1
|
Xu L, Yu Z, Xu Y, Wang Q, Wang G, Li B, Weng Q, Yi Y, Li J. An mRNA vaccine candidate encoding cholera toxin subunit B and conserved antigens of influenza viruses confers cross-protection against influenza a viruses in adult and aged mice. Hum Vaccin Immunother 2025; 21:2453304. [PMID: 39957235 PMCID: PMC11834421 DOI: 10.1080/21645515.2025.2453304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 02/18/2025] Open
Abstract
Currently, vaccination with influenza vaccines is still an effective strategy to prevent infection by seasonal influenza virus. However, seasonal influenza vaccines frequently fail to induce effective immune protection against rapidly changing seasonal influenza viruses and emerging zoonotic influenza viruses. In addition, seasonal influenza vaccines may not confer potent protection in elderly and immunocompromised individuals. There is an urgent need to develop potent broad-spectrum influenza vaccines to address this problem. Herein, we designed an mRNA-based broad-spectrum influenza vaccine candidate encoding cholera toxin subunit B and conserved antigens of influenza viruses. In both adult and aged mice, this universal influenza mRNA vaccine candidate stimulated robust T-cell and humoral immune responses and conferred effective protection against broad-spectrum influenza viruses in both adult and aged mice.
Collapse
Affiliation(s)
- Liang Xu
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhihao Yu
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yifan Xu
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Qin Wang
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Guiqin Wang
- Nanjing Advanced Academy of Life and Health, Nanjing, China
| | - Benchi Li
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian Weng
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yongxiang Yi
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Junwei Li
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Medical Innovation Center for Infectious Disease of Jiangsu Province, Nanjing, China
| |
Collapse
|
2
|
Liu Y, Yang M, Nie M, Wu S, Su R, Qiu D, Lu S, Xiong H, Zhang J, Ge S, Yuan Q, Zhao Q, Zhang T, Wang Y, Xia N. Immunological enhancement of micro-nanoparticle formulated with risedronate and zinc as vaccine adjuvant in aged mice. Immun Ageing 2025; 22:17. [PMID: 40361120 PMCID: PMC12070681 DOI: 10.1186/s12979-025-00512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Elderly individuals face heightened susceptibility to infectious diseases and diminished vaccine responses. Vaccine adjuvants offer a solution. Despite aluminum adjuvant's long history, its limitations in inducing strong cellular immunity and protecting immunocompromised individuals restrict its application. Building upon our previous development of zinc salt particle-based risedronate (Zn-RS), we systematically investigated the immunoenhancing effects of Zn-RS in aged mice and thoroughly explored the underlying mechanisms responsible for these observations in this study. RESULTS Compared to formulations using aluminum adjuvant, Zn-RS combined with either varicella-zoster virus glycoprotein E (gE) or SARS-CoV-2 monovalent STFK protein (STFK) elicited significantly higher IgG and neutralization titers, as well as superior long-term humoral immunity. Moreover, Zn-RS induced greater quantities of dendritic cells (DCs), antigen-presenting cells (APCs), follicular helper T (TFH) cells, Th1/Th2/Th9/Th17 type immune cells, germinal center B cells (GCBs) and plasma cells. CONCLUSIONS These findings support Zn-RS as a promising adjuvant candidate for elderly populations, warranting further exploration of its mechanisms and potential applications.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Man Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Meifeng Nie
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shuyu Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Rong Su
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Dekui Qiu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shouneng Lu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hualong Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jinlei Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shengxiang Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China.
| | - Tianying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health & School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| |
Collapse
|
3
|
Yang C, Li E, Guo X, Xie W, Wang Y, Huang X, Chiu S, Wu X. A Self-Assembled Nanovaccine with BA.4/5 Receptor-Binding Domain and CpG Oligodeoxynucleotides Induces Broad-Spectrum Neutralization against SARS-CoV-2 Omicron Subvariants. ACS NANO 2025; 19:16424-16437. [PMID: 40265996 DOI: 10.1021/acsnano.4c17269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Over the past 3 years, SARS-CoV-2 Omicron has been circulating globally with the emergence of multiple subvariants, including BA.5, BA.5.2, XBB, XBB.1, EG.5.1, HK.3, BA.2.86, JN.1, and KP.2. To combat these Omicron subvariants, several vaccines based on receptor-binding domain (RBD) dimers have been developed; however, RBD dimer vaccines require frequent updates to cope with the emergence of new variants. In contrast, the development of a safe, effective, and broad-spectrum vaccine against multiple Omicron subvariants, including the latest JN.1 and KP.2, would be a one-size-fits-all solution. Here, we designed BA.4/5 RBD-PC7A conjugate micelles by displaying the BA.4/5 RBD in PC7A micelles. Remarkably, the micelles elicited potent neutralizing antibodies (NAbs) in rabbits, effectively neutralizing BA.5.2, XBB.1.18, and HK.3 infections. Moreover, the micelles alone were able to induce NAbs in mice against the BA.5 variant. When a cytosine-phosphate-guanine (CpG) adjuvant was added and electrostatically adsorbed to the micelles, there was a significant increase in the antibody titers of IgG1, IgG2b, and IgG2c. This enhancement facilitated the broad neutralization of various strains, including BA.5.2, XBB.1.18, HK.3, JN.1, and KP.2. Furthermore, the micelles adsorbed with CpG protected golden hamsters from infection with the BA.5.2 strain. This study presents a potent and broadly neutralizing nanovaccine that includes the BA.4/5 RBD antigen and a CpG adjuvant. It demonstrates efficacy against multiple Omicron subvariants, including BA.5, BA.5.2, XBB.1.18, HK.3, JN.1, and KP.2, highlighting its potential for clinical translation.
Collapse
Affiliation(s)
- Chendong Yang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong Key Laboratory of Carbohydrate and Carbohydrate-conjugate Drugs, Shandong University, Qingdao, Shandong 266237, China
| | - Entao Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaoping Guo
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Wenyu Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yuanzhan Wang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong Key Laboratory of Carbohydrate and Carbohydrate-conjugate Drugs, Shandong University, Qingdao, Shandong 266237, China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Sandra Chiu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui 230027, China
| | - Xuanjun Wu
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong Key Laboratory of Carbohydrate and Carbohydrate-conjugate Drugs, Shandong University, Qingdao, Shandong 266237, China
| |
Collapse
|
4
|
Su R, Yao T, Cao C, Yang Y, Chen M, Wu J, Zhao Y, Liu X, Li S, Ding J, Yang R, Shen S, Zhang C, Zhan C, Gao X. Enhancing Immune Responses Through Modulation of Innate Cell Microenvironments in Lymph Nodes with Virus-Mimetic Vaccines. Angew Chem Int Ed Engl 2025; 64:e202503845. [PMID: 40072248 DOI: 10.1002/anie.202503845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Nanovaccines hold significant promise for the prevention and treatment of infectious diseases. However, the efficacy of many nanovaccines is often limited by inadequate stimulation of both innate and adaptive immune responses. Herein, we explore a rational vaccine strategy aimed at modulating innate cell microenvironments within lymph nodes (LNs) to enhance the generation of effective immune responses. Inspired by the structure and natural infection process of viruses, we developed a versatile antigen and adjuvant co-delivery platform, termed virus-mimetic vaccines (VMVs). Specifically, polyarginine-tagged antigens were noncovalently assembled onto nucleic acid nanogels containing cytosine-phosphate-guanine oligodeoxynucleotide via a salt-bridge zipper mechanism, which can activate Toll-like receptor 9. Upon intramuscular immunization, VMVs effectively drained into the LNs, recruiting and activating multiple innate cells, including CD8+ dendritic cells (DCs), CD103+ DCs, macrophages, plasmacytoid DCs, and neutrophils. This activation modulates the innate cell microenvironments and relocates antigen-presenting cells within LNs, optimizing adaptive immune responses. VMVs induced a robust antigen-specific immune response, characterized by high levels of neutralizing antibodies, augmented memory T cell activity, and enhanced development of germinal center B cells. Together, our findings demonstrate that dynamic modulation of innate cell microenvironments by VMVs leads to optimized generation of both humoral and cellular immunity against infectious diseases.
Collapse
Affiliation(s)
- Runping Su
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, P.R. China
| | - Tingting Yao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Chong Cao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
| | - Yaqi Yang
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Mingshan Chen
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Jianxiao Wu
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Yue Zhao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Xiaoxiao Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Sha Li
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Junqiang Ding
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Rong Yang
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
| | - Shun Shen
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Chuan Zhang
- Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Changyou Zhan
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, P.R. China
| | - Xihui Gao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| |
Collapse
|
5
|
Kanjo K, Lothe R, Nagar G, Rajurkar M, Rao H, Batwal S, Shaligram U, Varadarajan R. Destabilising Effect of Class B CpG Adjuvants on Different Proteins and Vaccine Candidates. Vaccines (Basel) 2025; 13:395. [PMID: 40333326 PMCID: PMC12031019 DOI: 10.3390/vaccines13040395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/19/2025] [Accepted: 04/02/2025] [Indexed: 05/09/2025] Open
Abstract
Background: Adjuvants function by enhancing the breadth, durability, and magnitude of the immune response, but little is known about their impact on vaccine stability. CpG is a widely used adjuvant that is included in several recently approved COVID-19 vaccines using Spike protein, RBD, or whole inactivated virus. Methods: Here, we investigate the in vitro stability of the Receptor-Binding Domain (RBD) of the SARS-CoV-2 Spike protein, as well as a number of other proteins formulated with a class B CpG adjuvant. Results: We show that RBD, BSA, and lysozyme proteins are less thermally stable, more aggregation-prone, and more protease-sensitive in the presence of CpG than without it, and that these effects are enhanced with prolonged incubation. For RBD, the effects of CpG are pH-independent but dependent on the salt concentration, with relative destabilisation decreasing with an increasing salt concentration, indicative of an electrostatic component to the interaction between CpG and the protein. The reduced thermal and proteolytic stability found in the presence of CpG is indicative of a preferential interaction of CpG with the unfolded state of the protein relative to its native state. It remains to be determined if these in vitro characteristics are unique to CpG or are also shared by other non-CpG commercial adjuvants, if they are antigen-dependent, and if and how they correlate with the in vivo immunogenicity of an adjuvanted vaccine. Conclusions: It is demonstrated that the CpG adjuvant is critical to enhancing immunogenicity and is a key reason for the success of multiple licensed commercial vaccines. Nonetheless, our work suggests that careful and systematic in vitro formulation studies may be warranted for the development of suitable, stable formulations of CpG-adjuvanted vaccine candidates.
Collapse
Affiliation(s)
- Kawkab Kanjo
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India;
| | - Rakesh Lothe
- Serum Institute of India Pvt. Ltd., Pune 411028, India; (R.L.); (G.N.); (M.R.); (H.R.); (S.B.); (U.S.)
| | - Gaurav Nagar
- Serum Institute of India Pvt. Ltd., Pune 411028, India; (R.L.); (G.N.); (M.R.); (H.R.); (S.B.); (U.S.)
| | - Meghraj Rajurkar
- Serum Institute of India Pvt. Ltd., Pune 411028, India; (R.L.); (G.N.); (M.R.); (H.R.); (S.B.); (U.S.)
| | - Harish Rao
- Serum Institute of India Pvt. Ltd., Pune 411028, India; (R.L.); (G.N.); (M.R.); (H.R.); (S.B.); (U.S.)
| | - Saurabh Batwal
- Serum Institute of India Pvt. Ltd., Pune 411028, India; (R.L.); (G.N.); (M.R.); (H.R.); (S.B.); (U.S.)
| | - Umesh Shaligram
- Serum Institute of India Pvt. Ltd., Pune 411028, India; (R.L.); (G.N.); (M.R.); (H.R.); (S.B.); (U.S.)
| | - Raghavan Varadarajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India;
| |
Collapse
|
6
|
Janes ME, Gottlieb AP, Park KS, Acharya S, Bibbey MG, Mitragotri S. Controlling vaccine kinetics using tannic acid for enhanced humoral immunity. J Control Release 2025; 379:135-146. [PMID: 39733913 DOI: 10.1016/j.jconrel.2024.12.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
Despite the success of global vaccination campaigns, vaccine access in low-resource settings is an ongoing challenge. Subunit vaccines are a well-established and clinically scalable intervention, yet they have achieved limited success for poorly immunogenic antigens such as those associated with SARS-CoV-2. Delivery strategies that promote gradual release of subunit vaccines from the injection site offer the potential to improve humoral immunity by enhancing lymph node exposure, however, clinical implementation of this strategy is challenging due to poor scalability and high costs. Here, we propose an approach that uses the polyphenol tannic acid (TA) as a simple and inexpensive strategy to enhance tissue residence of vaccines and subsequent humoral immunity. We show that TA mediates supramolecular interactions between vaccine components and tissue at the subcutaneous injection site to promote extended retention of protein antigens for over one week. In addition to enhancing the magnitude and duration of vaccine drainage to the lymph nodes, inclusion of TA improved accumulation of activated, antigen-laden monocyte-derived dendritic cells (moDCs), promoting long-lasting humoral immunity against the receptor-binding domain (RBD) of SARS-CoV-2 and variants of concern. This system, termed TAPER (Tannic Acid-Promoted Enhanced Retention) provides various translational advantages including one-pot synthesis, scalability, low cost, and modularity, towards realization of effective and accessible subunit vaccines.
Collapse
Affiliation(s)
- Morgan E Janes
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA; John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Alexander P Gottlieb
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Kyung Soo Park
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Shrinivas Acharya
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Michael Griffith Bibbey
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA.
| |
Collapse
|
7
|
Maniyamgama N, Bae KH, Chang ZW, Lee J, Ang MJY, Tan YJ, Ng LFP, Renia L, White KP, Yang YY. Muco-Penetrating Lipid Nanoparticles Having a Liquid Core for Enhanced Intranasal mRNA Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407383. [PMID: 39888252 PMCID: PMC11923898 DOI: 10.1002/advs.202407383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/15/2024] [Indexed: 02/01/2025]
Abstract
Intranasal delivery of mRNA vaccines offers promising opportunities to combat airborne viruses like SARS-CoV-2 by provoking mucosal immunity, which not only defends against respiratory infection but also prevents contagious transmission. However, the development of nasal mRNA vaccines has been hampered by the lack of effective means to overcome the mucus barrier. Herein, ionizable lipid-incorporated liquid lipid nanoparticles (iLLNs) capable of delivering mRNA cargo across airway mucosa are designed. Adjusting the ratios of ionizable and cationic lipids allows fine-tuning of the pKa of iLLNs to the range of nasal mucosal pH (5.5-6.5), thus facilitating mucus penetration via the formation of near-neutral, PEGylated muco-inert surfaces. When nasally administered to mice, the top candidate iLLN-2/mRNA complexes enable about 60-fold greater reporter gene expression in the nasal cavity, compared to the benchmark mRNA-lipid nanoparticles (ALC-LNP) having the same lipid composition as that of BNT162b2 vaccine. Moreover, a prime-boost intranasal immunization of iLLN-2/mRNA complexes elicits a greater magnitude of SARS-CoV-2 spike-specific mucosal IgA and IgG response than ALC-LNP, without triggering any noticeable inflammatory reactions. Taken together, these results provide useful insights for the design of nasally deliverable mRNA formulations for prophylactic applications.
Collapse
Affiliation(s)
- Nipuni Maniyamgama
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Zi Wei Chang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
| | - Jialing Lee
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Melgious J. Y. Ang
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Yong Jie Tan
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
| | - Lisa F. P. Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore138648Republic of Singapore
- School of Biological SciencesNanyang Technological UniversitySingapore138648Republic of Singapore
| | - Kevin P. White
- Department of Biochemistry and Precision Medicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore119228Republic of Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| |
Collapse
|
8
|
Ji J, Chen L, Wu Z, Tang T, Zhu L, Zhu M, Chen Y, Lu X, Yao H. Enhancing antibody levels and T cell activity of quadrivalent influenza vaccine by combining it with CpG HP021. Sci Rep 2024; 14:31424. [PMID: 39733119 PMCID: PMC11682164 DOI: 10.1038/s41598-024-83026-x] [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: 07/17/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Influenza virus infections are a serious danger to people's health worldwide as they are responsible for seasonal flu outbreaks. There is an urgent need to improve the effectiveness and durability longevity of the immune response to influenza vaccines. We synthesized the CpG HP021 and examined the impact of it on the immune response to an influenza vaccine. In BALB/c mice, hemagglutination inhibition (HI) titers to the vaccine were increased four- to eightfold against H1N1, H3N2, BV, and BY viruses by 3 μg IIV4 + 40 μg CpG HP021 compared with those of the non-adjuvanted IIV4 group, and the CpG HP021 group had a broader HI activity. Additionally, the immune response was directed towards Type 1 T helper (Th1) cells due to the CpG HP021 adjuvant. The CpG HP021-adjuvanted IIV4 induced a higher number of T cells secreting interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α), and increased the percentage of effector memory T cells in mice. In SD rats, the immune responses induced by IIV4 with CpG HP021 were similar to those in BALB/c mice. The development of CpG HP021 may expand the options for adjuvants in vaccines against infectious diseases.
Collapse
Affiliation(s)
- Jia Ji
- State Key Laboratory for Diagnosis, Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China
| | - Lei Chen
- Zhejiang Toyouvax Bio-pharmaceutical Co., Ltd, Hangzhou, 311100, China
| | - Zhigang Wu
- State Key Laboratory for Diagnosis, Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Taoming Tang
- State Key Laboratory for Diagnosis, Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Linwei Zhu
- State Key Laboratory for Diagnosis, Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Miaojin Zhu
- State Key Laboratory for Diagnosis, Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yan Chen
- Jiangsu Taipurui Biotechnology Co., Ltd, Taizhou, 225300, China
| | - Xiangyun Lu
- State Key Laboratory for Diagnosis, Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis, Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China.
| |
Collapse
|
9
|
He J, Ding X, Zhao J, Zeng J, Zhou Y, Xiao W, Hua D, Liu M, Guo H, Zhang Y, Qiu M, Li J. A novel pan-epitope based nanovaccine self-assembled with CpG enhances immune responses against flavivirus. J Nanobiotechnology 2024; 22:738. [PMID: 39609873 PMCID: PMC11603839 DOI: 10.1186/s12951-024-03031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Flavivirus is a highly prevalent and outbreak-prone disease, affecting billions of individuals annually and posing substantial public health challenges. Vaccination is critical to reducing the global impact of flavivirus infections, making the development of a safe and effective vaccine a top priority. The self-assembled pan-epitope vaccine presents key advantages for improving immunogenicity and safety without relying on external vectors or adding immunomodulatory elements, both of which are essential for successful vaccine development. RESULTS In this study, the pan-epitope peptide TBT was combined with adjuvant CpG to form the TBT-CpG nanovaccine (TBT-CpG NaVs), which was found to be spherical, uniform in shape, and demonstrated strong serum stability. In vitro studies showed that the TBT-CpG NaVs were efficiently taken up and internalized by bone marrow-derived dendritic cells (BMDCs). Flow cytometry and transcriptomic analysis indicated that the antigens were effectively presented to antigen-presenting cells (APCs) via the MHC II pathway, which facilitated BMDCs maturation and promoted the release of pro-inflammatory cytokines IL-1β, TNF-α, and IL-6. In vivo studies confirmed that TBT-CpG NaVs enhanced antigen-specific IgG levels, significantly increased IFN-γ and IL-4 expression in spleen cells, and offered protective effects against Dengue virus (DENV) and Zika virus (ZIKV) infections. Safety evaluations revealed no hepatotoxicity and no significant organ damage in immunized mice. CONCLUSION The self-assembled candidate nanovaccine TBT-CpG NaVs effectively activates BMDCs and triggers a targeted immune response, providing antiviral effects against DENV and ZIKV. This vaccine demonstrates good immunogenicity and safety, establishing a promising foundation and a new strategy for the development of safe and effective vaccines.
Collapse
Affiliation(s)
- Jiuxiang He
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Xiaoyan Ding
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Jing Zhao
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Jie Zeng
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuxin Zhou
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Wen Xiao
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Dong Hua
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Minchi Liu
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Hongxia Guo
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Yu Zhang
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Minyue Qiu
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Jintao Li
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China.
| |
Collapse
|
10
|
Brook B, Checkervarty AK, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The BNT162b2 mRNA vaccine demonstrates reduced age-associated T H1 support in vitro and in vivo. iScience 2024; 27:111055. [PMID: 39569372 PMCID: PMC11576392 DOI: 10.1016/j.isci.2024.111055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 07/05/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024] Open
Abstract
mRNA vaccines demonstrate impaired immunogenicity and durability in vulnerable older populations. We hypothesized that human in vitro modeling and proteomics could elucidate age-specific mRNA vaccine actions. BNT162b2-stimulation changed the plasma proteome of blood samples from young (18-50Y) and older adult (≥60Y) participants, assessed by mass spectrometry, proximity extension assay, and multiplex. Young adult up-regulation (e.g., PSMC6, CPN1) contrasted reduced induction in older adults (e.g., TPM4, APOF, APOC2, CPN1, PI16). 30-85% lower TH1-polarizing cytokines and chemokines were induced in elderly blood (e.g., IFNγ, CXCL10). Analytes lower in older adult samples included human in vivo mRNA immunogenicity biomarkers (e.g., IFNγ, CXCL10, CCL4, IL-1RA). BNT162b2 also demonstrated reduced CD4+ TH1 responses in aged vs. young adult mice. Our study demonstrates the utility of human in vitro platforms modeling age-specific mRNA vaccine immunogenicity, highlights impaired support of TH1 polarization in older adults, and provides a rationale for precision mRNA vaccine adjuvantation to induce greater immunogenicity.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC V6Z 2K5, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amy C Sherman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine- Chair of Pediatrics, University of Rome, 00133 Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dheeraj Soni
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hanno Steen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
11
|
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
|
12
|
Ji J, Tang T, Zhu M, Wu Z, Zhang J, Shi D, Zhu L, Zhang X, Lu X, Chen L, Yao H. Boosting the immune response in COVID-19 vaccines via an Alum:CpG complex adjuvant. Antiviral Res 2024; 229:105954. [PMID: 38964615 DOI: 10.1016/j.antiviral.2024.105954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/02/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Selecting appropriate adjuvants is crucial for developing an effective vaccine. However, studies on the immune responses triggered by different adjuvants in COVID-19 inactivated vaccines are scarce. Herein, we evaluated the efficacy of Alum, CpG HP021, Alum combined with CpG HP021 (Alum/CpG), or MF-59 adjuvants with COVID-19 inactivated vaccines in K18-hACE2 mice, and compared the different immune responses between K18-hACE2 and BALB/c mice. In K18-hACE2 mice, the Alum/CpG group produced a 6.5-fold increase in anti-receptor-binding domain (RBD) IgG antibody titers compared to the Alum group, and generated a comparable level of antibodies even when the antigen amount was reduced by two-thirds, possibly due to the significant activation of germinal center (GC) structures in the central region of the spleen. Different adjuvants induced a variety of binding antibody isotypes. CpG HP021 and Alum/CpG were biased towards Th1/IgG2c, while Alum and MF-59 were biased toward Th2/IgG1. Cytokines IFN-γ, IL-2, and TNF-α were significantly increased in the culture supernatants of splenocytes specifically stimulated in the Alum/CpG group. The antibody responses in BALB/c mice were similar to those in K18-hACE2 mice, but with lower levels of neutralizing antibodies (NAbs). Notably, the Alum/CpG-adjuvanted inactivated vaccine induced a higher number of T cells secreting IFN-γ and IL-2, increased the percentage of effector memory T (TEM) cells among CD8+ T cells, and effectively protected K18-hACE2 mice from Delta variant challenge. Our results showed that Alum/CpG complex adjuvant significantly enhanced the immune response to inactivated COVID-19 antigens and could induce a long-lasting immune response.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Mice
- Adjuvants, Immunologic/administration & dosage
- Mice, Inbred BALB C
- Alum Compounds/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Adjuvants, Vaccine/administration & dosage
- Female
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Cytokines/immunology
- Humans
Collapse
Affiliation(s)
- Jia Ji
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Taoming Tang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Miaojin Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhigang Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jiale Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Danrong Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Linwei Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xiaodi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xiangyun Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lei Chen
- Zhejiang Toyouvax Bio-pharmaceutical Co., Ltd., Hangzhou, 311103, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
13
|
Zou GQ, Li K, Yan C, Li YQ, Xian MY, Hu X, Luo R, Liu Z. Aluminum hydroxide and immunostimulatory glycolipid adjuvant combination for enhanced COVID-19 subunit vaccine immunogenicity. Vaccine 2024; 42:126145. [PMID: 39034218 DOI: 10.1016/j.vaccine.2024.07.046] [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: 03/23/2024] [Revised: 06/11/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Protein-based subunit vaccines like RBD-Fc are promising tools to fight COVID-19. RBD-Fc fuses the receptor-binding domain (RBD) of the SARS-CoV-2 virus spike protein with the Fc region of human IgG1, making it more immunogenic than RBD alone. Earlier work showed that combining RBD-Fc with iNKT cell agonists as adjuvants improved neutralizing antibodies but did not sufficiently enhance T cell responses, a limitation RBD-Fc vaccines share with common adjuvants. Here we demonstrate that aluminum hydroxide combined with α-C-GC, a C-glycoside iNKT cell agonist, significantly improved the RBD-Fc vaccine's induction of RBD-specific T-cell responses. Additionally, aluminum hydroxide with α-GC-CPOEt, a phosphonate diester derivative, synergistically elicited more robust neutralizing antibodies. Remarkably, modifying αGC with phosphate (OPO3H2) or phosphonate (CPO3H2) to potentially enhance aluminum hydroxide interaction did not improve efficacy over unmodified αGC with aluminum hydroxide. These findings underscore the straightforward yet potent potential of this approach in advancing COVID-19 vaccine development and provide insights for iNKT cell-based immunotherapy.
Collapse
Affiliation(s)
- Guo-Qing Zou
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Ke Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Cheng Yan
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Ya-Qian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Mao-Ying Xian
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xing Hu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China.
| | - Zheng Liu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China.
| |
Collapse
|
14
|
Yao Z, Liang Z, Li M, Wang H, Ma Y, Guo Y, Chen C, Xue C, Sun B. Aluminum oxyhydroxide-Poly(I:C) combination adjuvant with balanced immunostimulatory potentials for prophylactic vaccines. J Control Release 2024; 372:482-493. [PMID: 38914205 DOI: 10.1016/j.jconrel.2024.06.054] [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: 03/06/2024] [Revised: 05/20/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
The development of high-purity antigens promotes the urgent need of novel adjuvant with the capability to trigger high levels of immune response. Polyinosinic-polycytidylic (Poly(I:C)) is a synthetic double-stranded RNA (dsRNA) that can engage Toll-like receptor 3 (TLR3) to initiate immune responses. However, the Poly(I:C)-induced toxicity and inefficient delivery prevent its applications. In our study, combination adjuvants are formulated by aluminum oxyhydroxide nanorods (AlOOH NRs) and Poly(I:C), named Al-Poly(I:C), and the covalent interaction between the two components is further demonstrated. Al-Poly(I:C) mediates enhanced humoral and cellular immune responses in three antigen models, i.e., HBsAg virus-like particles (VLPs), human papilloma virus (HPV) VLPs and varicella-zoster virus (VZV) glycoprotein E (gE). Further mechanistic studies demonstrate that the dose and molecular weight (MW) of Poly(I:C) determine the physicochemical properties and adjuvanticity of the Al-Poly(I:C) combination adjuvants. Al-Poly(I:C) with higher Poly(I:C) dose promotes antigen-bearing dendritic cells (DCs) recruitment and B cells proliferation in lymph nodes. Al-Poly(I:C) formulated with higher MW Poly(I:C) induces higher activation of helper T cells, B cells, and CTLs. This study demonstrates that Al-Poly(I:C) potentiates the humoral and cellular responses in vaccine formulations. It offers insights for adjuvant design to meet the formulation requirements in both prophylactic and therapeutic vaccines.
Collapse
Affiliation(s)
- Zhiying Yao
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Zhihui Liang
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Min Li
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Huiyang Wang
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Yubin Ma
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Yiyang Guo
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Chen Chen
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; MOE Key Laboratory Bio-Intelligent Manufacturing, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; MOE Key Laboratory Bio-Intelligent Manufacturing, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Bingbing Sun
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China.
| |
Collapse
|
15
|
Brook B, Duval V, Barman S, Speciner L, Sweitzer C, Khanmohammed A, Menon M, Foster K, Ghosh P, Abedi K, Koster J, Nanishi E, Baden LR, Levy O, VanCott T, Micol R, Dowling DJ. Adjuvantation of a SARS-CoV-2 mRNA vaccine with controlled tissue-specific expression of an mRNA encoding IL-12p70. Sci Transl Med 2024; 16:eadm8451. [PMID: 39047117 DOI: 10.1126/scitranslmed.adm8451] [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: 11/08/2023] [Revised: 03/18/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024]
Abstract
Messenger RNA (mRNA) vaccines were pivotal in reducing severe acute respiratory syndrome 2 (SARS-CoV-2) infection burden, yet they have not demonstrated robust durability, especially in older adults. Here, we describe a molecular adjuvant comprising a lipid nanoparticle (LNP)-encapsulated mRNA encoding interleukin-12p70 (IL-12p70). The bioactive adjuvant was engineered with a multiorgan protection (MOP) sequence to restrict transcript expression to the intramuscular injection site. Admixing IL-12-MOP (CTX-1796) with the BNT162b2 SARS-CoV-2 vaccine increased spike protein-specific immune responses in mice. Specifically, the benefits of IL-12-MOP adjuvantation included amplified humoral and cellular immunity and increased immune durability for 1 year after vaccination in mice. An additional benefit included the restoration of immunity in aged mice to amounts comparable to those achieved in young adult animals, alongside amplification with a single immunization. Associated enhanced dendritic cell and germinal center responses were observed. Together, these data demonstrate that an LNP-encapsulated IL-12-MOP mRNA-encoded adjuvant can amplify immunogenicity independent of age, demonstrating translational potential to benefit vulnerable populations.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Valerie Duval
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Cali Sweitzer
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Manisha Menon
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Pallab Ghosh
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Kimia Abedi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jacob Koster
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thomas VanCott
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Romain Micol
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Soni D, Borriello F, Scott DA, Feru F, DeLeon M, Brightman SE, Cheng WK, Melhem G, Smith JA, Ramirez JC, Barman S, Cameron M, Kelly A, Walker K, Nanishi E, van Haren SD, Phan T, Qi Y, Kinsey R, Raczy MM, Ozonoff A, Pettengill MA, Hubbell JA, Fox CB, Dowling DJ, Levy O. From hit to vial: Precision discovery and development of an imidazopyrimidine TLR7/8 agonist adjuvant formulation. SCIENCE ADVANCES 2024; 10:eadg3747. [PMID: 38959314 PMCID: PMC11221515 DOI: 10.1126/sciadv.adg3747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/29/2024] [Indexed: 07/05/2024]
Abstract
Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.
Collapse
Affiliation(s)
- Dheeraj Soni
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Francesco Borriello
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - David A. Scott
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Frederic Feru
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria DeLeon
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Spencer E. Brightman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Wing Ki Cheng
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Gandolina Melhem
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Juan C. Ramirez
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Aisling Kelly
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Kristina Walker
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Simon Daniel van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tony Phan
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Yizhi Qi
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Robert Kinsey
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Michal M. Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Matthew A. Pettengill
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jeffery A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Christopher B. Fox
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA 98195, USA
| | - David J. Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
17
|
Kawai A, Noda M, Hirata H, Munakata L, Matsuda T, Omata D, Takemura N, Onoe S, Hirose M, Kato T, Saitoh T, Hirai T, Suzuki R, Yoshioka Y. Lipid Nanoparticle with 1,2-Di-O-octadecenyl-3-trimethylammonium-propane as a Component Lipid Confers Potent Responses of Th1 Cells and Antibody against Vaccine Antigen. ACS NANO 2024; 18:16589-16609. [PMID: 38885198 PMCID: PMC11223497 DOI: 10.1021/acsnano.4c00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024]
Abstract
Adjuvants are effective tools to enhance vaccine efficacy and control the type of immune responses such as antibody and T helper 1 (Th1)- or Th2-type responses. Several studies suggest that interferon (IFN)-γ-producing Th1 cells play a significant role against infections caused by intracellular bacteria and viruses; however, only a few adjuvants can induce a strong Th1-type immune response. Recently, several studies have shown that lipid nanoparticles (LNPs) can be used as vaccine adjuvants and that each LNP has a different adjuvant activity. In this study, we screened LNPs to develop an adjuvant that can induce Th1 cells and antibodies using a conventional influenza split vaccine (SV) as an antigen in mice. We observed that LNP with 1,2-di-O-octadecenyl-3-trimethylammonium-propane (DOTMA) as a component lipid (DOTMA-LNP) elicited robust SV-specific IgG1 and IgG2 responses compared with SV alone in mice and was as efficient as SV adjuvanted with other adjuvants in mice. Furthermore, DOTMA-LNPs induced robust IFN-γ-producing Th1 cells without inflammatory responses compared to those of other adjuvants, which conferred strong cross-protection in mice. We also demonstrated the high versatility of DOTMA-LNP as a Th1 cell-inducing vaccine adjuvant using vaccine antigens derived from severe acute respiratory syndrome coronavirus 2 and Streptococcus pneumoniae. Our findings suggest the potential of DOTMA-LNP as a safe and effective Th1 cell-inducing adjuvant and show that LNP formulations are potentially potent adjuvants to enhance the effectiveness of other subunit vaccines.
Collapse
Affiliation(s)
- Atsushi Kawai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Noda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Haruki Hirata
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Lisa Munakata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Teppei Matsuda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Daiki Omata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Naoki Takemura
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sakura Onoe
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mika Hirose
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kato
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tatsuya Saitoh
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshiro Hirai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Suzuki
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Yasuo Yoshioka
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, The Research Foundation for Microbial Diseases of
Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
18
|
Liang Z, Bao H, Yao Z, Li M, Chen C, Zhang L, Wang H, Guo Y, Ma Y, Yang X, Yu G, Zhang J, Xue C, Sun B, Mao C. The orientation of CpG conjugation on aluminum oxyhydroxide nanoparticles determines the immunostimulatory effects of combination adjuvants. Biomaterials 2024; 308:122569. [PMID: 38626556 DOI: 10.1016/j.biomaterials.2024.122569] [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: 01/27/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/18/2024]
Abstract
In subunit vaccines, aluminum salts (Alum) are commonly used as adjuvants, but with limited cellular immune responses. To overcome this limitation, CpG oligodeoxynucleotides (ODNs) have been used in combination with Alum. However, current combined usage of Alum and CpG is limited to linear mixtures, and the underlying interaction mechanism between CpG and Alum is not well understood. Thus, we propose to chemically conjugate Alum nanoparticles and CpG (with 5' or 3' end exposed) to design combination adjuvants. Our study demonstrates that compared to the 3'-end exposure, the 5'-end exposure of CpG in combination adjuvants (Al-CpG-5') enhances the activation of bone-marrow derived dendritic cells (BMDCs) and promotes Th1 and Th2 cytokine secretion. We used the SARS-CoV-2 receptor binding domain (RBD) and hepatitis B surface antigen (HBsAg) as model antigens to demonstrate that Al-CpG-5' enhanced antigen-specific antibody production and upregulated cytotoxic T lymphocyte markers. Additionally, Al-CpG-5' allows for coordinated adaptive immune responses even at lower doses of both CpG ODNs and HBsAg antigens, and enhances lymph node transport of antigens and activation of dendritic cells, promoting Tfh cell differentiation and B cell activation. Our novel Alum-CPG strategy points the way towards broadening the use of nanoadjuvants for both prophylactic and therapeutic vaccines.
Collapse
Affiliation(s)
- Zhihui Liang
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China; Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, PR China
| | - Hang Bao
- School of Bioengineering, Dalian University of Technology, Dalian 116024, PR China
| | - Zhiying Yao
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Min Li
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Chen Chen
- School of Bioengineering, Dalian University of Technology, Dalian 116024, PR China
| | - Lei Zhang
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Huiyang Wang
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Yiyang Guo
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Yubin Ma
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Xuecheng Yang
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Ge Yu
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Jiancheng Zhang
- AIM Honesty Biopharmaceutical Co., Ltd, Dalian, 116100, PR China
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, Dalian 116024, PR China.
| | - Bingbing Sun
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China.
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, PR China.
| |
Collapse
|
19
|
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
|
20
|
Goetz M, Thotathil N, Zhao Z, Mitragotri S. Vaccine adjuvants for infectious disease in the clinic. Bioeng Transl Med 2024; 9:e10663. [PMID: 39036089 PMCID: PMC11256182 DOI: 10.1002/btm2.10663] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Adjuvants, materials added to vaccines to enhance the resulting immune response, are important components of vaccination that are many times overlooked. While vaccines always include an antigen to tell the body what to vaccinate to, of equal importance the adjuvant provides the how, a significant factor in producing a complete response. The adjuvant space has been slow to develop with the first use of an adjuvant in a licensed vaccine occurring in the 1930s, and remaining the only adjuvant in licensed vaccines for the next 80 years. However, with vaccination at the forefront of protection against new and complex pathogens, it is important to consider all components when designing an effective vaccine. Here we summarize the adjuvant space in licensed vaccines as well as the novel adjuvant space in clinical trials with a specific focus on the materials utilized and their resulting impact on the immune response. We discuss five major categories of adjuvant materials: aluminum salts, nanoparticles, viral vectors, TLR agonists, and emulsions. For each category, we delve into the current clinical trials space, the impact of these materials on vaccination, as well as some of the ways in which they could be improved. Adjuvants present an exciting opportunity to improve vaccine responses and stability, this review will help inform about the current progress of this space. Translational impact statement In the aftermath of the COVID-19 pandemic, vaccines for infectious diseases have come into the spotlight. While antigens have always been an important focus of vaccine design, the adjuvant is a significant tool for enhancing the immune response to the vaccine that has been largely underdeveloped. This article provides a broad review of the history of adjuvants and, the current vaccine adjuvant space, and the progress seen in adjuvants in clinical trials. There is specific emphasis on the material landscape for adjuvants and their resulting mechanism of action. Looking ahead, while the novel vaccine adjuvant space features exciting new technologies and materials, there is still a need for more to meet the protective needs of new and complex pathogens.
Collapse
Affiliation(s)
- Morgan Goetz
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Naaz Thotathil
- University of Massachusetts AmherstAmherstMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
21
|
Nurdin A, Movieta Nency Y, Maddeppungeng M, Sekartini R, Mulia Sari R, Surachman F, Fitry Yani F, Raveinal, Anggrainy F, Hafiz A, Linosefa, Machmud R, Awaliyah Deza P, Rujiana V, Bella Rahimi M, Farhanah N, Gundi Pramudo S, Hapsari R, Tri Anantyo D, Mulyono, Mahati E, Maharani N, Darma S, Husni Esa Darussalam A, Shakinah S, Nasrum Massi M, Soedjatmiko. Immunogenicity and safety of SARS-CoV-2 recombinant protein subunit vaccine (IndoVac) adjuvanted with alum and CpG 1018 in Indonesian adults: A phase 3, randomized, active-controlled, multicenter trial. Vaccine 2024; 42:3009-3017. [PMID: 38575433 DOI: 10.1016/j.vaccine.2024.03.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Bio Farma has developed a recombinant protein subunit vaccine (IndoVac) that is indicated for active immunization in population of all ages. This article reported the results of the phase 3 immunogenicity and safety study in Indonesian adults aged 18 years and above. METHODS We conducted a randomized, active-controlled, multicenter, prospective intervention study to evaluate the immunogenicity and safety of IndoVac in adults aged 18 years and above. Participants who were SARS-CoV-2 vaccine-naïve received two doses of either IndoVac or control (Covovax) with 28 days interval between doses and were followed up until 12 months after complete vaccination. RESULTS A total of 4050 participants were enrolled from June to August 2022 and received at least one dose of vaccine. The geometric mean ratio (GMR) of neutralizing antibody at 14 days after the second dose was 1.01 (95 % confidence interval (CI) 0.89-1.16), which met the WHO non-inferiority criteria for immunobridging (95 % CI lower bound > 0.67). The antibody levels were maintained through 12 months after the second dose. The incidence rate of adverse events (AEs) were 27.95 % in IndoVac group and 32.15 % in Covovax group with mostly mild intensity (27.70 %). The most reported solicited AEs were pain (14.69 %) followed by myalgia (7.48 %) and fatigue (6.77 %). Unsolicited AEs varied, with each of the incidence rate under 5 %. There were no serious AEs assessed as possibly, probably, or likely related to vaccine. CONCLUSIONS IndoVac in adults showed favourable safety profile and elicited non-inferior immune response to Covovax. (ClinicalTrials.gov: NCT05433285, Indonesian Clinical Research Registry: INA-R5752S9).
Collapse
Affiliation(s)
| | | | | | - Rini Sekartini
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | | | | | - Raveinal
- Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | | | - Al Hafiz
- Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Linosefa
- Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | | | | | | | | | - Nur Farhanah
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | | | | | | | - Mulyono
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Endang Mahati
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Nani Maharani
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Sidrah Darma
- Faculty of Medicine, Universitas Muslim Indonesia, Makassar, Indonesia
| | | | | | | | - Soedjatmiko
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
22
|
Hou Y, Chen M, Bian Y, Hu Y, Chuan J, Zhong L, Zhu Y, Tong R. Insights into vaccines for elderly individuals: from the impacts of immunosenescence to delivery strategies. NPJ Vaccines 2024; 9:77. [PMID: 38600250 PMCID: PMC11006855 DOI: 10.1038/s41541-024-00874-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Immunosenescence increases the risk and severity of diseases in elderly individuals and leads to impaired vaccine-induced immunity. With aging of the global population and the emerging risk of epidemics, developing adjuvants and vaccines for elderly individuals to improve their immune protection is pivotal for healthy aging worldwide. Deepening our understanding of the role of immunosenescence in vaccine efficacy could accelerate research focused on optimizing vaccine delivery for elderly individuals. In this review, we analyzed the characteristics of immunosenescence at the cellular and molecular levels. Strategies to improve vaccination potency in elderly individuals are summarized, including increasing the antigen dose, preparing multivalent antigen vaccines, adding appropriate adjuvants, inhibiting chronic inflammation, and inhibiting immunosenescence. We hope that this review can provide a review of new findings with regards to the impacts of immunosenescence on vaccine-mediated protection and inspire the development of individualized vaccines for elderly individuals.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yuan Hu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Junlan Chuan
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Zhong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Yuxuan Zhu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
23
|
Lavelle EC, McEntee CP. Vaccine adjuvants: Tailoring innate recognition to send the right message. Immunity 2024; 57:772-789. [PMID: 38599170 DOI: 10.1016/j.immuni.2024.03.015] [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: 01/19/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Adjuvants play pivotal roles in vaccine development, enhancing immunization efficacy through prolonged retention and sustained release of antigen, lymph node targeting, and regulation of dendritic cell activation. Adjuvant-induced activation of innate immunity is achieved via diverse mechanisms: for example, adjuvants can serve as direct ligands for pathogen recognition receptors or as inducers of cell stress and death, leading to the release of immunostimulatory-damage-associated molecular patterns. Adjuvant systems increasingly stimulate multiple innate pathways to induce greater potency. Increased understanding of the principles dictating adjuvant-induced innate immunity will subsequently lead to programming specific types of adaptive immune responses. This tailored optimization is fundamental to next-generation vaccines capable of inducing robust and sustained adaptive immune memory across different cohorts.
Collapse
Affiliation(s)
- Ed C Lavelle
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Craig P McEntee
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
24
|
Thomas S, Pak J, Doss-Gollin S, Ryff K, Beijnen E, Pedersen GK, Christensen D, Levy O, van Haren SD. Human In vitro Modeling Identifies Adjuvant Combinations that Unlock Antigen Cross-presentation and Promote T-helper 1 Development in Newborns, Adults and Elders. J Mol Biol 2024; 436:168446. [PMID: 38242283 PMCID: PMC10922990 DOI: 10.1016/j.jmb.2024.168446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/08/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Adjuvants are vaccine components that can boost the type, magnitude, breadth, and durability of an immune response. We have previously demonstrated that certain adjuvant combinations can act synergistically to enhance and shape immunogenicity including promotion of Th1 and cytotoxic T-cell development. These combinations also promoted protective immunity in vulnerable populations such as newborns. In this study, we employed combined antigen-specific human in vitro models to identify adjuvant combinations that could synergistically promote the expansion of vaccine-specific CD4+ cells, induce cross-presentation on MHC class I, resulting in antigen-specific activation of CD8+ cells, and direct the balance of immune response to favor the production of Th1-promoting cytokines. A screen of 78 adjuvant combinations identified several T cell-potentiating adjuvant combinations. Remarkably, a combination of TLR9 and STING agonists (CpG + 2,3-cGAMP) promoted influenza-specific CD4+ and CD8+ T cell activation and selectively favored production of Th1-polarizing cytokines TNF and IL-12p70 over co-regulated cytokines IL-6 and IL-12p40, respectively. Phenotypic reprogramming towards cDC1-type dendritic cells by CpG + 2,3-cGAMP was also observed. Finally, we characterized the molecular mechanism of this adjuvant combination including the ability of 2,3-cGAMP to enhance DC expression of TLR9 and the dependency of antigen-presenting cell activation on the Sec22b protein important to endoplasmic reticulum-Golgi vesicle trafficking. The identification of the adjuvant combination CpG + 2,3-cGAMP may therefore prove key to the future development of vaccines against respiratory viral infections tailored for the functionally distinct immune systems of vulnerable populations such as older adults and newborns.
Collapse
Affiliation(s)
- Sanya Thomas
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jensen Pak
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Simon Doss-Gollin
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Kevin Ryff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Elisabeth Beijnen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Kim E, Khan MS, Ferrari A, Huang S, Kenniston TW, Cassaniti I, Baldanti F, Gambotto A. Second Boost of Omicron SARS-CoV-2 S1 Subunit Vaccine Induced Broad Humoral Immune Responses in Elderly Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578925. [PMID: 38370806 PMCID: PMC10871204 DOI: 10.1101/2024.02.05.578925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Currently approved COVID-19 vaccines prevent symptomatic infection, hospitalization, and death from the disease. However, repeated homologous boosters, while considered a solution for severe forms of the disease caused by new SARS-CoV-2 variants in elderly individuals and immunocompromised patients, cannot provide complete protection against breakthrough infections. This highlights the need for alternative platforms for booster vaccines. In our previous study, we assessed the boost effect of the SARS-CoV-2 Beta S1 recombinant protein subunit vaccine (rS1Beta) in aged mice primed with an adenovirus-based vaccine expressing SARS-CoV-2-S1 (Ad5.S1) via subcutaneous injection or intranasal delivery, which induced robust humoral immune responses (1). In this follow-up study, we demonstrated that a second booster dose of a non-adjuvanted recombinant Omicron (BA.1) S1 subunit vaccine with Toll-like receptor 4 (TLR4) agonist RS09 (rS1RS09OM) was effective in stimulating strong S1-specific immune responses and inducing significantly high neutralizing antibodies against the Wuhan, Delta, and Omicron variants in 100-week-old mice. Importantly, the second booster dose elicits cross-reactive antibody responses, resulting in ACE2 binding inhibition against the spike protein of SARS-CoV-2 variants, including Omicron (BA.1) and its subvariants. Interestingly, the levels of IgG and neutralizing antibodies correlated with the level of ACE2 inhibition in the booster serum samples, although Omicron S1-specific IgG level showed a weaker correlation compared to Wuhan S1-specific IgG level. Furthermore, we compared the immunogenic properties of the rS1 subunit vaccine in young, middle-aged, and elderly mice, resulting in reduced immunogenicity with age, especially an impaired Th1-biased immune response in aged mice. Our findings demonstrate that the new variant of concern (VOC) rS1 subunit vaccine as a second booster has the potential to offer cross-neutralization against a broad range of variants and to improve vaccine effectiveness against newly emerging breakthrough SARS-CoV-2 variants in elderly individuals who were previously primed with the authorized vaccines.
Collapse
Affiliation(s)
- Eun Kim
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, PA 15213, USA
| | - Muhammad S. Khan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, PA 15213, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Alessandro Ferrari
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Shaohua Huang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, PA 15213, USA
| | - Thomas W. Kenniston
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, PA 15213, USA
| | - Irene Cassaniti
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, PA 15213, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
Zhang T, Magazine N, McGee MC, Carossino M, Veggiani G, Kousoulas KG, August A, Huang W. Th2 and Th17-associated immunopathology following SARS-CoV-2 breakthrough infection in Spike-vaccinated ACE2-humanized mice. J Med Virol 2024; 96:e29408. [PMID: 38258331 PMCID: PMC10832989 DOI: 10.1002/jmv.29408] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024]
Abstract
Vaccines have demonstrated remarkable effectiveness in protecting against COVID-19; however, concerns regarding vaccine-associated enhanced respiratory diseases (VAERD) following breakthrough infections have emerged. Spike protein subunit vaccines for SARS-CoV-2 induce VAERD in hamsters, where aluminum adjuvants promote a Th2-biased immune response, leading to increased type 2 pulmonary inflammation in animals with breakthrough infections. To gain a deeper understanding of the potential risks and the underlying mechanisms of VAERD, we immunized ACE2-humanized mice with SARS-CoV-2 Spike protein adjuvanted with aluminum and CpG-ODN. Subsequently, we exposed them to increasing doses of SARS-CoV-2 to establish a breakthrough infection. The vaccine elicited robust neutralizing antibody responses, reduced viral titers, and enhanced host survival. However, following a breakthrough infection, vaccinated animals exhibited severe pulmonary immunopathology, characterized by a significant perivascular infiltration of eosinophils and CD4+ T cells, along with increased expression of Th2/Th17 cytokines. Intracellular flow cytometric analysis revealed a systemic Th17 inflammatory response, particularly pronounced in the lungs. Our data demonstrate that aluminum/CpG adjuvants induce strong antibody and Th1-associated immunity against COVID-19 but also prime a robust Th2/Th17 inflammatory response, which may contribute to the rapid onset of T cell-mediated pulmonary immunopathology following a breakthrough infection. These findings underscore the necessity for further research to unravel the complexities of VAERD in COVID-19 and to enhance vaccine formulations for broad protection and maximum safety.
Collapse
Affiliation(s)
- Tianyi Zhang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Nicholas Magazine
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michael C. McGee
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Gianluca Veggiani
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Konstantin G. Kousoulas
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
27
|
O'Meara TR, Nanishi E, McGrath ME, Barman S, Dong D, Dillen C, Menon M, Seo HS, Dhe-Paganon S, Ernst RK, Levy O, Frieman MB, Dowling DJ. Reduced SARS-CoV-2 mRNA vaccine immunogenicity and protection in mice with diet-induced obesity and insulin resistance. J Allergy Clin Immunol 2023; 152:1107-1120.e6. [PMID: 37595760 PMCID: PMC10841117 DOI: 10.1016/j.jaci.2023.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND Obesity and type 2 diabetes mellitus (T2DM) are associated with an increased risk of severe outcomes from infectious diseases, including coronavirus disease 2019. These conditions are also associated with distinct responses to immunization, including an impaired response to widely used severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines. OBJECTIVE We sought to establish a connection between reduced immunization efficacy via modeling the effects of metabolic diseases on vaccine immunogenicity that is essential for the development of more effective vaccines for this distinct vulnerable population. METHODS A murine model of diet-induced obesity and insulin resistance was used to model the effects of comorbid T2DM and obesity on vaccine immunogenicity and protection. RESULTS Mice fed a high-fat diet (HFD) developed obesity, hyperinsulinemia, and glucose intolerance. Relative to mice fed a normal diet, HFD mice vaccinated with a SARS-CoV-2 mRNA vaccine exhibited significantly lower anti-spike IgG titers, predominantly in the IgG2c subclass, associated with a lower type 1 response, along with a 3.83-fold decrease in neutralizing titers. Furthermore, enhanced vaccine-induced spike-specific CD8+ T-cell activation and protection from lung infection against SARS-CoV-2 challenge were seen only in mice fed a normal diet but not in HFD mice. CONCLUSIONS The study demonstrated impaired immunity following SARS-CoV-2 mRNA immunization in a murine model of comorbid T2DM and obesity, supporting the need for further research into the basis for impaired anti-SARS-CoV-2 immunity in T2DM and investigation of novel approaches to enhance vaccine immunogenicity among those with metabolic diseases.
Collapse
Affiliation(s)
- Timothy R O'Meara
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass
| | - Etsuro Nanishi
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Marisa E McGrath
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Md
| | - Soumik Barman
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Danica Dong
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass
| | - Carly Dillen
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Md
| | - Manisha Menon
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Md
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass
| | - Matthew B Frieman
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Md
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
28
|
Doss-Gollin S, Thomas S, Brook B, Abedi K, Lebas C, Auderset F, Lugo-Rodriguez Y, Sanchez-Schmitz G, Dowling DJ, Levy O, van Haren SD. Human in vitro modeling of adjuvant formulations demonstrates enhancement of immune responses to SARS-CoV-2 antigen. NPJ Vaccines 2023; 8:163. [PMID: 37884538 PMCID: PMC10603059 DOI: 10.1038/s41541-023-00759-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Adjuvants can enhance vaccine immunogenicity, but their mechanism of action is often incompletely understood, hampering rapid applicability for pandemic vaccines. Herein, we characterized the cellular and molecular activity of adjuvant formulations available for pre-clinical evaluation, including several developed for global open access. We applied four complementary human in vitro platforms to assess individual and combined adjuvants in unformulated, oil-in-water, and liposomal delivery platforms. Liposomal co-formulation of MPLA and QS-21 was most potent in promoting dendritic cell maturation, selective production of Th1-polarizing cytokines, and activation of SARS-CoV-2 Spike-specific CD4+ and CD8+ T cells in a co-culture assay. Select formulations also significantly enhanced Spike antigen-specific humoral immunity in vivo. This study confirms the utility of the cumulative use of human in vitro tools to predict adjuvanticity potential. Thus, human in vitro modeling may advance public health by accelerating the development of affordable and scalable adjuvants for vaccines tailored to vulnerable populations.
Collapse
Affiliation(s)
- Simon Doss-Gollin
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Sanya Thomas
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Byron Brook
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kimia Abedi
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Célia Lebas
- Vaccine Formulation Institute, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Floriane Auderset
- Vaccine Formulation Institute, 1228 Plan-les-Ouates, Geneva, Switzerland
| | | | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
29
|
Zhang T, Magazine N, McGee MC, Carossino M, Veggiani G, Kousoulas KG, August A, Huang W. Th2 and Th17-Associated Immunopathology Following SARS-CoV-2 Breakthrough Infection in Spike-Vaccinated ACE2-humanized Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.563016. [PMID: 37904941 PMCID: PMC10614945 DOI: 10.1101/2023.10.18.563016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Vaccines have demonstrated remarkable effectiveness in protecting against COVID-19; however, concerns regarding vaccine-associated enhanced respiratory diseases (VAERD) following breakthrough infections have emerged. Spike protein subunit vaccines for SARS-CoV-2 induce VAERD in hamsters, where aluminum adjuvants promote a Th2-biased immune response, leading to increased type 2 pulmonary inflammation in animals with breakthrough infections. To gain a deeper understanding of the potential risks and the underlying mechanisms of VAERD, we immunized ACE2-humanized mice with SARS-CoV-2 Spike protein adjuvanted with aluminum and CpG-ODN. Subsequently, we exposed them to increasing doses of SARS-CoV-2 to establish a breakthrough infection. The vaccine elicited robust neutralizing antibody responses, reduced viral titers, and enhanced host survival. However, following a breakthrough infection, vaccinated animals exhibited severe pulmonary immunopathology, characterized by a significant perivascular infiltration of eosinophils and CD4+ T cells, along with increased expression of Th2/Th17 cytokines. Intracellular flow cytometric analysis revealed a systemic Th17 inflammatory response, particularly pronounced in the lungs. Our data demonstrate that aluminum/CpG adjuvants induce strong antibody and Th1-associated immunity against COVID-19 but also prime a robust Th2/Th17 inflammatory response, which may contribute to the rapid onset of T cell-mediated pulmonary immunopathology following a breakthrough infection. These findings underscore the necessity for further research to unravel the complexities of VAERD in COVID-19 and to enhance vaccine formulations for broad protection and maximum safety.
Collapse
Affiliation(s)
- Tianyi Zhang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Nicholas Magazine
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michael C. McGee
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Gianluca Veggiani
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Konstantin G. Kousoulas
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
30
|
Kumru OS, Bajoria S, Kaur K, Hickey JM, Van Slyke G, Doering J, Berman K, Richardson C, Lien H, Kleanthous H, Mantis NJ, Joshi SB, Volkin DB. Effects of aluminum-salt, CpG and emulsion adjuvants on the stability and immunogenicity of a virus-like particle displaying the SARS-CoV-2 receptor-binding domain (RBD). Hum Vaccin Immunother 2023; 19:2264594. [PMID: 37932241 PMCID: PMC10760504 DOI: 10.1080/21645515.2023.2264594] [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: 07/13/2023] [Accepted: 09/25/2023] [Indexed: 11/08/2023] Open
Abstract
Second-generation COVID-19 vaccines with improved immunogenicity (e.g., breadth, duration) and availability (e.g., lower costs, refrigerator stable) are needed to enhance global coverage. In this work, we formulated a clinical-stage SARS-CoV-2 receptor-binding domain (RBD) virus-like particle (VLP) vaccine candidate (IVX-411) with widely available adjuvants. Specifically, we assessed the in vitro storage stability and in vivo mouse immunogenicity of IVX-411 formulated with aluminum-salt adjuvants (Alhydrogel™, AH and Adjuphos™, AP), without or with the TLR-9 agonist CpG-1018™ (CpG), and compared these profiles to IVX-411 adjuvanted with an oil-in-water nano-emulsion (AddaVax™, AV). Although IVX-411 bound both AH and AP, lower binding strength of antigen to AP was observed by Langmuir binding isotherms. Interestingly, AH- and AP-adsorbed IVX-411 had similar storage stability profiles as measured by antigen-binding assays (competitive ELISAs), but the latter displayed higher pseudovirus neutralizing titers (pNT) in mice, at levels comparable to titers elicited by AV-adjuvanted IVX-411. CpG addition to alum (AP or AH) resulted in a marginal trend of improved pNTs in stressed samples only, yet did not impact the storage stability profiles of IVX-411. In contrast, previous work with AH-formulations of a monomeric RBD antigen showed greatly improved immunogenicity and decreased stability upon CpG addition to alum. At elevated temperatures (25, 37°C), IVX-411 formulated with AH or AP displayed decreased in vitro stability compared to AV-formulated IVX-411and this rank-ordering correlated with in vivo performance (mouse pNT values). This case study highlights the importance of characterizing antigen-adjuvant interactions to develop low cost, aluminum-salt adjuvanted recombinant subunit vaccine candidates.
Collapse
Affiliation(s)
- Ozan S. Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Kawaljit Kaur
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - John M. Hickey
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Greta Van Slyke
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Jennifer Doering
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Katherine Berman
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | | | | | - Harry Kleanthous
- Discovery & Translational Sciences, Global Health, Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - Nicholas J. Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
31
|
Zhao T, Cai Y, Jiang Y, He X, Wei Y, Yu Y, Tian X. Vaccine adjuvants: mechanisms and platforms. Signal Transduct Target Ther 2023; 8:283. [PMID: 37468460 PMCID: PMC10356842 DOI: 10.1038/s41392-023-01557-7] [Citation(s) in RCA: 281] [Impact Index Per Article: 140.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
Adjuvants are indispensable components of vaccines. Despite being widely used in vaccines, their action mechanisms are not yet clear. With a greater understanding of the mechanisms by which the innate immune response controls the antigen-specific response, the adjuvants' action mechanisms are beginning to be elucidated. Adjuvants can be categorized as immunostimulants and delivery systems. Immunostimulants are danger signal molecules that lead to the maturation and activation of antigen-presenting cells (APCs) by targeting Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs) to promote the production of antigen signals and co-stimulatory signals, which in turn enhance the adaptive immune responses. On the other hand, delivery systems are carrier materials that facilitate antigen presentation by prolonging the bioavailability of the loaded antigens, as well as targeting antigens to lymph nodes or APCs. The adjuvants' action mechanisms are systematically summarized at the beginning of this review. This is followed by an introduction of the mechanisms, properties, and progress of classical vaccine adjuvants. Furthermore, since some of the adjuvants under investigation exhibit greater immune activation potency than classical adjuvants, which could compensate for the deficiencies of classical adjuvants, a summary of the adjuvant platforms under investigation is subsequently presented. Notably, we highlight the different action mechanisms and immunological properties of these adjuvant platforms, which will provide a wide range of options for the rational design of different vaccines. On this basis, this review points out the development prospects of vaccine adjuvants and the problems that should be paid attention to in the future.
Collapse
Affiliation(s)
- Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yulong Cai
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yujie Jiang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yifan Yu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
32
|
Singh A, Boggiano C, Eller MA, Maciel M, Marovich MA, Mehra VL, Mo AX, Singleton KL, Leitner WW. Optimizing the Immunogenicity of HIV Vaccines by Adjuvants - NIAID Workshop Report. Vaccine 2023; 41:4439-4446. [PMID: 37331838 DOI: 10.1016/j.vaccine.2023.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 05/11/2023] [Accepted: 06/06/2023] [Indexed: 06/20/2023]
Abstract
This report summarizes the highlights of a workshop convened by the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), on April 4-5, 2022, to provide a discussion forum for sharing insights on the current status, key challenges, and next steps to advance the current landscape of promising adjuvants in preclinical and clinical human immunodeficiency virus (HIV) vaccine studies. A key goal was to solicit and share recommendations on scientific, regulatory, and operational guidelines for bridging the gaps in rational selection, access, and formulation of clinically relevant adjuvants for HIV vaccine candidates. The NIAID Vaccine Adjuvant Program working group remains committed to accentuate promising adjuvants and nurturing collaborations between adjuvant and HIV vaccine developers.
Collapse
Affiliation(s)
- Anjali Singh
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - César Boggiano
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael A Eller
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Milton Maciel
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mary A Marovich
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vijay L Mehra
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Annie X Mo
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kentner L Singleton
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wolfgang W Leitner
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Kwong KWY, Xin Y, Lai NCY, Sung JCC, Wu KC, Hamied YK, Sze ETP, Lam DMK. Oral Vaccines: A Better Future of Immunization. Vaccines (Basel) 2023; 11:1232. [PMID: 37515047 PMCID: PMC10383709 DOI: 10.3390/vaccines11071232] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Oral vaccines are gaining more attention due to their ease of administration, lower invasiveness, generally greater safety, and lower cost than injectable vaccines. This review introduces certified oral vaccines for adenovirus, recombinant protein-based, and transgenic plant-based oral vaccines, and their mechanisms for inducing an immune response. Procedures for regulatory approval and clinical trials of injectable and oral vaccines are also covered. Challenges such as instability and reduced efficacy in low-income countries associated with oral vaccines are discussed, as well as recent developments, such as Bacillus-subtilis-based and nanoparticle-based delivery systems that have the potential to improve the effectiveness of oral vaccines.
Collapse
Affiliation(s)
- Keith Wai-Yeung Kwong
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Ying Xin
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
| | - Nelson Cheuk-Yin Lai
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Johnny Chun-Chau Sung
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Kam-Chau Wu
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
| | | | - Eric Tung-Po Sze
- School of Science and Technology, Hong Kong Metropolitan University, Hong Kong, China
| | - Dominic Man-Kit Lam
- DrD Novel Vaccines Limited, Hong Kong, China
- Torsten Wiesel International Research Institute, Sichuan University, Chengdu 610064, China
| |
Collapse
|
34
|
Ben-Akiva E, Karlsson J, Hemmati S, Yu H, Tzeng SY, Pardoll DM, Green JJ. Biodegradable lipophilic polymeric mRNA nanoparticles for ligand-free targeting of splenic dendritic cells for cancer vaccination. Proc Natl Acad Sci U S A 2023; 120:e2301606120. [PMID: 37339211 PMCID: PMC10293809 DOI: 10.1073/pnas.2301606120] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
Nanoparticle (NP)-based mRNA cancer vaccines hold great promise to realize personalized cancer treatments. To advance this technology requires delivery formulations for efficient intracellular delivery to antigen-presenting cells. We developed a class of bioreducible lipophilic poly(beta-amino ester) nanocarriers with quadpolymer architecture. The platform is agnostic to the mRNA sequence, with one-step self-assembly allowing for delivery of multiple antigen-encoding mRNAs as well as codelivery of nucleic acid-based adjuvants. We examined structure-function relationships for NP-mediated mRNA delivery to dendritic cells (DCs) and identified that a lipid subunit of the polymer structure was critical. Following intravenous administration, the engineered NP design facilitated targeted delivery to the spleen and preferential transfection of DCs without the need for surface functionalization with targeting ligands. Treatment with engineered NPs codelivering antigen-encoding mRNA and toll-like receptor agonist adjuvants led to robust antigen-specific CD8+ T cell responses, resulting in efficient antitumor therapy in in vivo models of murine melanoma and colon adenocarcinoma.
Collapse
Affiliation(s)
- Elana Ben-Akiva
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD21287
| | - Johan Karlsson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Chemistry–Ångström Laboratory, Uppsala University, UppsalaSE-75121, Sweden
| | - Shayan Hemmati
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Hongzhe Yu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Drew M. Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD21287
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Jordan J. Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD21287
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science & Engineering, Johns Hopkins University, Baltimore, MD21231
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21231
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD21231
| |
Collapse
|
35
|
Lee B, Nanishi E, Levy O, Dowling DJ. Precision Vaccinology Approaches for the Development of Adjuvanted Vaccines Targeted to Distinct Vulnerable Populations. Pharmaceutics 2023; 15:1766. [PMID: 37376214 PMCID: PMC10305121 DOI: 10.3390/pharmaceutics15061766] [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: 05/04/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Infection persists as one of the leading global causes of morbidity and mortality, with particular burden at the extremes of age and in populations who are immunocompromised or suffer chronic co-morbid diseases. By focusing discovery and innovation efforts to better understand the phenotypic and mechanistic differences in the immune systems of diverse vulnerable populations, emerging research in precision vaccine discovery and development has explored how to optimize immunizations across the lifespan. Here, we focus on two key elements of precision vaccinology, as applied to epidemic/pandemic response and preparedness, including (a) selecting robust combinations of adjuvants and antigens, and (b) coupling these platforms with appropriate formulation systems. In this context, several considerations exist, including the intended goals of immunization (e.g., achieving immunogenicity versus lessening transmission), reducing the likelihood of adverse reactogenicity, and optimizing the route of administration. Each of these considerations is accompanied by several key challenges. On-going innovation in precision vaccinology will expand and target the arsenal of vaccine components for protection of vulnerable populations.
Collapse
Affiliation(s)
- Branden Lee
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
| | - Etsuro Nanishi
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David J. Dowling
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
36
|
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
|
37
|
Kim E, Khan MS, Ferrari A, Huang S, Sammartino JC, Percivalle E, Kenniston TW, Cassaniti I, Baldanti F, Gambotto A. SARS-CoV-2 S1 Subunit Booster Vaccination Elicits Robust Humoral Immune Responses in Aged Mice. Microbiol Spectr 2023; 11:e0436322. [PMID: 37162333 PMCID: PMC10269910 DOI: 10.1128/spectrum.04363-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/16/2023] [Indexed: 05/11/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has raised concerns about reduced vaccine effectiveness and the increased risk of infection, and while repeated homologous booster shots are recommended for elderly and immunocompromised individuals, they cannot completely protect against breakthrough infections. In our previous study, we assessed the immunogenicity of an adenovirus-based vaccine expressing SARS-CoV-2 S1 (Ad5.S1) in mice, which induced robust humoral and cellular immune responses (E. Kim, F. J. Weisel, S. C. Balmert, M. S. Khan, et al., Eur J Immunol 51:1774-1784, 2021, https://doi.org/10.1002/eji.202149167). In this follow-up study, we found that the mice had high titers of anti-S1 antibodies 1 year after vaccination, and one booster dose of the nonadjuvanted rS1Beta (recombinant S1 protein of SARS-CoV-2 Beta [B.1.351]) subunit vaccine was effective at stimulating strong long-lived S1-specific immune responses and inducing significantly high neutralizing antibodies against Wuhan, Beta, and Delta strains, with 3.6- to 19.5-fold increases. Importantly, the booster dose also elicited cross-reactive antibodies, resulting in angiotensin-converting enzyme 2 (ACE2) binding inhibition against spikes of SARS-CoV-2, including Omicron variants, persisting for >28 weeks after booster vaccination. Interestingly, the levels of neutralizing antibodies were correlated not only with the level of S1 binding IgG but also with ACE2 inhibition. Our findings suggest that the rS1Beta subunit vaccine candidate as a booster has the potential to offer cross-neutralization against broad variants and has important implications for the vaccine control of newly emerging breakthrough SARS-CoV-2 variants in elderly individuals primed with adenovirus-based vaccines like AZD1222 and Ad26.COV2.S. IMPORTANCE Vaccines have significantly reduced the incidences of severe coronavirus disease 2019 (COVID-19) cases and deaths. However, the emergence of SARS-CoV-2 variants has raised concerns about their increased transmissibility and ability to evade neutralizing antibodies, especially among elderly individuals who are at higher risks of mortality and reductions of vaccine effectiveness. To address this, a heterologous booster vaccination strategy has been considered as a solution to protect the elderly population against breakthrough infections caused by emerging variants. This study evaluated the booster effect of an S1 subunit vaccine in aged mice that had been previously primed with adenoviral vaccines, providing valuable preclinical evidence for elderly people vaccinated with the currently approved COVID-19 vaccines. This study confirms the potential for using the S1 subunit vaccine as a booster to enhance cross-neutralizing antibodies against emerging variants of concern.
Collapse
Affiliation(s)
- Eun Kim
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Muhammad S. Khan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Alessandro Ferrari
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Shaohua Huang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Josè C. Sammartino
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elena Percivalle
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Thomas W. Kenniston
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Irene Cassaniti
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, Division of Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
38
|
Afewerki S, Stocco TD, Rosa da Silva AD, Aguiar Furtado AS, Fernandes de Sousa G, Ruiz-Esparza GU, Webster TJ, Marciano FR, Strømme M, Zhang YS, Lobo AO. In vitro high-content tissue models to address precision medicine challenges. Mol Aspects Med 2023; 91:101108. [PMID: 35987701 PMCID: PMC9384546 DOI: 10.1016/j.mam.2022.101108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023]
Abstract
The field of precision medicine allows for tailor-made treatments specific to a patient and thereby improve the efficiency and accuracy of disease prevention, diagnosis, and treatment and at the same time would reduce the cost, redundant treatment, and side effects of current treatments. Here, the combination of organ-on-a-chip and bioprinting into engineering high-content in vitro tissue models is envisioned to address some precision medicine challenges. This strategy could be employed to tackle the current coronavirus disease 2019 (COVID-19), which has made a significant impact and paradigm shift in our society. Nevertheless, despite that vaccines against COVID-19 have been successfully developed and vaccination programs are already being deployed worldwide, it will likely require some time before it is available to everyone. Furthermore, there are still some uncertainties and lack of a full understanding of the virus as demonstrated in the high number new mutations arising worldwide and reinfections of already vaccinated individuals. To this end, efficient diagnostic tools and treatments are still urgently needed. In this context, the convergence of bioprinting and organ-on-a-chip technologies, either used alone or in combination, could possibly function as a prominent tool in addressing the current pandemic. This could enable facile advances of important tools, diagnostics, and better physiologically representative in vitro models specific to individuals allowing for faster and more accurate screening of therapeutics evaluating their efficacy and toxicity. This review will cover such technological advances and highlight what is needed for the field to mature for tackling the various needs for current and future pandemics as well as their relevancy towards precision medicine.
Collapse
Affiliation(s)
- Samson Afewerki
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Thiago Domingues Stocco
- Bioengineering Program, Technological and Scientific Institute, Brazil University, 08230-030, São Paulo, SP, Brazil; Faculty of Medical Sciences, Unicamp - State University of Campinas, 13083-877, Campinas, SP, Brazil
| | | | - André Sales Aguiar Furtado
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo Fernandes de Sousa
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Guillermo U Ruiz-Esparza
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA
| | - Thomas J Webster
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil; Hebei University of Technology, Tianjin, China
| | - Fernanda R Marciano
- Department of Physics, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Maria Strømme
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA.
| | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil.
| |
Collapse
|
39
|
Bai Y, An C, Zhang X, Li K, Cheng F, Cui B, Song Z, Liu D, Zhang J, He Q, Liu J, Mao Q, Liang Z. A Novel Targeted RIG-I Receptor 5'Triphosphate Double Strain RNA-Based Adjuvant Significantly Improves the Immunogenicity of the SARS-CoV-2 Delta-Omicron Chimeric RBD-Dimer Recombinant Protein Vaccine. Viruses 2023; 15:v15051099. [PMID: 37243185 DOI: 10.3390/v15051099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The rapid mutation and spread of SARS-CoV-2 variants recently, especially through the emerging variants Omicron BA5, BF7, XBB and BQ1, necessitate the development of universal vaccines to provide broad spectrum protection against variants. For the SARS-CoV-2 universal recombinant protein vaccines, an effective approach is necessary to design broad-spectrum antigens and combine them with novel adjuvants that can induce high immunogenicity. In this study, we designed a novel targeted retinoic acid-inducible gene-I (RIG-I) receptor 5'triphosphate double strain RNA (5'PPP dsRNA)-based vaccine adjuvant (named AT149) and combined it with the SARS-CoV-2 Delta and Omicron chimeric RBD-dimer recombinant protein (D-O RBD) to immunize mice. The results showed that AT149 activated the P65 NF-κB signaling pathway, which subsequently activated the interferon signal pathway by targeting the RIG-I receptor. The D-O RBD + AT149 and D-O RBD + aluminum hydroxide adjuvant (Al) + AT149 groups showed elevated levels of neutralizing antibodies against the authentic Delta variant, and Omicron subvariants, BA1, BA5, and BF7, pseudovirus BQ1.1, and XBB compared with D-O RBD + Al and D-O RBD + Al + CpG7909/Poly (I:C) groups at 14 d after the second immunization, respectively. In addition, D-O RBD + AT149 and D-O RBD + Al + AT149 groups presented higher levels of the T-cell-secreted IFN-γ immune response. Overall, we designed a novel targeted RIG-I receptor 5'PPP dsRNA-based vaccine adjuvant to significantly improve the immunogenicity and broad spectrum of the SARS-CoV-2 recombinant protein vaccine.
Collapse
Affiliation(s)
- Yu Bai
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Chaoqiang An
- Shanghai JunTuo Biotechnology Co., Ltd., Shanghai 201210, China
| | - Xuanxuan Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Kelei Li
- Beijing Minhai Biotechnology Co., Ltd., Beijning 102600, China
| | - Feiran Cheng
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Bopei Cui
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Ziyang Song
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Dong Liu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Jialu Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Qian He
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Jianyang Liu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Qunying Mao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| | - Zhenglun Liang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102600, China
| |
Collapse
|
40
|
Campbell E, Dobkin J, Osorio LJ, Kolloli A, Ramasamy S, Kumar R, Sant'Angelo DB, Subbian S, Denzin LK, Anderson S. A SARS-CoV-2 Vaccine Designed for Manufacturability Results in Unexpected Potency and Non-Waning Humoral Response. Vaccines (Basel) 2023; 11:vaccines11040832. [PMID: 37112744 PMCID: PMC10145385 DOI: 10.3390/vaccines11040832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The rapid development of several highly efficacious SARS-CoV-2 vaccines was an unprecedented scientific achievement that saved millions of lives. However, now that SARS-CoV-2 is transitioning to the endemic stage, there exists an unmet need for new vaccines that provide durable immunity and protection against variants and can be more easily manufactured and distributed. Here, we describe a novel protein component vaccine candidate, MT-001, based on a fragment of the SARS-CoV-2 spike protein that encompasses the receptor binding domain (RBD). Mice and hamsters immunized with a prime-boost regimen of MT-001 demonstrated extremely high anti-spike IgG titers, and remarkably this humoral response did not appreciably wane for up to 12 months following vaccination. Further, virus neutralization titers, including titers against variants such as Delta and Omicron BA.1, remained high without the requirement for subsequent boosting. MT-001 was designed for manufacturability and ease of distribution, and we demonstrate that these attributes are not inconsistent with a highly immunogenic vaccine that confers durable and broad immunity to SARS-CoV-2 and its emerging variants. These properties suggest MT-001 could be a valuable new addition to the toolbox of SARS-CoV-2 vaccines and other interventions to prevent infection and curtail additional morbidity and mortality from the ongoing worldwide pandemic.
Collapse
Affiliation(s)
- Elliot Campbell
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA
- Macrotope, Inc., Princeton, NJ 08540, USA
| | - Julie Dobkin
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Louis J Osorio
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Afsal Kolloli
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Santhamani Ramasamy
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Ranjeet Kumar
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Derek B Sant'Angelo
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Lisa K Denzin
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Stephen Anderson
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA
- Macrotope, Inc., Princeton, NJ 08540, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
41
|
Gary EN, Tursi NJ, Warner BM, Cuismano G, Connors J, Parzych EM, Griffin BD, Bell MR, Ali AR, Frase D, Hojecki CE, Canziani GA, Chaiken I, Kannan T, Moffat E, Embury-Hyatt C, Wooton SK, Kossenkov A, Patel A, Kobasa D, Kutzler MA, Haddad EK, Weiner DB. Adenosine deaminase augments SARS-CoV-2 specific cellular and humoral responses in aged mouse models of immunization and challenge. Front Immunol 2023; 14:1138609. [PMID: 36999023 PMCID: PMC10043169 DOI: 10.3389/fimmu.2023.1138609] [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: 01/05/2023] [Accepted: 02/23/2023] [Indexed: 03/16/2023] Open
Abstract
Despite numerous clinically available vaccines and therapeutics, aged patients remain at increased risk for COVID-19 morbidity. Furthermore, various patient populations, including the aged can have suboptimal responses to SARS-CoV-2 vaccine antigens. Here, we characterized vaccine-induced responses to SARS-CoV-2 synthetic DNA vaccine antigens in aged mice. Aged mice exhibited altered cellular responses, including decreased IFNγ secretion and increased TNFα and IL-4 secretion suggestive of TH2-skewed responses. Aged mice exhibited decreased total binding and neutralizing antibodies in their serum but significantly increased TH2-type antigen-specific IgG1 antibody compared to their young counterparts. Strategies to enhance vaccine-induced immune responses are important, especially in aged patient populations. We observed that co-immunization with plasmid-encoded adenosine deaminase (pADA)enhanced immune responses in young animals. Ageing is associated with decreases in ADA function and expression. Here, we report that co-immunization with pADA enhanced IFNγ secretion while decreasing TNFα and IL-4 secretion. pADA expanded the breadth and affinity SARS-CoV-2 spike-specific antibodies while supporting TH1-type humoral responses in aged mice. scRNAseq analysis of aged lymph nodes revealed that pADA co-immunization supported a TH1 gene profile and decreased FoxP3 gene expression. Upon challenge, pADA co-immunization decreased viral loads in aged mice. These data support the use of mice as a model for age-associated decreased vaccine immunogenicity and infection-mediated morbidity and mortality in the context of SARS-CoV-2 vaccines and provide support for the use of adenosine deaminase as a molecular adjuvant in immune-challenged populations.
Collapse
Affiliation(s)
- Ebony N. Gary
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Nicholas J. Tursi
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Bryce M. Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Gina Cuismano
- The Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jennifer Connors
- The Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Elizabeth M. Parzych
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Bryan D. Griffin
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Matthew R. Bell
- The Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ali R. Ali
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Drew Frase
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Casey E. Hojecki
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Gabriela A. Canziani
- The Department of Biochemistry, Drexel University college of Medicine, Philadelphia, PA, United States
| | - Irwin Chaiken
- The Department of Biochemistry, Drexel University college of Medicine, Philadelphia, PA, United States
| | - Toshitha Kannan
- The Genomics Core, The Wistar Institute, Philadelphia, PA, United States
| | - Estella Moffat
- National Center for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Carissa Embury-Hyatt
- National Center for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Sarah K. Wooton
- Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Andrew Kossenkov
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
- The Genomics Core, The Wistar Institute, Philadelphia, PA, United States
| | - Ami Patel
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Michele A. Kutzler
- The Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Elias K. Haddad
- The Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - David B. Weiner
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
42
|
He C, Chen L, Yang J, Chen Z, Lei H, Hong W, Song X, Yang L, Li J, Wang W, Shen G, Lu G, Wei X. Trimeric protein vaccine based on Beta variant elicits robust immune response against BA.4/5-included SARS-CoV-2 Omicron variants. MOLECULAR BIOMEDICINE 2023; 4:9. [PMID: 36894743 PMCID: PMC9998262 DOI: 10.1186/s43556-023-00121-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/07/2023] [Indexed: 03/11/2023] Open
Abstract
The current Coronavirus Disease 2019 (COVID-19) pandemic, induced by newly emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variants, posed great threats to global public health security. There is an urgent need to design effective next‑generation vaccines against Omicron lineages. Here, we investigated the immunogenic capacity of the vaccine candidate based on the receptor binding domain (RBD). An RBDβ-HR self-assembled trimer vaccine including RBD of Beta variant (containing K417, E484 and N501) and heptad repeat (HR) subunits was developed using an insect cell expression platform. Sera obtained from immunized mice effectively blocked RBD-human angiotensin-converting enzyme 2 (hACE2) binding for different viral variants, showing robust inhibitory activity. In addition, RBDβ-HR/trimer vaccine durably exhibited high titers of specific binding antibodies and high levels of cross-protective neutralizing antibodies against newly emerging Omicron lineages, as well as other major variants including Alpha, Beta, and Delta. Consistently, the vaccine also promoted a broad and potent cellular immune response involving the participation of T follicular helper (Tfh) cells, germinal center (GC) B cells, activated T cells, effector memory T cells, and central memory T cells, which are critical facets of protective immunity. These results demonstrated that RBDβ-HR/trimer vaccine candidates provided an attractive next-generation vaccine strategy against Omicron variants in the global effort to halt the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zimin Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Lei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiangrong Song
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiong Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guobo Shen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guangwen Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
43
|
Cao L, Guo J, Li H, Ren H, Xiao K, Zhang Y, Zhu S, Song Y, Zhao W, Wu D, Chen Z, Zhang Y, Xia B, Ji T, Yan D, Wang D, Yang Q, Zhou Y, Li X, Hou Z, Xu W. A Beta Strain-Based Spike Glycoprotein Vaccine Candidate Induces Broad Neutralization and Protection against SARS-CoV-2 Variants of Concern. Microbiol Spectr 2023; 11:e0268722. [PMID: 36847495 PMCID: PMC10100794 DOI: 10.1128/spectrum.02687-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 02/04/2023] [Indexed: 03/01/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is still ongoing. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) are circulating worldwide, making it resistant to existing vaccines and antiviral drugs. Therefore, the evaluation of variant-based expanded spectrum vaccines to optimize the immune response and provide broad protectiveness is very important. In this study, we expressed spike trimer protein (S-TM) based on the Beta variant in a GMP-grade workshop using CHO cells. Mice were immunized twice with S-TM protein combined with aluminum hydroxide (Al) and CpG Oligonucleotides (CpG) adjuvant to evaluate its safety and efficacy. BALB/c immunized with S-TM + Al + CpG induced high neutralizing antibody titers against the Wuhan-Hu-1 strain (wild-type, WT), the Beta and Delta variants, and even the Omicron variant. In addition, compared with the S-TM + Al group, the S-TM + Al + CpG group effectively induced a stronger Th1-biased cell immune response in mice. Furthermore, after the second immunization, H11-K18 hACE2 mice were well protected from challenge with the SARS-CoV-2 Beta strain, with a 100% survival rate. The virus load and pathological lesions in the lungs were significantly reduced, and no virus was detected in mouse brain tissue. Our vaccine candidate is practical and effective for current SARS-CoV-2 VOCs, which will support its further clinical development for potential sequential immune and primary immunization. IMPORTANCE Continuous emergence of adaptive mutations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to challenge the use and development of existing vaccines and drugs. The value of variant-based vaccines that are capable of inducing a higher and broader protection immune response against SARS-CoV-2 variants is currently being evaluated. This article shows that a recombinant prefusion spike protein based on a Beta variant was highly immunogenic and could induced a stronger Th1-biased cell immune response in mice and was effectively protective against challenge with the SARS-CoV-2 Beta variant. Importantly, this Beta-based SARS-CoV-2 vaccine could also offer a robust humoral immune response with effectively broad neutralization ability against the wild type and different variants of concern (VOCs): the Beta, Delta, and Omicron BA.1 variants. To date, the vaccine described here has been produced in a pilot scale (200L), and the development, filling process, and toxicological safety evaluation have also been completed, which provides a timely response to the emerging SARS-CoV-2 variants and vaccine development.
Collapse
Affiliation(s)
- Lei Cao
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jinyuan Guo
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hai Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hu Ren
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuangli Zhu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yang Song
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Weijia Zhao
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Dan Wu
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Zhihui Chen
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Yanan Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Baicheng Xia
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Tianjiao Ji
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dongmei Yan
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dongyan Wang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qian Yang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yangzi Zhou
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaolei Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhanjun Hou
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Wenbo Xu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| |
Collapse
|
44
|
Nanishi E, Borriello F, Seo HS, O’Meara TR, McGrath ME, Saito Y, Chen J, Diray-Arce J, Song K, Xu AZ, Barman S, Menon M, Dong D, Caradonna TM, Feldman J, Hauser BM, Schmidt AG, Baden LR, Ernst RK, Dillen C, Yu J, Chang A, Hilgers L, Platenburg PP, Dhe-Paganon S, Barouch DH, Ozonoff A, Zanoni I, Frieman MB, Dowling DJ, Levy O. Carbohydrate fatty acid monosulphate: oil-in-water adjuvant enhances SARS-CoV-2 RBD nanoparticle-induced immunogenicity and protection in mice. NPJ Vaccines 2023; 8:18. [PMID: 36788219 PMCID: PMC9927065 DOI: 10.1038/s41541-023-00610-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
Development of SARS-CoV-2 vaccines that protect vulnerable populations is a public health priority. Here, we took a systematic and iterative approach by testing several adjuvants and SARS-CoV-2 antigens to identify a combination that elicits antibodies and protection in young and aged mice. While demonstrating superior immunogenicity to soluble receptor-binding domain (RBD), RBD displayed as a protein nanoparticle (RBD-NP) generated limited antibody responses. Comparison of multiple adjuvants including AddaVax, AddaS03, and AS01B in young and aged mice demonstrated that an oil-in-water emulsion containing carbohydrate fatty acid monosulphate derivative (CMS:O/W) most effectively enhanced RBD-NP-induced cross-neutralizing antibodies and protection across age groups. CMS:O/W enhanced antigen retention in the draining lymph node, induced injection site, and lymph node cytokines, with CMS inducing MyD88-dependent Th1 cytokine polarization. Furthermore, CMS and O/W synergistically induced chemokine production from human PBMCs. Overall, CMS:O/W adjuvant may enhance immunogenicity and protection of vulnerable populations against SARS-CoV-2 and other infectious pathogens.
Collapse
Affiliation(s)
- Etsuro Nanishi
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Francesco Borriello
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA ,grid.2515.30000 0004 0378 8438Division of Immunology, Boston Children’s Hospital, Boston, MA USA ,Present Address: Generate Biomedicines, Cambridge, MA USA
| | - Hyuk-Soo Seo
- grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA USA
| | - Timothy R. O’Meara
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA
| | - Marisa E. McGrath
- grid.411024.20000 0001 2175 4264Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, MD USA
| | - Yoshine Saito
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA
| | - Jing Chen
- grid.2515.30000 0004 0378 8438Research Computing Group, Boston Children’s Hospital, Boston, MA USA
| | - Joann Diray-Arce
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Kijun Song
- grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Andrew Z. Xu
- grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Soumik Barman
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Manisha Menon
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA
| | - Danica Dong
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA
| | - Timothy M. Caradonna
- grid.461656.60000 0004 0489 3491Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA USA
| | - Jared Feldman
- grid.461656.60000 0004 0489 3491Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA USA
| | - Blake M. Hauser
- grid.461656.60000 0004 0489 3491Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA USA
| | - Aaron G. Schmidt
- grid.461656.60000 0004 0489 3491Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA USA ,grid.38142.3c000000041936754XDepartment of Microbiology, Harvard Medical School, Boston, MA USA
| | - Lindsey R. Baden
- grid.62560.370000 0004 0378 8294Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Robert K. Ernst
- grid.411024.20000 0001 2175 4264Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD USA
| | - Carly Dillen
- grid.411024.20000 0001 2175 4264Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, MD USA
| | - Jingyou Yu
- grid.38142.3c000000041936754XCenter for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Aiquan Chang
- grid.38142.3c000000041936754XCenter for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | | | | | - Sirano Dhe-Paganon
- grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA USA
| | - Dan H. Barouch
- grid.38142.3c000000041936754XCenter for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Al Ozonoff
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA ,grid.66859.340000 0004 0546 1623Broad Institute of MIT & Harvard, Cambridge, MA USA
| | - Ivan Zanoni
- grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA ,grid.2515.30000 0004 0378 8438Division of Immunology, Boston Children’s Hospital, Boston, MA USA
| | - Matthew B. Frieman
- grid.411024.20000 0001 2175 4264Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, MD USA
| | - David J. Dowling
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA. .,Department of Pediatrics, Harvard Medical School, Boston, MA, USA. .,Broad Institute of MIT & Harvard, Cambridge, MA, USA.
| |
Collapse
|
45
|
Germanó MJ, Giai C, Cargnelutti DE, Colombo MI, Blanco S, Konigheim B, Spinsanti L, Aguilar J, Gallego S, Valdez HA, Mackern-Oberti JP, Sanchez MV. Receptor-binding domain-based SARS-CoV-2 vaccine adjuvanted with cyclic di-adenosine monophosphate enhances humoral and cellular immunity in mice. J Med Virol 2023; 95:e28584. [PMID: 36794675 DOI: 10.1002/jmv.28584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/25/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
Novel adjuvants are highly desired to improve immune responses of SARS-CoV-2 vaccines. This work reports the potential of the stimulator of interferon genes (STING) agonist adjuvant, the cyclic di-adenosine monophosphate (c-di-AMP), in a SARS-CoV-2 vaccine based on the receptor binding domain (RBD). Here, mice immunized with two doses of monomeric RBD adjuvanted with c-di-AMP intramuscularly were found to exhibit stronger immune responses compared to mice vaccinated with RBD adjuvanted with aluminum hydroxide (Al(OH)3 ) or without adjuvant. After two immunizations, consistent enhancements in the magnitude of RBD-specific immunoglobulin G (IgG) antibody response were observed by RBD + c-di-AMP (mean: 15360) compared to RBD + Al(OH)3 (mean: 3280) and RBD alone (n.d.). Analysis of IgG subtypes indicated a predominantly Th1-biased immune response (IgG2c, mean: 14480; IgG2b, mean: 1040, IgG1, mean: 470) in mice vaccinated with RBD + c-di-AMP compared to a Th2-biased response in those vaccinated with RBD + Al(OH)3 (IgG2c, mean: 60; IgG2b: n.d.; IgG1, mean: 16660). In addition, the RBD + c-di-AMP group showed better neutralizing antibody responses as determined by pseudovirus neutralization assay and by plaque reduction neutralization assay with SARS-CoV-2 wild type. Moreover, the RBD + c-di-AMP vaccine promoted interferon-γ secretion of spleen cell cultures after RBD stimulation. Furthermore, evaluation of IgG-antibody titers in aged mice showed that di-AMP was able to improve RBD-immunogenicity at old age after 3 doses (mean: 4000). These data suggest that c-di-AMP improves immune responses of a SARS-CoV-2 vaccine based on RBD, and would be considered a promising option for future COVID-19 vaccines.
Collapse
Affiliation(s)
- María José Germanó
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Constanza Giai
- Instituto de Histología y Embriología de Mendoza, CONICET-Mendoza, Universidad Nacional de Cuyo-(UNCuyo) CONICET, Mendoza, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Esteban Cargnelutti
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Isabel Colombo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Histología y Embriología de Mendoza, CONICET-Mendoza, Universidad Nacional de Cuyo-(UNCuyo) CONICET, Mendoza, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Sebastián Blanco
- Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Brenda Konigheim
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lorena Spinsanti
- Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Javier Aguilar
- Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sandra Gallego
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Hugo Alberto Valdez
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Juan Pablo Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Maria Victoria Sanchez
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
46
|
DNA Oligonucleotides as Antivirals and Vaccine Constituents against SARS Coronaviruses: A Prospective Tool for Immune System Tuning. Int J Mol Sci 2023; 24:ijms24021553. [PMID: 36675069 PMCID: PMC9862924 DOI: 10.3390/ijms24021553] [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: 11/15/2022] [Revised: 12/26/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The SARS-CoV-2 pandemic has demonstrated the need to create highly effective antivirals and vaccines against various RNA viruses, including SARS coronaviruses. This paper provides a short review of innovative strategies in the development of antivirals and vaccines against SARS coronaviruses, with a focus on antisense antivirals, oligonucleotide adjuvants in vaccines, and oligonucleotide vaccines. Well-developed viral genomic databases create new opportunities for the development of innovative vaccines and antivirals using a post-genomic platform. The most effective vaccines against SARS coronaviruses are those able to form highly effective memory cells for both humoral and cellular immunity. The most effective antivirals need to efficiently stop viral replication without side effects. Oligonucleotide antivirals and vaccines can resist the rapidly changing genomic sequences of SARS coronaviruses using conserved regions of their genomes to generate a long-term immune response. Oligonucleotides have been used as excellent adjuvants for decades, and increasing data show that oligonucleotides could serve as antisense antivirals and antigens in vaccine formulations, becoming a prospective tool for immune system tuning.
Collapse
|
47
|
Ontiveros-Padilla L, Batty CJ, Hendy DA, Pena ES, Roque JA, Stiepel RT, Carlock MA, Simpson SR, Ross TM, Abraham SN, Staats HF, Bachelder EM, Ainslie KM. Development of a broadly active influenza intranasal vaccine adjuvanted with self-assembled particles composed of mastoparan-7 and CpG. Front Immunol 2023; 14:1103765. [PMID: 37033992 PMCID: PMC10081679 DOI: 10.3389/fimmu.2023.1103765] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Currently licensed vaccine adjuvants offer limited mucosal immunity, which is needed to better combat respiratory infections such as influenza. Mast cells (MCs) are emerging as a target for a new class of mucosal vaccine adjuvants. Here, we developed and characterized a nanoparticulate adjuvant composed of an MC activator [mastoparan-7 (M7)] and a TLR ligand (CpG). This novel nanoparticle (NP) adjuvant was co-formulated with a computationally optimized broadly reactive antigen (COBRA) for hemagglutinin (HA), which is broadly reactive against influenza strains. M7 was combined at different ratios with CpG and tested for in vitro immune responses and cytotoxicity. We observed significantly higher cytokine production in dendritic cells and MCs with the lowest cytotoxicity at a charge-neutralizing ratio of nitrogen/phosphate = 1 for M7 and CpG. This combination formed spherical NPs approximately 200 nm in diameter with self-assembling capacity. Mice were vaccinated intranasally with COBRA HA and M7-CpG NPs in a prime-boost-boost schedule. Vaccinated mice had significantly higher antigen-specific antibody responses (IgG and IgA) in serum and mucosa compared with controls. Splenocytes from vaccinated mice had significantly increased cytokine production upon antigen recall and the presence of central and effector memory T cells in draining lymph nodes. Finally, co-immunization with NPs and COBRA HA induced influenza H3N2-specific HA inhibition antibody titers across multiple strains and partially protected mice from a challenge against an H3N2 virus. These results illustrate that the M7-CpG NP adjuvant combination can induce a protective immune response with a broadly reactive influenza antigen via mucosal vaccination.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Erik S. Pena
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
| | - John A. Roque
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rebeca T. Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael A. Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Sean R. Simpson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ted M. Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Soman N. Abraham
- Departments of Pathology, Molecular Genetics and Microbiology and Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Herman F. Staats
- Department of Pathology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccines Institute, School of Medicine, Duke University, Durham, NC, United States
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Kristy M. Ainslie,
| |
Collapse
|
48
|
Gao Y, Wang W, Yang Y, Zhao Q, Yang C, Jia X, Liu Y, Zhou M, Zeng W, Huang X, Chiu S, Jin T, Wu X. Developing Next-Generation Protein-Based Vaccines Using High-Affinity Glycan Ligand-Decorated Glyconanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204598. [PMID: 36398611 PMCID: PMC9839878 DOI: 10.1002/advs.202204598] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Major diseases, such as cancer and COVID-19, are frightening global health problems, and sustained action is necessary to develop vaccines. Here, for the first time, ethoxy acetalated dextran nanoparticles (Ace-Dex-NPs) are functionalized with 9-N-(4H-thieno[3,2-c]chromene-2-carbamoyl)-Siaα2-3Galβ1-4GlcNAc (TCC Sia-LacNAc) targeting macrophages as a universal vaccine design platform. First, azide-containing oxidized Ace-Dex-NPs are synthesized. After the NPs are conjugated with ovalbumin (OVA) and resiquimod (Rd), they are coupled to TCC Sia-LacNAc-DBCO to produce TCC Sia-Ace-Dex-OVA-Rd, which induce a potent, long-lasting OVA-specific cytotoxic T-lymphocyte (CTL) response and high anti-OVA IgG, providing mice with superior protection against tumors. Next, this strategy is exploited to develop vaccines against infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein is the main target for neutralizing antibodies. The TCC Sia-Ace-Dex platform is preferentially used for designing an RBD-based vaccine. Strikingly, the synthetic TCC Sia-Ace-Dex-RBD-Rd elicited potent RBD-neutralizing antibodies against live SARS-CoV-2 infected Vero E6 cells. To develop a universal SARS-CoV-2 vaccine, the TCC Sia-Ace-Dex-N-Rd vaccine carrying SARS-CoV-2 nucleocapsid protein (N) is also prepared, which is highly conserved among SARS-CoV-2 and its variants of concern (VOCs), including Omicron (BA.1 to BA.5); this vaccine can trigger strong N-specific CTL responses against target cells infected with SARS-CoV-2 and its VOCs.
Collapse
Affiliation(s)
- Yanan Gao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Wei Wang
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Yunru Yang
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Qingyu Zhao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Chendong Yang
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Xiaoying Jia
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Yang Liu
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
| | - Minmin Zhou
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Weihong Zeng
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical EngineeringInstitute for Quantitative Health Science and EngineeringMichigan State UniversityEast LansingMichigan48824United States
| | - Sandra Chiu
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Tengchuan Jin
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Xuanjun Wu
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
- Suzhou Research InstituteShandong UniversitySuzhouJiangsu215123China
| |
Collapse
|
49
|
Chen YF, Wang Y, Wang Y, Luo YL, Lu ZD, Du XJ, Xu CF, Wang J. Optimized Cationic Lipid-assisted Nanoparticle for Delivering CpG Oligodeoxynucleotides to Treat Hepatitis B Virus Infection. Pharm Res 2023; 40:145-156. [PMID: 36002611 DOI: 10.1007/s11095-022-03307-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/25/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Hepatitis B virus (HBV) infection is such a global health problem that hundreds of millions of people are HBV carriers. Current anti-viral agents can inhibit HBV replication, but can hardly eradicate HBV. Cytosine-phosphate-guanosine (CpG) oligodeoxynucleotides (ODNs) are an adjuvant that can activate plasmacytoid dendritic cells (pDCs) and conventional dendritic cells (cDCs) to induce therapeutic immunity for HBV eradication. However, efficient delivery of CpG ODNs into pDCs and cDCs remains a challenge. In this study, we constructed a series of cationic lipid-assisted nanoparticles (CLANs) using different cationic lipids to screen an optimal nanoparticle for delivering CpG ODNs into pDCs and cDCs. METHODS We constructed different CLANCpG using six cationic lipids and analyzed the cellular uptake of different CLANCpG by pDCs and cDCs in vitro and in vivo, and further analyzed the efficiency of different CLANCpG for activating pDCs and cDCs in both wild type mice and HBV-carrier mice. RESULTS We found that CLAN fabricated with 1,2-Dioleoyl-3-trimethylammonium propane (DOTAP) showed the highest efficiency for delivering CpG ODNs into pDCs and cDCs, resulting in strong therapeutic immunity in HBV-carrier mice. By using CLANCpG as an immune adjuvant in combination with the injection of recombinant hepatitis B surface antigen (rHBsAg), HBV was successfully eradicated and the chronic liver inflammation in HBV-carrier mice was reduced. CONCLUSION We screened an optimized CLAN fabricated with DOTAP for efficient delivery of CpG ODNs to pDCs and cDCs, which can act as a therapeutic vaccine adjuvant for treating HBV infection.
Collapse
Affiliation(s)
- Yi-Fang Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, People's Republic of China
| | - Yan Wang
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Yue Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, People's Republic of China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Ying-Li Luo
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Zi-Dong Lu
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Xiao-Jiao Du
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, People's Republic of China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, People's Republic of China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.,Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China
| |
Collapse
|
50
|
Zeng Y, Zou F, Xia N, Li S. In-depth review of delivery carriers associated with vaccine adjuvants: current status and future perspectives. Expert Rev Vaccines 2023; 22:681-695. [PMID: 37496496 DOI: 10.1080/14760584.2023.2238807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Vaccines are powerful tools for controlling microbial infections and preventing epidemics. To enhance the immune response to antigens, effective subunit vaccines or mRNA vaccines often require the combination of adjuvants or delivery carriers. In recent years, with the rapid development of immune mechanism research and nanotechnology, various studies based on the optimization of traditional adjuvants or various novel carriers have been intensified, and the construction of vaccine adjuvant delivery systems (VADS) with both adjuvant activity and antigen delivery has become more and more important in vaccine research. AREAS COVERED This paper reviews the common types of vaccine adjuvant delivery carriers, classifies the VADS according to their basic carrier types, introduces the current research status and future development trend, and emphasizes the important role of VADS in novel vaccine research. EXPERT OPINION As the number of vaccine types increases, conventional aluminum adjuvants show limitations in effectively stimulating cellular immune responses, limiting their use in therapeutic vaccines for intracellular infections or tumors. In contrast, the use of conventional adjuvants as VADS to carry immunostimulatory molecules or deliver antigens can greatly enhance the immune boosting effect of classical adjuvants. A comprehensive understanding of the various delivery vehicles will further facilitate the development of vaccine adjuvant research.
Collapse
Affiliation(s)
- Yarong Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Feihong Zou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
- The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| |
Collapse
|