-to-batch consistency evaluation, and stability study of the trimeric SARS-CoV-2 RBD, as part of a quality control strategy during the potential vaccine production.
Collapse
Affiliation(s)
- Jialiang Du
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, 102629, China
| | - Gang Wu
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, 102629, China
| | - Quanyao Chen
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, 102629, China,School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Chuanfei Yu
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, 102629, China
| | - Gangling Xu
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, 102629, China
| | - Anhui Liu
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Lan Wang
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, 102629, China.
| |
Collapse
|
33
|
Wahba MA, Mofed D, Ghareeb DA, Omran JI, Salem TZ. Baculovirus displaying SARS-CoV-2 spike RBD promotes neutralizing antibody production in a mouse model. J Genet Eng Biotechnol 2023; 21:16. [PMID: 36759349 PMCID: PMC9910779 DOI: 10.1186/s43141-023-00472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND There is always a need for a safe and efficient vaccine platform, especially when facing a pandemic such as COVID-19. Most of the SARS-CoV-2-based vaccines are based on the full spike protein, which is presented as a trimerized protein, and many viral vector vaccines express the spike protein into the host cells and do not display it on virus surfaces. However, the spike receptor-binding domain (RBD)-based vaccines are efficient and are currently under investigation and clinical trials. METHODOLOGY In this study, we are testing the efficacy of the RBD displayed on a baculovirus as a mean to formulate a safe and stable carrier to induce the immune system against SARS-CoV-2. Therefore, two pseudotyped baculoviruses were constructed to display the RBD, AcRBD-sfGFP-64, and AcRBD-sfGFP-V, using two different displaying strategies based on gp64 and VSV-G envelope glycoproteins, from Autographa californica multiple nucleopolyhedrovirus (AcMNPV) and vesicular stomatitis virus (VSV), respectively. BALB/C mice were immunized with the pseudotyped baculoviruses in a dose-optimized manner. Dot blot and Western blot were used to screen and validate the polyclonal antibodies' specificity to the SARS-CoV-2 RBD. A plaque reduction neutralization test (PRNT) was used to measure the sera neutralization capacity against a SARS-CoV-2 wild-type isolate from Egypt. ELISA was used to quantify certain cytokines for the assessment of the immune response. RESULT The outcome of our investigation showed that the monomeric RBD proteins were properly displayed on baculovirus and efficiently triggered the mouse immune system. The produced sera efficiently neutralized about 50% of SARS-CoV-2 in more than 100-fold serum dilution. The immunized mice showed a significant increase (p<0.01) in the levels of IL-2 and IFN-γ and a significant decrease (p<0.01) and (p<0.001) in the levels of IL-4 and IL-10, respectively, which suggest that AcRBD-sfGFP-64 and AcRBD-sfGFP-V induce Th1 cellular immune response. CONCLUSION The produced recombinant viruses can induce the immune response without adjuvant, which needs dose optimization and further stability tests. Neutralizing antibodies were induced without affecting the health of immunized mice. Th1 response can be attainable through the system, which is of great benefit in SARS CoV-2 infection and the system can be tested for future applications including vaccine development and polyclonal antibody production.
Collapse
Affiliation(s)
- Mohamed A. Wahba
- grid.440881.10000 0004 0576 5483Molecular Biology and Virology lab, Biomedical Sciences program, UST, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza 12578 Egypt
| | - Dina Mofed
- grid.440881.10000 0004 0576 5483Molecular Biology and Virology lab, Biomedical Sciences program, UST, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza 12578 Egypt
| | - Doaa A. Ghareeb
- grid.7155.60000 0001 2260 6941Bio-screening and preclinical trial lab, Biochemistry Department, Faculty of Science, Alexandria University, P.O. Box 21511, Alexandria, Egypt
| | - Jihad I. Omran
- grid.440881.10000 0004 0576 5483Molecular Biology and Virology lab, Biomedical Sciences program, UST, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza 12578 Egypt
| | - Tamer Z. Salem
- grid.440881.10000 0004 0576 5483Molecular Biology and Virology lab, Biomedical Sciences program, UST, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza 12578 Egypt ,grid.482515.f0000 0004 7553 2175Department of Microbial Genetics, Agricultural Genetic Engineering Research Institute (AGERl), ARC, Giza, 12619 Egypt ,National Biotechnology Network of Expertise (NBNE), Academy of Science Research and Technology (ASRT), Cairo, 11334 Egypt
| |
Collapse
|
34
|
Wen D, Liang T, Chen G, Li H, Wang Z, Wang J, Fu R, Han X, Ci T, Zhang Y, Abdou P, Li R, Bu L, Dotti G, Gu Z. Adipocytes Encapsulating Telratolimod Recruit and Polarize Tumor-Associated Macrophages for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206001. [PMID: 36526596 PMCID: PMC9929126 DOI: 10.1002/advs.202206001] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/06/2022] [Indexed: 05/09/2023]
Abstract
Tumor-associated adipocytes (TAAs) recruit monocytes and promote their differentiation into tumor-associated macrophages (TAMs) that support tumor development. Here, TAAs are engineered to promote the polarization of TAMs to the tumor suppressive M1 phenotype. Telratolimod, a toll-like receptor 7/8 agonist, is loaded into the lipid droplets of adipocytes to be released at the tumor site upon tumor cell-triggered lipolysis. Locally administered drug-loaded adipocytes increased tumor suppressive M1 macrophages in both primary and distant tumors and suppressed tumor growth in a melanoma model. Furthermore, drug-loaded adipocytes improved CD8+ T cell-mediated immune responses within the tumor microenvironment and favored dendritic cell maturation in the tumor draining lymph nodes.
Collapse
Affiliation(s)
- Di Wen
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
- Earle A. Chiles Research InstituteRobert W. Franz Cancer CenterProvidence Portland Medical CenterPortlandOregon97213USA
| | - Tingxizi Liang
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang ProvinceCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Guojun Chen
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Hongjun Li
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang ProvinceCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Zejun Wang
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Jinqiang Wang
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang ProvinceCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Ruxing Fu
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Xiao Han
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Tianyuan Ci
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Yuqi Zhang
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang ProvinceCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Peter Abdou
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Ruoxin Li
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Linlin Bu
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
| | - Gianpietro Dotti
- Department of Microbiology and ImmunologySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - Zhen Gu
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang ProvinceCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Jinhua Institute of Zhejiang UniversityJinhua321299P. R. China
- Department of General SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| |
Collapse
|
35
|
Qin S, Huang H, Xiao W, Chen K, He X, Tang X, Huang Z, Zhang Y, Duan X, Fan N, Zheng Q, Wu M, Lu G, Wei Y, Wei X, Song X. A novel heterologous receptor-binding domain dodecamer universal mRNA vaccine against SARS-CoV-2 variants. Acta Pharm Sin B 2023; 13:S2211-3835(23)00010-2. [PMID: 36647424 PMCID: PMC9833852 DOI: 10.1016/j.apsb.2023.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/16/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
There are currently approximately 4,000 mutations in the SARS-CoV-2 S protein gene and emerging SARS-CoV-2 variants continue to spread rapidly worldwide. Universal vaccines with high efficacy and safety urgently need to be developed to prevent SARS-CoV-2 variants pandemic. Here, we described a novel self-assembling universal mRNA vaccine containing a heterologous receptor-binding domain (HRBD)-based dodecamer (HRBDdodecamer) against SARS-CoV-2 variants, including Alpha (B.1.1.7), Beta (B.1.351), Gamma (B.1.1.28.1), Delta (B.1.617.2) and Omicron (B.1.1.529). HRBD containing four heterologous RBD (Delta, Beta, Gamma, and Wild-type) can form a stable dodecameric conformation under T4 trimerization tag (Flodon, FD). The HRBDdodecamer -encoding mRNA was then encapsulated into the newly-constructed LNPs consisting of a novel ionizable lipid (4N4T). The obtained universal mRNA vaccine (4N4T-HRBDdodecamer) presented higher efficiency in mRNA transfection and expression than the approved ALC-0315 LNPs, initiating potent immune protection against the immune escape of SARS-CoV-2 caused by evolutionary mutation. These findings demonstrated the first evidence that structure-based antigen design and mRNA delivery carrier optimization may facilitate the development of effective universal mRNA vaccines to tackle SARS-CoV-2 variants pandemic.
Collapse
Affiliation(s)
| | | | | | | | - Xi He
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoshan Tang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiying Huang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yupei Zhang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xing Duan
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Fan
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qian Zheng
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Wu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guangwen Lu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiawei Wei
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
36
|
Huang H, Zhou Z, Xiong X, Liu Z, Zheng X, Quan Q, Yu M. SARS-CoV-2 delta (B.1.617.2) spike protein adjuvanted with Alum-3M-052 enhances antibody production and neutralization ability. Front Public Health 2023; 10:976686. [PMID: 36684881 PMCID: PMC9853420 DOI: 10.3389/fpubh.2022.976686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
Background Optimizing adjuvant is one of the critical methods to improve the vaccine. 3M-052, a novel TLR7/8 agonist which was designed for slow dissemination at the injection site, has a potential as adjuvant, but its performance as a vaccine adjuvant for SARS-CoV-2 (B.1.617.2) spike protein has not been studied. The present study aimed to evaluate the effect of Alum-3M-052 as an adjuvant to improve mice serum antibody titers and pseudovirus neutralization efficiency. Method Female Balb/c mice were immunized 3 times at day 0, 7 and 21 intramuscularly with SARS-CoV-2 (B.1.617.2) spike protein and adjuvant (Alum or Alum-3M-052). Mice serum was collected weekly since day 7. Antibody titers of mice serum anti-SARS-CoV-2 (B.1.617.2) IgG and IgM were detected by ELISA. Inhibition rates of mice serum blocking SARS-CoV-2 (B.1.617.2) spike protein binding to ACE2 were detected by SARS-CoV-2 (B.1.617.2) Inhibitor Screening Kit. Neutralization efficiencies of mice serum against both SARS-CoV-2 (BA.2.12.1) pseudovirus and SARS-CoV-2 (B.1.617.2) pseudovirus were detected by pseudovirus neutralizing assay. Result Serum of mice immunized by SARS-CoV-2 (B.1.617.2) spike protein adjuvanted with Alum-3M-052 had highest antibody titers and higher neutralization efficiency against both SARS-CoV-2 (BA.2.12.1) pseudovirus and SARS-CoV-2 (B.1.617.2) pseudovirus. Besides, neutralization efficiency of anti-SARS-CoV-2 (B.1.617.2) spike protein antibody against SARS-CoV-2 (BA.2.12.1) pseudovirus was lower than that of SARS-CoV-2 (B.1.617.2) pseudovirus. Conclusion Alum-3M-052 rapidly increased the titer of anti-SARS-CoV-2 (B.1.617.2) spike protein neutralizing antibodies and enhanced the neutralization ability against pseudoviruses and variants. This study provided evidence for the application of Alum-3M-052 as an adjuvant in COVID-19 vaccines production.
Collapse
Affiliation(s)
- Hong Huang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhongcheng Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xinxin Xiong
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhihai Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoxue Zheng
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qingli Quan
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - Meixing Yu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
37
|
Guan X, Yang Y, Du L. Advances in SARS-CoV-2 receptor-binding domain-based COVID-19 vaccines. Expert Rev Vaccines 2023; 22:422-439. [PMID: 37161869 PMCID: PMC10355161 DOI: 10.1080/14760584.2023.2211153] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
INTRODUCTION The Coronavirus Disease 2019 (COVID-19) pandemic has caused devastating human and economic costs. Vaccination is an important step in controlling the pandemic. Severe acute respiratory coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19, infects cells by binding a cellular receptor through the receptor-binding domain (RBD) within the S1 subunit of the spike (S) protein. Viral entry and membrane fusion are mediated by the S2 subunit. AREAS COVERED SARS-CoV-2 S protein, particularly RBD, serves as an important target for vaccines. Here we review the structure and function of SARS-CoV-2 S protein and its RBD, summarize current COVID-19 vaccines targeting the RBD, and outline potential strategies for improving RBD-based vaccines. Overall, this review provides important information that will facilitate rational design and development of safer and more effective COVID-19 vaccines. EXPERT OPINION The S protein of SARS-CoV-2 harbors numerous mutations, mostly in the RBD, resulting in multiple variant strains. Although many COVID-19 vaccines targeting the RBD of original virus strain (and previous variants) can prevent infection of these strains, their ability against recent dominant variants, particularly Omicron and its offspring, is significantly reduced. Collective efforts are needed to develop effective broad-spectrum vaccines to control current and future variants that have pandemic potential.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
38
|
Zhao F, Zai X, Zhang Z, Xu J, Chen W. Challenges and developments in universal vaccine design against SARS-CoV-2 variants. NPJ Vaccines 2022; 7:167. [PMID: 36535982 PMCID: PMC9761649 DOI: 10.1038/s41541-022-00597-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had become a global concern because of its unexpectedly high pathogenicity and transmissibility. SARS-CoV-2 variants that reduce the immune protection elicited from previous vaccination or natural infection raise challenges in controlling the spread of the pandemic. The development of universal vaccines against these variants seems to be a practical solution to alleviate the physical and economic effects caused by this disease, but it is hard to achieve. In this review, we describe the high mutation rate of RNA viruses and dynamic molecular structures of SARS-CoV-2 variants in several major neutralizing epitopes, trying to answer the question of why universal vaccines are difficult to design. Understanding the biological basis of immune evasion is crucial for combating these obstacles. We then summarize several advancements worthy of further study, including heterologous prime-boost regimens, construction of chimeric immunogens, design of protein nanoparticle antigens, and utilization of conserved neutralizing epitopes. The fact that some immunogens can induce cross-reactive immune responses against heterologous coronaviruses provides hints for universal vaccine development. We hope this review can provide inspiration to current universal vaccine studies.
Collapse
Affiliation(s)
- Fangxin Zhao
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiaodong Zai
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China
| | - Zhiling Zhang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China.
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China.
| |
Collapse
|
39
|
Adam A, Shi Q, Wang B, Zou J, Mai J, Osman SR, Wu W, Xie X, Aguilar PV, Bao X, Shi PY, Shen H, Wang T. A modified porous silicon microparticle potentiates protective systemic and mucosal immunity for SARS-CoV-2 subunit vaccine. Transl Res 2022; 249:13-27. [PMID: 35688318 PMCID: PMC9173827 DOI: 10.1016/j.trsl.2022.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 11/28/2022]
Abstract
Development of optimal SARS-CoV-2 vaccines to induce potent, long-lasting immunity and provide cross-reactive protection against emerging variants remains a high priority. Here, we report that a modified porous silicon microparticle (mPSM) adjuvant to SARS-CoV-2 receptor-binding domain (RBD) vaccine activated dendritic cells and generated more potent and durable systemic humoral and type 1 helper T (Th) cell- mediated immune responses than alum-formulated RBD following parenteral vaccination, and protected mice from SARS-CoV-2 and Beta variant challenge. Notably, mPSM facilitated the uptake of SARS-CoV-2 RBD antigens by nasal and airway epithelial cells. Parenteral and intranasal prime and boost vaccinations with mPSM-RBD elicited stronger lung resident T and B cells and IgA responses compared to parenteral vaccination alone, which led to markedly diminished viral loads and inflammation in the lung following SARS-CoV-2 Delta variant challenge. Overall, our results suggest that mPSM is effective adjuvant for SARS-CoV-2 subunit vaccine in both systemic and mucosal vaccinations.
Collapse
Key Words
- mpsm, modified porous silicon microparticle
- covid-19, coronavirus disease 2019
- rbd, receptor-binding domain
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
- β-cov, betacoronavirus
- e, envelope
- m, membrane
- n, nucleocapsid
- hace2, human angiotensin-converting enzyme 2
- nabs, neutralizing antibodies
- dc, dendritic cell
- th1, t helper 1
- cpg, cytosine guanosine dinucleotide
- cgamp, cyclic gamp
- bm, bone marrow
- i.p., intraperitoneally
- i.d., intradermally
- i.m., or intramuscularly
- tmb, tetramethylbenzidine
- pbs-t, phosphate-buffered saline containing tween-20
- bal, bronchoalveolar lavage
- hrp, horseradish peroxidase
- elisa, enzyme-linked immunosorbent assay
- elispot, enzyme-linked immune absorbent spot
- sfc, spot-forming cells
- ics, intracellular cytokine staining
- moi, multiplicity of infection
- apc, antigen presenting cells
- mbc, memory b cell
- asc, antibody secreting cells
- prnt, plaque reduction neutralization test
- saec, small airway epithelial cells
- nalt, nasal-associated lymphoid tissue
- ade, antibody-dependent enhancement
- q-pcr, quantitative pcr
Collapse
Affiliation(s)
- Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Qing Shi
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, Texas
| | - Binbin Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Jing Zou
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, Texas
| | - Samantha R Osman
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas
| | - Xuping Xie
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas
| | - Patricia V Aguilar
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, Texas; Innovative Therapeutic Program, Houston Methodist Cancer Center, Houston, Texas; ImmunoQ Therapeutics, Houston, Texas.
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Pathology, University of Texas Medical Branch, Galveston, Texas; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
40
|
Luo S, Zhang J, Kreutzberger AJ, Eaton A, Edwards RJ, Jing C, Dai HQ, Sempowski GD, Cronin K, Parks R, Ye AY, Mansouri K, Barr M, Pishesha N, Williams AC, Vieira Francisco L, Saminathan A, Peng H, Batra H, Bellusci L, Khurana S, Alam SM, Montefiori DC, Saunders KO, Tian M, Ploegh H, Kirchhausen T, Chen B, Haynes BF, Alt FW. An antibody from single human V H-rearranging mouse neutralizes all SARS-CoV-2 variants through BA.5 by inhibiting membrane fusion. Sci Immunol 2022; 7:eadd5446. [PMID: 35951767 PMCID: PMC9407951 DOI: 10.1126/sciimmunol.add5446] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/03/2022] [Indexed: 12/14/2022]
Abstract
SARS-CoV-2 Omicron subvariants have generated a worldwide health crisis due to resistance to most approved SARS-CoV-2 neutralizing antibodies and evasion of vaccination-induced antibodies. To manage Omicron subvariants and prepare for new ones, additional means of isolating broad and potent humanized SARS-CoV-2 neutralizing antibodies are desirable. Here, we describe a mouse model in which the primary B cell receptor (BCR) repertoire is generated solely through V(D)J recombination of a human VH1-2 heavy chain (HC) and, substantially, a human Vκ1-33 light chain (LC). Thus, primary humanized BCR repertoire diversity in these mice derives from immensely diverse HC and LC antigen-contact CDR3 sequences generated by nontemplated junctional modifications during V(D)J recombination. Immunizing this mouse model with SARS-CoV-2 (Wuhan-Hu-1) spike protein immunogens elicited several VH1-2/Vκ1-33-based neutralizing antibodies that bound RBD in a different mode from each other and from those of many prior patient-derived VH1-2-based neutralizing antibodies. Of these, SP1-77 potently and broadly neutralized all SARS-CoV-2 variants through BA.5. Cryo-EM studies revealed that SP1-77 bound RBD away from the receptor-binding motif via a CDR3-dominated recognition mode. Lattice light-sheet microscopy-based studies showed that SP1-77 did not block ACE2-mediated viral attachment or endocytosis but rather blocked viral-host membrane fusion. The broad and potent SP1-77 neutralization activity and nontraditional mechanism of action suggest that it might have therapeutic potential. Likewise, the SP1-77 binding epitope may inform vaccine strategies. Last, the type of humanized mouse models that we have described may contribute to identifying therapeutic antibodies against future SARS-CoV-2 variants and other pathogens.
Collapse
Affiliation(s)
- Sai Luo
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Zhang
- Division of Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Alex J.B. Kreutzberger
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Amanda Eaton
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert J. Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Changbin Jing
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Hai-Qiang Dai
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth Cronin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Adam Yongxin Ye
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Aimee Chapdelaine Williams
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Lucas Vieira Francisco
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Anand Saminathan
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hanqin Peng
- Division of Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Himanshu Batra
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Lorenza Bellusci
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - S. Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - David C. Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Kevin O. Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University, Durham, NC 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ming Tian
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Hidde Ploegh
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Tom Kirchhausen
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bing Chen
- Division of Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Frederick W. Alt
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
41
|
Li D, Martinez DR, Schäfer A, Chen H, Barr M, Sutherland LL, Lee E, Parks R, Mielke D, Edwards W, Newman A, Bock KW, Minai M, Nagata BM, Gagne M, Douek DC, DeMarco CT, Denny TN, Oguin TH, Brown A, Rountree W, Wang Y, Mansouri K, Edwards RJ, Ferrari G, Sempowski GD, Eaton A, Tang J, Cain DW, Santra S, Pardi N, Weissman D, Tomai MA, Fox CB, Moore IN, Andersen H, Lewis MG, Golding H, Seder R, Khurana S, Baric RS, Montefiori DC, Saunders KO, Haynes BF. Breadth of SARS-CoV-2 neutralization and protection induced by a nanoparticle vaccine. Nat Commun 2022; 13:6309. [PMID: 36274085 PMCID: PMC9588772 DOI: 10.1038/s41467-022-33985-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/11/2022] [Indexed: 12/25/2022] Open
Abstract
Coronavirus vaccines that are highly effective against current and anticipated SARS-CoV-2 variants are needed to control COVID-19. We previously reported a receptor-binding domain (RBD)-sortase A-conjugated ferritin nanoparticle (scNP) vaccine that induced neutralizing antibodies against SARS-CoV-2 and pre-emergent sarbecoviruses and protected non-human primates (NHPs) from SARS-CoV-2 WA-1 infection. Here, we find the RBD-scNP induced neutralizing antibodies in NHPs against pseudoviruses of SARS-CoV and SARS-CoV-2 variants including 614G, Beta, Delta, Omicron BA.1, BA.2, BA.2.12.1, and BA.4/BA.5, and a designed variant with escape mutations, PMS20. Adjuvant studies demonstrate variant neutralization titers are highest with 3M-052-aqueous formulation (AF). Immunization twice with RBD-scNPs protect NHPs from SARS-CoV-2 WA-1, Beta, and Delta variant challenge, and protect mice from challenges of SARS-CoV-2 Beta variant and two other heterologous sarbecoviruses. These results demonstrate the ability of RBD-scNPs to induce broad neutralization of SARS-CoV-2 variants and to protect animals from multiple different SARS-related viruses. Such a vaccine could provide broad immunity to SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Dapeng Li
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - David R. Martinez
- grid.10698.360000000122483208Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Alexandra Schäfer
- grid.10698.360000000122483208Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Haiyan Chen
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Maggie Barr
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA
| | - Laura L. Sutherland
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA
| | - Esther Lee
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Robert Parks
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA
| | - Dieter Mielke
- grid.26009.3d0000 0004 1936 7961Department of Surgery, Duke University School of Medicine, Durham, NC 27710 USA
| | - Whitney Edwards
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA
| | - Amanda Newman
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Kevin W. Bock
- grid.94365.3d0000 0001 2297 5165Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814 USA
| | - Mahnaz Minai
- grid.94365.3d0000 0001 2297 5165Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814 USA
| | - Bianca M. Nagata
- grid.94365.3d0000 0001 2297 5165Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814 USA
| | - Matthew Gagne
- grid.94365.3d0000 0001 2297 5165Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814 USA
| | - Daniel C. Douek
- grid.94365.3d0000 0001 2297 5165Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814 USA
| | - C. Todd DeMarco
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Thomas N. Denny
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Thomas H. Oguin
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Alecia Brown
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Wes Rountree
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Yunfei Wang
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Katayoun Mansouri
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA
| | - Robert J. Edwards
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Guido Ferrari
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Surgery, Duke University School of Medicine, Durham, NC 27710 USA
| | - Gregory D. Sempowski
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Amanda Eaton
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Surgery, Duke University School of Medicine, Durham, NC 27710 USA
| | - Juanjie Tang
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20871 USA
| | - Derek W. Cain
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA
| | - Sampa Santra
- grid.239395.70000 0000 9011 8547Beth Israel Deaconess Medical Center, Boston, MA 02215 USA
| | - Norbert Pardi
- grid.25879.310000 0004 1936 8972Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Drew Weissman
- grid.25879.310000 0004 1936 8972Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mark A. Tomai
- grid.417536.20000 0001 0695 6319Corporate Research Materials Lab, 3M Company, St Paul, MN 55144 USA
| | - Christopher B. Fox
- grid.53959.330000 0004 1794 8076Infectious Disease Research Institute, Seattle, WA 98104 USA
| | - Ian N. Moore
- grid.94365.3d0000 0001 2297 5165Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814 USA
| | - Hanne Andersen
- grid.282501.c0000 0000 8739 6829BIOQUAL, Rockville, MD 20850 USA
| | - Mark G. Lewis
- grid.282501.c0000 0000 8739 6829BIOQUAL, Rockville, MD 20850 USA
| | - Hana Golding
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20871 USA
| | - Robert Seder
- grid.94365.3d0000 0001 2297 5165Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814 USA
| | - Surender Khurana
- grid.417587.80000 0001 2243 3366Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20871 USA
| | - Ralph S. Baric
- grid.10698.360000000122483208Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - David C. Montefiori
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Surgery, Duke University School of Medicine, Durham, NC 27710 USA
| | - Kevin O. Saunders
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Surgery, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Immunology, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710 USA
| | - Barton F. Haynes
- grid.26009.3d0000 0004 1936 7961Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University School of Medicine, Durham, NC 27710 USA ,grid.26009.3d0000 0004 1936 7961Department of Immunology, Duke University School of Medicine, Durham, NC 27710 USA
| |
Collapse
|
42
|
Shi R, Zeng J, Xu L, Wang F, Duan X, Wang Y, Wu Z, Yu D, Huang Q, Yao YG, Yan J. A combination vaccine against SARS-CoV-2 and H1N1 influenza based on receptor binding domain trimerized by six-helix bundle fusion core. EBioMedicine 2022; 85:104297. [PMID: 36206623 PMCID: PMC9530591 DOI: 10.1016/j.ebiom.2022.104297] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/07/2022] Open
Abstract
Background Increasing severe morbidity and mortality by simultaneous or sequential infections with SARS-CoV-2 and influenza A viruses (IAV), especially in the elderly and obese patients, highlight the urgency of developing a combination vaccine against COVID-19 and influenza. Methods Self-assembling SARS-CoV-2 RBD-trimer and Influenza H1N1 HA1-trimer antigens were constructed, upon the stable fusion core in post-fusion conformation. Immunogenicity of SARS-CoV-2 RBD-trimer vaccine and H1N1 HA1-trimer antigens candidates were evaluated in mice. Protection efficacy of a combination vaccine candidate against SARS-CoV-2 and IAV challenge was identified using the K18-hACE2 mouse model. Findings Both the resultant RBD-trimer for SARS-CoV-2 and HA1-trimer for H1N1 influenza fully exposed receptor-binding motifs (RBM) or receptor-binding site (RBS). Two-dose RBD-trimer induced significantly higher binding and neutralizing antibody titers, and also a strong Th1/Th2 balanced cellular immune response in mice. Similarly, the HA1-trimer vaccine was confirmed to exhibit potent immunogenicity in mice. A combination vaccine candidate, composed of RBD-trimer and HA1-trimer, afforded high protection efficacy in mouse models against stringent lethal SARS-CoV-2 and homogenous H1N1 influenza co-infection, characterized by 100% survival rate. Interpretation Our results represent a proof of concept for a combined vaccine candidate based on trimerized receptor binding domain against co-epidemics of COVID-19 and influenza. Funding This project was funded by the Strategic Priority Research Program of CAS (XDB29040201), the National Natural Science Foundation of China (81830050, 81901680, and 32070569) and China Postdoctoral Science Foundation (2021M703450).
Collapse
Affiliation(s)
- Rui Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiawei Zeng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China,Kunming National High-level Biosafety Research Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Fengze Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaomin Duan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheng Wu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,Institute of Physical Science and Information, Anhui University, Hefei, 230039, China
| | - Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China,Kunming National High-level Biosafety Research Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Qingrui Huang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,Corresponding authors.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China,Kunming National High-level Biosafety Research Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China,National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China,Corresponding authors.
| | - Jinghua Yan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China,Corresponding authors.
| |
Collapse
|
43
|
Ghaemi A, Roshani Asl P, Zargaran H, Ahmadi D, Hashimi AA, Abdolalipour E, Bathaeian S, Miri SM. Recombinant COVID-19 vaccine based on recombinant RBD/Nucleoprotein and saponin adjuvant induces long-lasting neutralizing antibodies and cellular immunity. Front Immunol 2022; 13:974364. [PMID: 36159845 PMCID: PMC9494508 DOI: 10.3389/fimmu.2022.974364] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022] Open
Abstract
SARS-CoV-2 has caused a global pandemic, infecting millions of people. An effective preventive vaccine against this virus is urgently needed. Here, we designed and developed a novel formulated recombinant receptor-binding domain (RBD) nucleocapsid (N) recombinant vaccine candidates. The RBD and N were separately expressed in E. coli and purified using column chromatography. The female Balb/c mice were immunized subcutaneously with the combination of purified RBD and N alone or formulated with saponin adjuvant in a two-week interval in three doses. Neutralization antibody (Nabs) titers against the SARS-CoV-2 were detected by a Surrogate Virus Neutralization (sVNT) Test. Also, total IgG and IgG1, and IgG2a isotypes and the balance of cytokines in the spleen (IFN-γ, Granzyme B, IL-4, and IL-12) were measured by ELISA. The percentages of CD4+ and CD8+ T cells were quantified by flow cytometry. The lymphoproliferative activity of restimulated spleen cells was also determined. The findings showed that the combination of RBD and N proteins formulated with saponin significantly promoted specific total IgG and neutralization antibodies, elicited robust specific lymphoproliferative and T cell response responses. Moreover, marked increase in CD4+ and CD8+ T cells were observed in the adjuvanted RBD and N vaccine group compared with other groups. The results suggest that the formulations are able to elicit a specific long-lasting mixed Th1/Th2 balanced immune response. Our data indicate the significance of the saponin-adjuvanted RBD/N vaccine in the design of SARS-CoV-2 vaccines and provide a rationale for the development of a protective long-lasting and strong vaccine.
Collapse
Affiliation(s)
- Amir Ghaemi
- Department of Influenza and other respiratory viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Parisa Roshani Asl
- Department of Influenza and other respiratory viruses, Pasteur Institute of Iran, Tehran, Iran
| | | | - Delaram Ahmadi
- Department of Influenza and other respiratory viruses, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Sahar Bathaeian
- Department of Influenza and other respiratory viruses, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
44
|
Chen L, Qi X, Liang D, Li G, Peng X, Li X, Ke B, Zheng H, Liu Z, Ke C, Liao G, Liu L, Feng Q. Human Fc-Conjugated Receptor Binding Domain-Based Recombinant Subunit Vaccines with Short Linker Induce Potent Neutralizing Antibodies against Multiple SARS-CoV-2 Variants. Vaccines (Basel) 2022; 10:vaccines10091502. [PMID: 36146579 PMCID: PMC9505662 DOI: 10.3390/vaccines10091502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
The coronavirus disease-19 (COVID-19) pandemic has been ongoing since December 2019, with more than 6.3 million deaths reported globally as of August 2022. Despite the success of several SARS-CoV-2 vaccines, the rise in variants, some of which are resistant to the effects of vaccination, highlights the need for a so-called pan-coronavirus (universal) vaccine. Here, we performed an immunogenicity comparison of prototype vaccines containing spike protein receptor-binding domain (RBD) residues 319–541, or spike protein regions S1, S2 and S fused to a histidine-tagged or human IgG1 Fc (hFC) fragment with either a longer (six residues) or shorter (three residues) linker. While all recombinant protein vaccines developed were effective in eliciting humoral immunity, the RBD-hFc vaccine was able to generate a potent neutralizing antibody response as well as a cellular immune response. We then compared the effects of recombinant protein length and linker size on immunogenicity in vivo. We found that a longer recombinant RBD protein (residues 319–583; RBD-Plus-hFc) containing a small alanine linker (AAA) was able to trigger long-lasting, high-titer neutralizing antibodies in mice. Finally, we evaluated cross-neutralization of wild-type and mutant RBD-Plus-hFc vaccines against wild-type, Alpha, Beta, Delta and Omicron SARS-CoV-2 variants. Significantly, at the same antigen dose, wild-type RBD-Plus-hFc immune sera induced broadly neutralizing antibodies against wild-type, Alpha, Beta, Delta and Omicron variants. Taken together, our findings provide valuable information for the continued development of recombinant protein-based SARS-CoV-2 vaccines and a basic foundation for booster vaccinations to avoid reinfection with SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Liqing Chen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaoxiao Qi
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Dan Liang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510006, China
| | - Guiqi Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaofang Peng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510006, China
| | - Xiaohui Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Bixia Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510006, China
| | - Huanying Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510006, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Changwen Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510006, China
| | - Guochao Liao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Guangdong Hengda Biomedical Technology Co., Ltd., Guangzhou 510006, China
- Correspondence: (G.L.); (L.L.); (Q.F.)
| | - Liang Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Guangdong Hengda Biomedical Technology Co., Ltd., Guangzhou 510006, China
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou 510006, China
- Correspondence: (G.L.); (L.L.); (Q.F.)
| | - Qian Feng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Correspondence: (G.L.); (L.L.); (Q.F.)
| |
Collapse
|
45
|
Cao X, Zai J, Zhao Q, Xie L, Li Y. Intranasal immunization with recombinant Vaccinia virus encoding trimeric SARS-CoV-2 spike receptor-binding domain induces neutralizing antibody. Vaccine 2022; 40:5757-5763. [PMID: 36055873 PMCID: PMC9420698 DOI: 10.1016/j.vaccine.2022.08.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/01/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022]
Abstract
Respiratory transmission of SARS-CoV-2 is considered to be the major dissemination route for COVID-19, therefore, mucosal immune responses have great importance in preventing SARS-CoV-2 from infection. In this study, we constructed a recombinant Vaccinia virus (VV) harboring trimeric receptor-binding domain (RBD) of SARS-CoV-2 spike protein (VV-tRBD), and evaluated the immune responses towards RBD following intranasal immunization against mice and rabbits. In BALB/c mice, intranasal immunization with VV-tRBD elicited robust humoral and cellular immune responses, with high-level of both neutralizing IgG and IgA in sera against SARS-CoV-2 psudoviruses, and a number of RBD-specific IFN-γ-secreting lymphocytes. Sera from immunized rabbits also exhibited neutralization effects. Notably, RBD-specific secretory IgA (sIgA) in both nasal washes and bronchoalveolar lavage fluids (BALs) were detectable and showed substantial neutralization activities. Collectively, a recombinant VV expressing trimeric RBD confers robust systemic immune response and mucosal neutralizing antibodies, thus warranting further exploration as a mucosal vaccine.
Collapse
Affiliation(s)
- Xiaoling Cao
- College of Life Science and Technology, Wuhan University of Bioengineering, Wuhan, China
| | - Junjie Zai
- Immunology Innovation Team, School of Medicine, Ningbo University, Ningbo, China.
| | - Qingzhen Zhao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China.
| | - Lilan Xie
- College of Life Science and Technology, Wuhan University of Bioengineering, Wuhan, China; Hubei Engineering Research Center of Viral Vector, Applied Biotechnology Research Center, Wuhan University of Bioengineering, Wuhan, China.
| | - Yaoming Li
- College of Life Science and Technology, Wuhan University of Bioengineering, Wuhan, China; Hubei Engineering Research Center of Viral Vector, Applied Biotechnology Research Center, Wuhan University of Bioengineering, Wuhan, China.
| |
Collapse
|
46
|
Zhang J, Han ZB, Liang Y, Zhang XF, Jin YQ, Du LF, Shao S, Wang H, Hou JW, Xu K, Lei W, Lei ZH, Liu ZM, Zhang J, Hou YN, Liu N, Shen FJ, Wu JJ, Zheng X, Li XY, Li X, Huang WJ, Wu GZ, Su JG, Li QM. A mosaic-type trimeric RBD-based COVID-19 vaccine candidate induces potent neutralization against Omicron and other SARS-CoV-2 variants. eLife 2022; 11:e78633. [PMID: 36004719 PMCID: PMC9481243 DOI: 10.7554/elife.78633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Large-scale populations in the world have been vaccinated with COVID-19 vaccines, however, breakthrough infections of SARS-CoV-2 are still growing rapidly due to the emergence of immune-evasive variants, especially Omicron. It is urgent to develop effective broad-spectrum vaccines to better control the pandemic of these variants. Here, we present a mosaic-type trimeric form of spike receptor-binding domain (mos-tri-RBD) as a broad-spectrum vaccine candidate, which carries the key mutations from Omicron and other circulating variants. Tests in rats showed that the designed mos-tri-RBD, whether used alone or as a booster shot, elicited potent cross-neutralizing antibodies against not only Omicron but also other immune-evasive variants. Neutralizing antibody ID50 titers induced by mos-tri-RBD were substantially higher than those elicited by homo-tri-RBD (containing homologous RBDs from prototype strain) or the BIBP inactivated COVID-19 vaccine (BBIBP-CorV). Our study indicates that mos-tri-RBD is highly immunogenic, which may serve as a broad-spectrum vaccine candidate in combating SARS-CoV-2 variants including Omicron.
Collapse
Affiliation(s)
- Jing Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zi Bo Han
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Liang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xue Feng Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Qin Jin
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
| | - Li Fang Du
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Shuai Shao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Hui Wang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Jun Wei Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ke Xu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Wenwen Lei
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ze Hua Lei
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zhao Ming Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Zhang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Ya Nan Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ning Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Fu Jie Shen
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Juan Wu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xiang Zheng
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Yu Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Wei Jin Huang
- National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Gui Zhen Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ji Guo Su
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Qi Ming Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| |
Collapse
|
47
|
Overduin M, Kervin TA, Tran A. Progressive membrane-binding mechanism of SARS-CoV-2 variant spike proteins. iScience 2022; 25:104722. [PMID: 35813872 PMCID: PMC9251956 DOI: 10.1016/j.isci.2022.104722] [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: 03/09/2022] [Revised: 05/15/2022] [Accepted: 06/28/2022] [Indexed: 12/09/2022] Open
Abstract
Membrane recognition by viral spike proteins is critical for infection. Here we show the host cell membrane-binding surfaces of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike variants Alpha, Beta, Gamma, Delta, Epsilon, Kappa, and Omicron as well as SARS-CoV-1 and pangolin and bat relatives. They show increases in membrane binding propensities over time, with all spike head mutations in variants, and particularly BA.1, impacting the protein's affinity to cell membranes. Comparison of hundreds of structures yields a progressive model of membrane docking in which spike protein trimers shift from initial perpendicular stances to increasingly tilted positions that draw viral particles alongside host cell membranes before optionally engaging angiotensin-converting enzyme 2 (ACE2) receptors. This culminates in the assembly of the symmetric fusion apparatus, with enhanced membrane interactions of variants explaining their unique cell fusion capacities and COVID-19 disease transmission rates.
Collapse
Affiliation(s)
- Michael Overduin
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Troy A. Kervin
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Anh Tran
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
48
|
Firmino-Cruz L, dos-Santos JS, da Fonseca-Martins AM, Oliveira-Maciel D, Guadagnini-Perez G, Roncaglia-Pereira VA, Dumard CH, Guedes-da-Silva FH, Vicente Santos AC, Alvim RGF, Lima TM, Marsili FF, Abreu DPB, Rossi-Bergmann B, Vale AM, Filardy AD, Silva JL, de Oliveira AC, Gomes AMO, de Matos Guedes HL. Intradermal Immunization of SARS-CoV-2 Original Strain Trimeric Spike Protein Associated to CpG and AddaS03 Adjuvants, but Not MPL, Provide Strong Humoral and Cellular Response in Mice. Vaccines (Basel) 2022; 10:1305. [PMID: 36016193 PMCID: PMC9415730 DOI: 10.3390/vaccines10081305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Despite the intramuscular route being the most used vaccination strategy against SARS-CoV-2, the intradermal route has been studied around the globe as a strong candidate for immunization against SARS-CoV-2. Adjuvants have shown to be essential vaccine components that are capable of driving robust immune responses and increasing the vaccination efficacy. In this work, our group aimed to develop a vaccination strategy for SARS-CoV-2 using a trimeric spike protein, by testing the best route with formulations containing the adjuvants AddaS03, CpG, MPL, Alum, or a combination of two of them. Our results showed that formulations that were made with AddaS03 or CpG alone or AddaS03 combined with CpG were able to induce high levels of IgG, IgG1, and IgG2a; high titers of neutralizing antibodies against SARS-CoV-2 original strain; and also induced high hypersensitivity during the challenge with Spike protein and a high level of IFN-γ producing CD4+ T-cells in mice. Altogether, those data indicate that AddaS03, CpG, or both combined may be used as adjuvants in vaccines for COVID-19.
Collapse
Affiliation(s)
- Luan Firmino-Cruz
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Júlio Souza dos-Santos
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Alessandra Marcia da Fonseca-Martins
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Diogo Oliveira-Maciel
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Gustavo Guadagnini-Perez
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Victor A. Roncaglia-Pereira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
| | - Carlos H. Dumard
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
| | - Francisca H. Guedes-da-Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
| | - Ana C. Vicente Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
| | - Renata G. F. Alvim
- Cell Culture Engineering Lab., COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-598, RJ, Brazil
| | - Tulio M. Lima
- Cell Culture Engineering Lab., COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-598, RJ, Brazil
| | - Federico F. Marsili
- Cell Culture Engineering Lab., COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-598, RJ, Brazil
| | - Daniel P. B. Abreu
- Cell Culture Engineering Lab., COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-598, RJ, Brazil
| | - Bartira Rossi-Bergmann
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Andre M. Vale
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Alessandra D’Almeida Filardy
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Jerson Lima Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
| | - Andrea Cheble de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
| | - Andre M. O. Gomes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, RJ, Brazil
| | - Herbert Leonel de Matos Guedes
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
- Clinical Immunology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
| |
Collapse
|
49
|
Induction of Broadly Cross-Reactive Antibody Responses to SARS-CoV-2 Variants by S1 Nanoparticle Vaccines. J Virol 2022; 96:e0038322. [PMID: 35699445 PMCID: PMC9278117 DOI: 10.1128/jvi.00383-22] [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] [Indexed: 01/09/2023] Open
Abstract
Despite the rapid deployment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines, the emergence of SARS-CoV-2 variants and reports of their immune evasion characteristics have led to an urgent need for novel vaccines that confer potent cross-protective immunity. In this study, we constructed three different SARS-CoV-2 spike S1-conjugated nanoparticle vaccine candidates that exhibited high structural homogeneity and stability. Notably, these vaccines elicited up to 50-times-higher neutralizing antibody titers than the S1 monomer in mice. Crucially, it was found that the S1-conjugated nanoparticle vaccine could elicit comparable levels of neutralizing antibodies against wild-type or emerging variant SARS-CoV-2, with cross-reactivity to SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV), the effect of which could be further enhanced using our designed nanoparticles. Our results indicate that the S1-conjugated nanoparticles are promising vaccine candidates with the potential to elicit potent and cross-reactive immunity against not only wild-type SARS-CoV-2, but also its variants of concern, variants of interest, and even other pathogenic betacoronaviruses. IMPORTANCE The emergence of SARS-CoV-2 variants led to an urgent demand for a broadly effective vaccine against the threat of variant infection. The spike protein S1-based nanoparticle designed in our study could elicit a comprehensive humoral response toward different SARS-CoV-2 variants of concern and variants of interest and will be helpful to combat COVID-19 globally.
Collapse
|
50
|
Qu J, Yang F, Zhu T, Wang Y, Fang W, Ding Y, Zhao X, Qi X, Xie Q, Chen M, Xu Q, Xie Y, Sun Y, Chen D. A reference single-cell regulomic and transcriptomic map of cynomolgus monkeys. Nat Commun 2022; 13:4069. [PMID: 35831300 PMCID: PMC9279386 DOI: 10.1038/s41467-022-31770-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/01/2022] [Indexed: 12/24/2022] Open
Abstract
Non-human primates are attractive laboratory animal models that accurately reflect both developmental and pathological features of humans. Here we present a compendium of cell types across multiple organs in cynomolgus monkeys (Macaca fascicularis) using both single-cell chromatin accessibility and RNA sequencing data. The integrated cell map enables in-depth dissection and comparison of molecular dynamics, cell-type compositions and cellular heterogeneity across multiple tissues and organs. Using single-cell transcriptomic data, we infer pseudotime cell trajectories and cell-cell communications to uncover key molecular signatures underlying their cellular processes. Furthermore, we identify various cell-specific cis-regulatory elements and construct organ-specific gene regulatory networks at the single-cell level. Finally, we perform comparative analyses of single-cell landscapes among mouse, monkey and human. We show that cynomolgus monkey has strikingly higher degree of similarities in terms of immune-associated gene expression patterns and cellular communications to human than mouse. Taken together, our study provides a valuable resource for non-human primate cell biology. Non-human primates are attractive laboratory animal models that can accurately reflect some developmental and pathological features of humans. Here the authors chart a reference cell map of cynomolgus monkeys using both scATAC-seq and scRNA-seq data across multiple organs, providing insights into the molecular dynamics and cellular heterogeneity of this organism.
Collapse
Affiliation(s)
- Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Fa Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Tao Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Yingshuo Wang
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 310052, Hangzhou, China
| | - Wen Fang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Yan Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Xue Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Xianjia Qi
- Shanghai XuRan Biotechnology Co., Ltd., 1088 Zhongchun Road, 201109, Shanghai, China
| | - Qiangmin Xie
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 310052, Hangzhou, China
| | - Ming Chen
- College of Life Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Yicheng Xie
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 310052, Hangzhou, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China. .,Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, China.
| | - Dijun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China.
| |
Collapse
|