1
|
Coish JM, MacNeil LA, MacNeil AJ. The SARS-CoV-2 antibody-dependent enhancement façade. Microbes Infect 2025; 27:105464. [PMID: 39662700 DOI: 10.1016/j.micinf.2024.105464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Antibody-dependent enhancement (ADE) is an immunological paradox whereby sensitization following a primary viral infection results in the subsequent enhancement of a similar secondary infection. This idiosyncratic immune response has been established in dengue virus infections, driven by four antigenically related serotypes co-circulating in endemic regions. Several coronaviruses exhibit antibody-mediated mechanisms of viral entry, which has led to speculation of an ADE capacity for SARS-CoV-2, though in vivo and epidemiological evidence do not currently support this phenomenon. Three distinct antibody-dependent mechanisms for SARS-CoV-2 entry have recently been demonstrated: 1. FcR-dependent, 2. ACE2-FcR-interdependent, and 3. FcR-independent. These mechanisms of viral entry may be dependent on SARS-CoV-2 antibody specificity; antibodies targeting the receptor binding domain (RBD) typically result in Fc-dependent and ACE2-FcR-interdependent entry, whereas antibodies targeting the N-terminal domain can induce a conformational change to the RBD that optimizes ACE2-receptor binding domain interactions independent of Fc receptors. Whether these antibody-dependent entry mechanisms of SARS-CoV-2 result in the generation of infectious progenies and enhancement of infection has not been robustly demonstrated. Furthermore, ADE of SARS-CoV-2 mediated by antigenic seniority remains a theoretical concern, as no evidence suggests that SARS-CoV-2 imprinting blunts a subsequent immune response, contributing to severe COVID-19 disease.
Collapse
Affiliation(s)
- Jeremia M Coish
- Department of Health Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| | - Lori A MacNeil
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada.
| |
Collapse
|
2
|
Tao T, Tian L, Ke J, Zhang C, Li M, Xu X, Fan J, Tong Y, Fan H. Antibody-dependent enhancement of coronaviruses. Int J Biol Sci 2025; 21:1686-1704. [PMID: 39990674 PMCID: PMC11844293 DOI: 10.7150/ijbs.96112] [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: 03/08/2024] [Accepted: 01/11/2025] [Indexed: 02/25/2025] Open
Abstract
The COVID-19 pandemic presents a significant challenge to the global health and the world economy, with humanity engaged in an extended struggle against the virus. Notable advancements have been achieved in the development of vaccines and therapeutic interventions, including the application of neutralizing antibodies (NAbs) and convalescent plasma (CP). While antibody-dependent enhancement (ADE) has not been observed in human clinical studies related to SARS-CoV-2, the potential for ADE remains a critical concern and challenge in addressing SARS-CoV-2 infections. Moreover, the causal relationship between ADE and viral characteristics remains to be clearly elucidated. Viruses that present with severe clinical manifestations of ADE have demonstrated the capacity to replicate in macrophages or other immune cells, or to alter the immunological status of these cells, which induces abortive infections characterized by systemic inflammation. In this review, we summarize experimental observations and clinical evidence concerning the ADE effect associated with coronaviruses. We critically examine the potential mechanisms through which coronaviruses mediate ADE, and propose strategies to mitigate this phenomenon in the context of viral infection treatment. Our aim is to offer informed recommendations for the containment of the COVID-19 pandemic and to strengthen the response to SARS-CoV-2, as well as to prepare for potential future coronavirus threats.
Collapse
Affiliation(s)
- Tao Tao
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiayi Ke
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Chuxie Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Huahao Fan
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| |
Collapse
|
3
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
4
|
Rathore D, Chauhan P, Bonagiri A, Gandhi L, Maisnam D, Kumar R, Row AT, Kesavulu MM, Venkataramana M. Non-RBD peptides of SARS-CoV-2 spike protein exhibit immunodominance as they elicit both innate and adaptive immune responses. Heliyon 2024; 10:e39941. [PMID: 39568852 PMCID: PMC11577203 DOI: 10.1016/j.heliyon.2024.e39941] [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: 10/02/2023] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Severe acute respiratory coronavirus-2 (SARS-CoV-2) emerged in 2019 as a new virus and caused worldwide outbreaks, quickly turning into a pandemic disease called coronavirus disease-19 (COVID-19). All the existing methodologies were used for developing vaccines for this virus. But sporadic infections of this virus and the emergence of new strains to date suggest the incomplete protection offered by the developed vaccines and the need for new research. In this direction, we identified five epitopes present in the non-RBD region and on the surface of the spike protein by in silico analysis. They are epitope I (aa 80-90), epitope II (aa 262-270), and a small protein with three epitopes (aa 1059-1124). Antigenicity scores of these epitopes were found to be higher than the full length spike protein and its RBD region. These epitopes showed high conserveness across the emerging strains, high immunogenicity, non-toxicity, no homology with human sequences and high affinity for MHC class I & II molecules. Antibodies raised against these epitopes interacted with the bacterially expressed spike protein in western blotting. The antiserum of COVID-19 recovered participants reacted with the developed epitopes (small protein). Furthermore, in the presence of the respective antiserum and COVID-19 convalescent serum, these epitopes successfully fixed the complement, implying a possible role in innate immunity. The epitopes were also found to activate the peripheral blood mononuclear cells (PBMCs) isolated from the blood samples of COVID-19 recovered/vaccinated participants, suggesting a possible role in adaptive immunity. The need for the new SARS-CoV-2 vaccines is further highlighted in light of current reports about the side effects of a developed vaccine (AstraZeneca) and the circulating new strains. The epitopes presented in this study represent the potential immunogens and expect certain pitfalls of the existing vaccines would be sealed.
Collapse
Affiliation(s)
- Deepika Rathore
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| | - Preeti Chauhan
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| | - Anvesh Bonagiri
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| | - Lekha Gandhi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| | - Deepti Maisnam
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| | - Ramesh Kumar
- Health Centre, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| | - Anupama T Row
- Health Centre, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| | - M M Kesavulu
- Department of Basic Sciences and Humanities, Sree Vidyanikethan Engineering College, Tirupati, Andhra Pradesh, India
| | - Musturi Venkataramana
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, 500046, Hyderabad, Telangana State, India
| |
Collapse
|
5
|
Feng Q, Cheng K, Zhang L, Wang D, Gao X, Liang J, Liu G, Ma N, Xu C, Tang M, Chen L, Wang X, Ma X, Zou J, Shi Q, Du P, Wang Q, Wang H, Nie G, Zhao X. Rationally designed multimeric nanovaccines using icosahedral DNA origami for display of SARS-CoV-2 receptor binding domain. Nat Commun 2024; 15:9581. [PMID: 39505890 PMCID: PMC11542012 DOI: 10.1038/s41467-024-53937-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Multivalent antigen display on nanoparticles can enhance the immunogenicity of nanovaccines targeting viral moieties, such as the receptor binding domain (RBD) of SARS-CoV-2. However, particle morphology and size of current nanovaccines are significantly different from those of SARS-CoV-2. Additionally, surface antigen patterns are not controllable to enable the optimization of B cell activation. Herein, we employ an icosahedral DNA origami (ICO) as a display particle for RBD nanovaccines, achieving morphology and diameter like the virus (91 ± 11 nm). The surface addressability of DNA origami permits facile modification of the ICO surface with numerous RBD antigen clusters (ICO-RBD) to form various antigen patterns. Using an in vitro screening system, we demonstrate that the antigen spacing, antigen copies within clusters and cluster number parameters of the surface antigen pattern all impact the ability of the nanovaccines to activate B cells. Importantly, the optimized ICO-RBD nanovaccines evoke stronger and more enduring humoral and T cell immune responses in female mouse models compared to soluble RBD antigens, and the multivalent display broaden the protection range of B cell responses to more mutant strains. Our vaccines activate similar humoral immunity, observable stronger cellular immunity and more memory immune cells compared to trimeric mRNA vaccines.
Collapse
Affiliation(s)
- Qingqing Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lizhuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Dongshu Wang
- State Key Laboratory of Pathogens and Biosecurity, Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing, 100071, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Nana Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ming Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liting Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuehui Ma
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Jiajia Zou
- Beijing Intell Nanomedicine, No. 9, Chengwan Street, Haidian District, Beijing, 100000, China
| | - Quanwei Shi
- Beijing Intell Nanomedicine, No. 9, Chengwan Street, Haidian District, Beijing, 100000, China
| | - Pei Du
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Qihui Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Hengliang Wang
- State Key Laboratory of Pathogens and Biosecurity, Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing, 100071, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
6
|
Stepanova E, Isakova-Sivak I, Matyushenko V, Mezhenskaya D, Kudryavtsev I, Kostromitina A, Chistiakova A, Rak A, Bazhenova E, Prokopenko P, Kotomina T, Donina S, Novitskaya V, Sivak K, Karal-Ogly D, Rudenko L. Safety and Immunogenicity Study of a Bivalent Vaccine for Combined Prophylaxis of COVID-19 and Influenza in Non-Human Primates. Vaccines (Basel) 2024; 12:1099. [PMID: 39460266 PMCID: PMC11511058 DOI: 10.3390/vaccines12101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. METHODS Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. RESULTS There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. CONCLUSIONS The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Arina Kostromitina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Anna Chistiakova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Alexandra Rak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Svetlana Donina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Vlada Novitskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint-Petersburg 197376, Russia;
| | - Dzhina Karal-Ogly
- Center of Preclinical Research, Research Institute of Medical Primatology, Sochi 354376, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| |
Collapse
|
7
|
Dashti N, Golsaz-Shirazi F, Soltanghoraee H, Zarnani AH, Mohammadi M, Imani D, Jeddi-Tehrani M, Amiri MM, Shokri F. Preclinical assessment of a recombinant RBD-Fc fusion protein as SARS-CoV-2 candidate vaccine. Eur J Microbiol Immunol (Bp) 2024; 14:228-242. [PMID: 38753442 PMCID: PMC11393645 DOI: 10.1556/1886.2024.00045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Background Waning immunity and emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlight the need for further research in vaccine development. Methods A recombinant fusion protein containing the receptor-binding domain (RBD) fused to the human IgG1 Fc (RBD-Fc) was produced in CHO-K1 cells. RBD-Fc was emulsified with four adjuvants to evaluate its immunogenicity. The RBD-specific humoral and cellular immune responses were assessed by ELISA. The virus neutralizing potency of the vaccine was investigated using four neutralization methods. Safety was studied in mice and rabbits, and Antibody-Dependent Enhancement (ADE) effects were investigated by flow cytometry. Results RBD-Fc emulsified in Alum induced a high titer of anti-RBD antibodies with remarkable efficacy in neutralizing both pseudotyped and live SARS-CoV-2 Delta variant. The neutralization potency dropped significantly in response to the Omicron variant. RBD-Fc induced both TH2 and particularly TH1 immune responses. Histopathologic examinations demonstrated no substantial pathologic changes in different organs. No changes in serum biochemical and hematologic parameters were observed. ADE effect was not observed following immunization with RBD-Fc. Conclusion RBD-Fc elicits highly robust neutralizing antibodies and cellular immune responses, with no adverse effects. Therefore, it could be considered a promising and safe subunit vaccine against SARS-CoV-2.
Collapse
Affiliation(s)
- Navid Dashti
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Golsaz-Shirazi
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Soltanghoraee
- 2Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- 3Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mehdi Mohammadi
- 4Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Danyal Imani
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- 5Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Mehdi Amiri
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fazel Shokri
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Evtushenko E, Ryabchevskaya E, Kovalenko A, Granovskiy D, Arkhipenko M, Vasiliev Y, Nikitin N, Karpova O. Wuhan Sequence-Based Recombinant Antigens Expressed in E. coli Elicit Antibodies Capable of Binding with Omicron S-Protein. Int J Mol Sci 2024; 25:9016. [PMID: 39201702 PMCID: PMC11354337 DOI: 10.3390/ijms25169016] [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: 07/01/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
The development of cross-reactive vaccines is one of the central aims of modern vaccinology. Continuous mutation and the emergence of new SARS-CoV-2 variants and subvariants create the problem of universal coronavirus vaccine design. Previously, the authors devised three recombinant coronavirus antigens, which were based on the sequence collected in 2019 (the Wuhan variant) and produced in an E. coli bacterial expression system. The present work has shown, for the first time, that these recombinant antigens induce the production of antibodies that clearly interact with produced in CHO full-length S-protein of the Omicron variant. The immunogenicity of these recombinant antigens was studied in formulations with different adjuvants: Freund's adjuvant, Al(OH)3 and an adjuvant based on spherical particles (SPs), which are structurally modified plant virus. All adjuvanted formulations effectively stimulated Omicron-specific IgG production in mice. These universal coronavirus antigens could be considered the main component for the further development of broad-spectrum coronavirus vaccines for the prevention of SARS-CoV-2 infection. The present work also provides evidence that the synthetic biology approach is a promising strategy for the development of highly cross-reactive vaccines. Moreover, it is important to note that the bacterial expression system might be appropriate for the production of antigenically active universal antigens.
Collapse
Affiliation(s)
- Ekaterina Evtushenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (E.R.); (A.K.); (D.G.); (M.A.); (N.N.); (O.K.)
| | - Ekaterina Ryabchevskaya
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (E.R.); (A.K.); (D.G.); (M.A.); (N.N.); (O.K.)
| | - Angelina Kovalenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (E.R.); (A.K.); (D.G.); (M.A.); (N.N.); (O.K.)
| | - Dmitriy Granovskiy
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (E.R.); (A.K.); (D.G.); (M.A.); (N.N.); (O.K.)
| | - Marina Arkhipenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (E.R.); (A.K.); (D.G.); (M.A.); (N.N.); (O.K.)
| | | | - Nikolai Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (E.R.); (A.K.); (D.G.); (M.A.); (N.N.); (O.K.)
| | - Olga Karpova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (E.R.); (A.K.); (D.G.); (M.A.); (N.N.); (O.K.)
| |
Collapse
|
9
|
Gao T, Irie A, Kouwaki T, Oshiumi H. Development of a single-chain variable antibody fragment against a conserved region of the SARS-CoV-2 spike protein. Sci Rep 2024; 14:14419. [PMID: 38909102 PMCID: PMC11193732 DOI: 10.1038/s41598-024-64103-7] [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: 11/24/2023] [Accepted: 06/05/2024] [Indexed: 06/24/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has prolonged the duration of the pandemic because of the continuous emergence of new variant strains. The emergence of these mutant strains makes it difficult to detect the virus with the existing antibodies; thus, the development of novel antibodies that can target both the variants as well as the original strain is necessary. In this study, we generated a high-affinity monoclonal antibody (5G2) against the highly conserved region of the SARS-CoV-2 spike protein to detect the protein variants. Moreover, we generated its single-chain variable antibody fragment (sc5G2). The sc5G2 expressed in mammalian and bacterial cells detected the spike protein of the original SARS-CoV-2 and variant strains. The resulting sc5G2 will be a useful tool to detect the original SARS-CoV-2 and variant strains.
Collapse
Affiliation(s)
- Tingyu Gao
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Atsushi Irie
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| | - Takahisa Kouwaki
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| |
Collapse
|
10
|
Reed DS, McElroy AK, Barbeau DJ, McMillen CM, Tilston-Lunel NL, Nambulli S, Cottle E, Gilliland TC, Rannulu H, Lundy J, Olsen EL, O’Malley KJ, Xia M, Hartman AL, Luke TC, Egland K, Bausch C, Wu H, Sullivan EJ, Klimstra WB, Duprex WP. No evidence for enhanced disease with human polyclonal SARS-CoV-2 antibody in the ferret model. PLoS One 2024; 19:e0290909. [PMID: 38900732 PMCID: PMC11189238 DOI: 10.1371/journal.pone.0290909] [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: 08/17/2023] [Accepted: 05/06/2024] [Indexed: 06/22/2024] Open
Abstract
Since SARS-CoV-2 emerged in late 2019, it spread from China to the rest of the world. An initial concern was the potential for vaccine- or antibody-dependent enhancement (ADE) of disease as had been reported with other coronaviruses. To evaluate this, we first developed a ferret model by exposing ferrets to SARS-CoV-2 by either mucosal inoculation (intranasal/oral/ocular) or inhalation using a small particle aerosol. Mucosal inoculation caused a mild fever and weight loss that resolved quickly; inoculation via either route resulted in virus shedding detected in the nares, throat, and rectum for 7-10 days post-infection. To evaluate the potential for ADE, we then inoculated groups of ferrets intravenously with 0.1, 0.5, or 1 mg/kg doses of a human polyclonal anti-SARS-CoV-2 IgG from hyper-immunized transchromosomic bovines (SAB-185). Twelve hours later, ferrets were challenged by mucosal inoculation with SARS-CoV-2. We found no significant differences in fever, weight loss, or viral shedding after infection between the three antibody groups or the controls. Signs of pathology in the lungs were noted in infected ferrets but no differences were found between control and antibody groups. The results of this study indicate that healthy, young adult ferrets of both sexes are a suitable model of mild COVID-19 and that low doses of specific IgG in SAB-185 are unlikely to enhance the disease caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Douglas S. Reed
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Anita K. McElroy
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Division of Pediatric Infectious Disease, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Dominique J. Barbeau
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Division of Pediatric Infectious Disease, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Cynthia M. McMillen
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Natasha L. Tilston-Lunel
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Indiana University–Purdue University Indianapolis, Indianapolis, IN, United States of America
| | - Shamkumar Nambulli
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Emily Cottle
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Theron C. Gilliland
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hasala Rannulu
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jeneveve Lundy
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Emily L. Olsen
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Katherine J. O’Malley
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mengying Xia
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Amy L. Hartman
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Thomas C. Luke
- SAB Biotherapetuics, Sioux Falls, SD, United States of America
| | - Kristi Egland
- SAB Biotherapetuics, Sioux Falls, SD, United States of America
| | | | - Hua Wu
- SAB Biotherapetuics, Sioux Falls, SD, United States of America
| | | | - William B. Klimstra
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - W. Paul Duprex
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
11
|
Antonyan T, Chilingaryan G, Zagorski K, Ghazaryan M, Hovakimyan A, Davtyan H, Petrushina I, King O, Kniazev R, Petrovsky N, Ghochikyan A. MultiTEP-Based Vaccines Targeting SARS-CoV-2 Spike Protein IgG Epitopes Elicit Robust Binding Antibody Titers with Limited Virus-Neutralizing Activity. Pathogens 2024; 13:520. [PMID: 38921817 PMCID: PMC11206316 DOI: 10.3390/pathogens13060520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Within the last two decades, SARS-CoV-2 was the third zoonotic severe acute respiratory betacoronavirus (sarbecovirus) to infect humans, following SARS and MERS. The disruptions caused by the pandemic underscore the need for a universal vaccine against respiratory betacoronaviruses. Our group previously developed the universal platform for vaccine development, MultiTEP, which has been utilized in this study to generate a range of SARS-CoV-2 epitope vaccine candidates. We prepared and characterized 18 vaccines incorporating small peptide fragments from SARS-CoV-2 Spike protein fused with the MultiTEP sequence using overlapping PCR. Wild-type mice were immunized intramuscularly with the immunogen formulated in AdvaxCpG adjuvant. Serum antibodies were detected by ELISA, surrogate neutralization, and pseudovirus neutralization assays. Finally, the most promising vaccine candidate was administered to three non-human primates. All vaccines generated high titers of spike-binding IgG antibodies. However, only three vaccines generated antibodies that blocked RBD binding to the ACE2 receptor in a surrogate virus neutralization assay. However, none of the vaccines induced antibodies able to neutralize pseudotype viruses, including after the administration of the lead vaccine to NHPs. MultiTEP-based COVID-19 vaccines elicited robust, IgG-binding responses against the Spike protein in mice and non-human primates, but these antibodies were not neutralizing, underscoring the need to refine this approach further.
Collapse
Affiliation(s)
- Tatevik Antonyan
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| | - Garri Chilingaryan
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| | - Karen Zagorski
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| | - Manush Ghazaryan
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| | - Armine Hovakimyan
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| | - Hayk Davtyan
- Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Olga King
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| | - Roman Kniazev
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| | | | - Anahit Ghochikyan
- Department of Molecular Immunology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (T.A.)
| |
Collapse
|
12
|
Dillard JA, Taft-Benz SA, Knight AC, Anderson EJ, Pressey KD, Parotti B, Martinez SA, Diaz JL, Sarkar S, Madden EA, De la Cruz G, Adams LE, Dinnon KH, Leist SR, Martinez DR, Schäfer A, Powers JM, Yount BL, Castillo IN, Morales NL, Burdick J, Evangelista MKD, Ralph LM, Pankow NC, Linnertz CL, Lakshmanane P, Montgomery SA, Ferris MT, Baric RS, Baxter VK, Heise MT. Adjuvant-dependent impact of inactivated SARS-CoV-2 vaccines during heterologous infection by a SARS-related coronavirus. Nat Commun 2024; 15:3738. [PMID: 38702297 PMCID: PMC11068739 DOI: 10.1038/s41467-024-47450-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: 10/06/2023] [Accepted: 04/02/2024] [Indexed: 05/06/2024] Open
Abstract
Whole virus-based inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous coronavirus infection, the emergence of novel variants and the presence of large zoonotic reservoirs harboring novel heterologous coronaviruses provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes like vaccine-associated enhanced respiratory disease. Here, we use a female mouse model of coronavirus disease to evaluate inactivated vaccine performance against either homologous challenge with SARS-CoV-2 or heterologous challenge with a bat-derived coronavirus that represents a potential emerging disease threat. We show that inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide can cause enhanced respiratory disease during heterologous infection, while use of an alternative adjuvant does not drive disease and promotes heterologous viral clearance. In this work, we highlight the impact of adjuvant selection on inactivated vaccine safety and efficacy against heterologous coronavirus infection.
Collapse
Affiliation(s)
- Jacob A Dillard
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sharon A Taft-Benz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Audrey C Knight
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth J Anderson
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katia D Pressey
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Breantié Parotti
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sabian A Martinez
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer L Diaz
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sanjay Sarkar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily A Madden
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabriela De la Cruz
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lily E Adams
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth H Dinnon
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Boyd L Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Izabella N Castillo
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Noah L Morales
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jane Burdick
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Lauren M Ralph
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholas C Pankow
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Colton L Linnertz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Premkumar Lakshmanane
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephanie A Montgomery
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Dallas Tissue Research, Farmers Branch, TX, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria K Baxter
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Texas Biomedical Research Institute, San Antonio, TX, USA.
| | - Mark T Heise
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
13
|
Gao R, Feng C, Sheng Z, Li F, Wang D. Research progress in Fc-effector functions against SARS-CoV-2. J Med Virol 2024; 96:e29638. [PMID: 38682662 DOI: 10.1002/jmv.29638] [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: 03/12/2024] [Revised: 03/31/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has caused more than 676 million cases in the global human population with approximately 7 million deaths and vaccination has been proved as the most effective countermeasure in reducing clinical complications and mortality rate of SARS-CoV-2 infection in people. However, the protective elements and correlation of protection induced by vaccination are still not completely understood. Various antibodies with multiple protective mechanisms can be induced simultaneously by vaccination in vivo, thereby complicating the identification and characterization of individual correlate of protection. Recently, an increasing body of observations suggests that antibody-induced Fc-effector functions play a crucial role in combating SARS-CoV-2 infections, including neutralizing antibodies-escaping variants. Here, we review the recent progress in understanding the impact of Fc-effector functions in broadly disarming SARS-CoV-2 infectivity and discuss various efforts in harnessing this conserved antibody function to develop an effective SARS-CoV-2 vaccine that can protect humans against infections by SARS-CoV-2 virus and its variants of concern.
Collapse
Affiliation(s)
- Rongyuan Gao
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Chenchen Feng
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Zizhang Sheng
- Zuckerman Mind Brian Behavior Institute, Columbia University, New York, New York, USA
| | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| | - Dan Wang
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
14
|
Sakai Y, Kuwahara K. Carcinogenesis caused by transcription-coupled DNA damage through GANP and other components of the TREX-2 complex. Pathol Int 2024; 74:103-118. [PMID: 38411330 DOI: 10.1111/pin.13415] [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: 08/27/2023] [Revised: 02/05/2024] [Accepted: 02/10/2024] [Indexed: 02/28/2024]
Abstract
Perturbation of genes is important for somatic hypermutation to increase antibody affinity during B-cell immunity; however, it may also promote carcinogenesis. Previous studies have revealed that transcription is an important process that can induce DNA damage and genomic instability. Transciption-export-2 (TREX-2) complex, which regulates messenger RNA (mRNA) nuclear export, has been studied in the budding yeast Saccharomyces cerevisiae; however, recent studies have started investigating the molecular function of the mammalian TREX-2 complex. The central molecule in the TREX-2 complex, that is, germinal center-associated nuclear protein (GANP), is closely associated with antibody affinity maturation as well as cancer etiology. In this review, we focus on carcinogenesis, lymphomagenesis, and teratomagenesis caused by transcription-coupled DNA damage through GANP and other components of the TREX-2 complex. We review the basic machinery of mRNA nuclear export and transcription-coupled DNA damage. We then briefly describe the immunological relationship between GANP and the affinity maturation of antibodies. Finally, we illustrate that the aberrant expression of the components of the TREX-2 complex, especially GANP, is associated with the etiology of various solid tumors, lymphomas, and testicular teratoma. These components serve as reliable predictors of cancer prognosis and response to chemotherapy.
Collapse
Affiliation(s)
- Yasuhiro Sakai
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Aichi, Japan
| | - Kazuhiko Kuwahara
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka, Japan
| |
Collapse
|
15
|
Ko HL, Lee DK, Kim Y, Jang HJ, Lee YW, Lee HY, Seok SH, Park JW, Limb JK, On DI, Yun JW, Lyoo KS, Song D, Yeom M, Lee H, Seong JK, Lee S. Development of a neutralization monoclonal antibody with a broad neutralizing effect against SARS-CoV-2 variants. Virol J 2023; 20:285. [PMID: 38041113 PMCID: PMC10693169 DOI: 10.1186/s12985-023-02230-9] [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: 08/14/2023] [Accepted: 11/06/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has challenged the effectiveness of current therapeutic regimens. Here, we aimed to develop a potent SARS-CoV-2 antibody with broad neutralizing effect by screening a scFv library with the spike protein receptor-binding domain (RBD) via phage display. METHODS SKAI-DS84 was identified through phage display, and we performed pseudovirus neutralization assays, authentic virus neutralization assays, and in vivo neutralization efficacy evaluations. Furthermore, surface plasmon resonance (SPR) analysis was conducted to assess the physical characteristics of the antibody, including binding kinetics and measure its affinity for variant RBDs. RESULTS The selected clones were converted to human IgG, and among them, SKAI-DS84 was selected for further analyses based on its binding affinity with the variant RBDs. Using pseudoviruses, we confirmed that SKAI-DS84 was strongly neutralizing against wild-type, B.1.617.2, B.1.1.529, and subvariants of SARS-CoV-2. We also tested the neutralizing effect of SKAI-DS84 on authentic viruses, in vivo and observed a reduction in viral replication and improved lung pathology. We performed binding and epitope mapping experiments to understand the mechanisms underlying neutralization and identified quaternary epitopes formed by the interaction between RBDs as the target of SKAI-DS84. CONCLUSIONS We identified, produced, and tested the neutralizing effect of SKAI-DS84 antibody. Our results highlight that SKAI-DS84 could be a potential neutralizing antibody against SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Hae Li Ko
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
- Department of Microbiology, College of Medical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Deuk-Ki Lee
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
- Department of Microbiology, College of Medical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Younghyeon Kim
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
- Department of Microbiology, College of Medical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Hui Jeong Jang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Youn Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Jin-Kyung Limb
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, South Korea
| | - Da In On
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, South Korea
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Kwang-Soo Lyoo
- College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Daesub Song
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Minjoo Yeom
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hanbyeul Lee
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, South Korea.
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
- Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, 08826, South Korea.
| | - Sungjin Lee
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
16
|
Buzas D, Bunzel AH, Staufer O, Milodowski EJ, Edmunds GL, Bufton JC, Vidana Mateo BV, Yadav SKN, Gupta K, Fletcher C, Williamson MK, Harrison A, Borucu U, Capin J, Francis O, Balchin G, Hall S, Vega MV, Durbesson F, Lingappa S, Vincentelli R, Roe J, Wooldridge L, Burt R, Anderson RJL, Mulholland AJ, Bristol UNCOVER Group, Hare J, Bailey M, Davidson AD, Finn A, Morgan D, Mann J, Spatz J, Garzoni F, Schaffitzel C, Berger I. In vitro generated antibodies guide thermostable ADDomer nanoparticle design for nasal vaccination and passive immunization against SARS-CoV-2. Antib Ther 2023; 6:277-297. [PMID: 38075238 PMCID: PMC10702856 DOI: 10.1093/abt/tbad024] [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: 06/20/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 01/10/2024] Open
Abstract
Background Due to COVID-19, pandemic preparedness emerges as a key imperative, necessitating new approaches to accelerate development of reagents against infectious pathogens. Methods Here, we developed an integrated approach combining synthetic, computational and structural methods with in vitro antibody selection and in vivo immunization to design, produce and validate nature-inspired nanoparticle-based reagents against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Results Our approach resulted in two innovations: (i) a thermostable nasal vaccine called ADDoCoV, displaying multiple copies of a SARS-CoV-2 receptor binding motif derived epitope and (ii) a multivalent nanoparticle superbinder, called Gigabody, against SARS-CoV-2 including immune-evasive variants of concern (VOCs). In vitro generated neutralizing nanobodies and electron cryo-microscopy established authenticity and accessibility of epitopes displayed by ADDoCoV. Gigabody comprising multimerized nanobodies prevented SARS-CoV-2 virion attachment with picomolar EC50. Vaccinating mice resulted in antibodies cross-reacting with VOCs including Delta and Omicron. Conclusion Our study elucidates Adenovirus-derived dodecamer (ADDomer)-based nanoparticles for use in active and passive immunization and provides a blueprint for crafting reagents to combat respiratory viral infections.
Collapse
Affiliation(s)
- Dora Buzas
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Adrian H Bunzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Oskar Staufer
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Leibniz Institute for New Materials, Helmholtz Institute for Pharmaceutical Research and Center for Biophysics, Saarland University, Saarbrücken 66123, Germany
| | | | - Grace L Edmunds
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joshua C Bufton
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | - Kapil Gupta
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | | | - Maia K Williamson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Ufuk Borucu
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Julien Capin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Ore Francis
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Georgia Balchin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Sophie Hall
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Mirella V Vega
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Fabien Durbesson
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | | | - Renaud Vincentelli
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | - Joe Roe
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Linda Wooldridge
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Rachel Burt
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | | | | | | | - Jonathan Hare
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Mick Bailey
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Andrew D Davidson
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Adam Finn
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
- Children's Vaccine Centre, Bristol Medical School, Bristol BS2 8EF UK
| | - David Morgan
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Jamie Mann
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joachim Spatz
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Frederic Garzoni
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Christiane Schaffitzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| | - Imre Berger
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| |
Collapse
|
17
|
Wang X, Li M, Lu P, Li C, Zhao C, Zhao X, Qiao R, Cui Y, Chen Y, Li J, Cai G, Wang P. In Vitro Antibody-Dependent Enhancement of SARS-CoV-2 Infection Could Be Abolished by Adding Human IgG. Pathogens 2023; 12:1108. [PMID: 37764916 PMCID: PMC10535176 DOI: 10.3390/pathogens12091108] [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: 07/18/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Evidence of antibody-dependent enhancement (ADE) of other viruses has raised concerns about the safety of SARS-CoV-2 vaccines and antibody therapeutics. In vitro studies have shown ADE of SARS-CoV-2 infection. In this study, we also found that vaccination/convalescent sera and some approved monoclonal antibodies can enhance SARS-CoV-2 infection of FcR-expressing B cells in vitro. However, the enhancement of SARS-CoV-2 infection can be prevented by blocking Fc-FcR interaction through the addition of human serum/IgG or the introduction of mutations in the Fc portion of the antibody. It should be noted that ADE activity observed on FcR-expressing cells in vitro may not necessarily reflect the situation in vivo; therefore, animal and clinical data should be included for ADE evaluation.
Collapse
Affiliation(s)
- Xun Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Minghui Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Panpan Lu
- Reproductive Center, Women and Children's Hospital, Qingdao University, Qingdao 266001, China
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chaoyue Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaoyu Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Rui Qiao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yuchen Cui
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yanjia Chen
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jiayan Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Guonan Cai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
18
|
Ziganshina MM, Shilova NV, Khalturina EO, Dolgushina NV, V Borisevich S, Yarotskaya EL, Bovin NV, Sukhikh GT. Antibody-Dependent Enhancement with a Focus on SARS-CoV-2 and Anti-Glycan Antibodies. Viruses 2023; 15:1584. [PMID: 37515270 PMCID: PMC10384250 DOI: 10.3390/v15071584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon where virus-specific antibodies paradoxically cause enhanced viral replication and/or excessive immune responses, leading to infection exacerbation, tissue damage, and multiple organ failure. ADE has been observed in many viral infections and is supposed to complicate the course of COVID-19. However, the evidence is insufficient. Since no specific laboratory markers have been described, the prediction and confirmation of ADE are very challenging. The only possible predictor is the presence of already existing (after previous infection) antibodies that can bind to viral epitopes and promote the disease enhancement. At the same time, the virus-specific antibodies are also a part of immune response against a pathogen. These opposite effects of antibodies make ADE research controversial. The assignment of immunoglobulins to ADE-associated or virus neutralizing is based on their affinity, avidity, and content in blood. However, these criteria are not clearly defined. Another debatable issue (rather terminological, but no less important) is that in most publications about ADE, all immunoglobulins produced by the immune system against pathogens are qualified as pre-existing antibodies, thus ignoring the conventional use of this term for natural antibodies produced without any stimulation by pathogens. Anti-glycan antibodies (AGA) make up a significant part of the natural immunoglobulins pool, and there is some evidence of their antiviral effect, particularly in COVID-19. AGA have been shown to be involved in ADE in bacterial infections, but their role in the development of ADE in viral infections has not been studied. This review focuses on pros and cons for AGA as an ADE trigger. We also present the results of our pilot studies, suggesting that AGAs, which bind to complex epitopes (glycan plus something else in tight proximity), may be involved in the development of the ADE phenomenon.
Collapse
Affiliation(s)
- Marina M Ziganshina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
| | - Nadezhda V Shilova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Eugenia O Khalturina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Natalya V Dolgushina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | | | - Ekaterina L Yarotskaya
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
| | - Nicolai V Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Gennady T Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
19
|
Lobaina Y, Chen R, Suzarte E, Ai P, Huerta V, Tan C, Alvarez-Lajonchere L, Liling Y, Musacchio A, Silva R, Guillén G, Zaixue J, Yang K, Perera Y, Hermida L. Broad humoral immunity generated in mice by a formulation composed of two antigens from the Delta variant of SARS-CoV-2. Arch Virol 2023; 168:190. [PMID: 37351679 DOI: 10.1007/s00705-023-05812-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/05/2023] [Indexed: 06/24/2023]
Abstract
Due to the rapid development of new variants of SARS-CoV-2 as well as the real threat of new coronavirus zoonosis events, the development of a preventive vaccine with a broader scope of functionality is highly desirable. Previously, we reported the functionality of a nasal formulation containing the nucleocapsid protein and the receptor-binding domain (RBD) of the spike protein of the Delta variant of SARS-CoV-2 combined with the ODN-39M adjuvant. This combination induced cross-reactive immunity in mucosal and systemic compartments at the sarbecovirus level. In the present study, we explored the magnitude of the immunity generated in BALB/c mice by the same formulation with alum added as an additional adjuvant, to enhance the humoral immunity against the two antigens. Animals were immunized with three doses of the bivalent formulation, administered by subcutaneous route. Humoral immunity was tested by ELISA, and the neutralizing capacity of the resulting antibodies (Abs) was evaluated using a surrogate test and a vesicular stomatitis virus (VSV) pseudovirus-based assay. Cell-mediated immunity was also investigated using an IFN-γ ELISpot assay. High levels of antibodies against both antigens (N and RBD) were obtained upon immunization. Anti-RBD Abs with neutralizing capacity reacted with the RBD of three SARS-CoV-2 variants tested, including Omicron. Abs recognizing the nucleocapsid proteins of SARS-CoV-1 and the SARS-CoV-2 Delta and Omicron variants were also detected. Taken together, these results suggest that this bivalent formulation could be an attractive component of a pancorona vaccine able to broaden the scope of humoral immunity against both antigens. This will be particularly important for the reinforcement of immunity in previously vaccinated and/or infected populations.
Collapse
Affiliation(s)
- Yadira Lobaina
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China
- Research Department, Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou, 425000, Hunan, China
| | - Rong Chen
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou, Hunan, China
| | - Edith Suzarte
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Panchao Ai
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou, Hunan, China
| | - Vivian Huerta
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Changyuan Tan
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou, Hunan, China
| | - Liz Alvarez-Lajonchere
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Yang Liling
- Department of Laboratory Medicine, Dongguan Ninth People's Hospital, No. 88, Shaditang, Guancheng District, Dongguan, Guangdong, China
| | - Alexis Musacchio
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Ricardo Silva
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China
- Scientific Department, Representative Office of BioCubaFarma in China, Jingtai Tower, No. 24 Jianguomen Wai Street, Chaoyang District, Beijing, 100022, China
| | - Gerardo Guillén
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Jiang Zaixue
- Guangdong Eighth People's Hospital, No. 68 South, Shilong Xihu 3rd Road, Shilong Town, Dongguan, Guangdong, China
| | - Ke Yang
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China.
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou, Hunan, China.
| | - Yasser Perera
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China.
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba.
- Research Department, Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou, 425000, Hunan, China.
| | - Lisset Hermida
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou, 425000, Hunan, China.
- Scientific Department, Representative Office of BioCubaFarma in China, Jingtai Tower, No. 24 Jianguomen Wai Street, Chaoyang District, Beijing, 100022, China.
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou, Hunan, China.
| |
Collapse
|
20
|
Wen Z, Yuan Y, Zhao Y, Wang H, Han Z, Li M, Yuan J, Sun C. Enhancement of SARS-CoV-2 N Antigen-Specific T Cell Functionality by Modulating the Autophagy-Mediated Signal Pathway in Mice. Viruses 2023; 15:1316. [PMID: 37376617 DOI: 10.3390/v15061316] [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: 05/03/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
The frequent SARS-CoV-2 variants have caused a continual challenge, weakening the effectiveness of current vaccines, and thus it is of great importance to induce robust and conserved T cellular immunity for developing the next-generation vaccine against SARS-CoV-2 variants. In this study, we proposed a conception of enhancing the SARS-CoV-2 specific T cell functionality by fusing autophagosome-associated LC3b protein to the nucleocapsid (N) (N-LC3b). When compared to N protein alone, the N-LC3b protein was more effectively targeted to the autophagosome/lysosome/MHC II compartment signal pathway and thus elicited stronger CD4+ and CD8+ T cell immune responses in mice. Importantly, the frequency of N-specific polyfunctional CD4+ and CD8+ T cells, which can simultaneously secrete multiple cytokines (IFN-γ+/IL-2+/TNF-α+), in the N-LC3b group was significantly higher than that in the N alone group. Moreover, there was a significantly improved T cell proliferation, especially for CD8+ T cells in the N-LC3b group. In addition, the N-LC3b also induced a robust humoral immune response, characterized by the Th1-biased IgG2a subclass antibodies against the SARS-CoV-2 N protein. Overall, these findings demonstrated that our strategy could effectively induce a potential SARS-CoV-2 specific T cellular immunity with enhanced magnitude, polyfunctionality, and proliferation, and thus provided insights to develop a promising strategy for the design of a novel universal vaccine against SARS-CoV-2 variants and other emerging infectious diseases.
Collapse
Affiliation(s)
- Ziyu Wen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Yue Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Yangguo Zhao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Haohang Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Zirong Han
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Minchao Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen 518000, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China
| |
Collapse
|
21
|
Moustafa RI, Faraag AHI, El-Shenawy R, Agwa MM, Elsayed H. Harnessing immunoinformatics for developing a multiple-epitope peptide-based vaccination approach against SARS-CoV-2 spike protein. Saudi J Biol Sci 2023; 30:103661. [PMID: 37163156 PMCID: PMC10141799 DOI: 10.1016/j.sjbs.2023.103661] [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: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
COVID-19 has spread to over 200 countries with variable severity and mortality rates. Computational analysis is a valuable tool for developing B-cell and T-cell epitope-based vaccines. In this study, by harnessing immunoinformatics tools, we designed a multiple-epitope vaccine to protect against COVID-19. The candidate epitopes were designed from highly conserved regions of the SARS-CoV-2 spike (S) glycoprotein. The consensus amino acids sequence of ten SARS-CoV-2 variants including Gamma, Beta, Epsilon, Delta, Alpha, Kappa, Iota, Lambda, Mu, and Omicron was involved. Applying the multiple sequence alignment plugin and the antigenic prediction tools of Geneious prime 2021, ten predicted variants were identified and consensus S-protein sequences were used to predict the antigenic part. According to ElliPro analysis of S-protein B-cell prediction, we explored 22 continuous linear epitopes with high scores ranging from 0.879 to 0.522. First, we reported five promising epitopes: BE1 1115-1192, BE2 481-563, BE3 287-313, BE4 62-75, and BE5 112-131 with antigenicity scores of 0.879, 0.86, 0.813, 0.779, and 0.765, respectively, while only nine discontinuous epitopes scored between 0.971 and 0.511. Next, we identified 194 Major Histocompatibility Complex (MHC) - I and 156 MHC - II epitopes with antigenic characteristics. These spike-specific peptide-epitopes with characteristically high immunogenic and antigenic scores have the potential as a SARS-CoV-2 multiple-epitope peptide-based vaccination strategy. Nevertheless, further experimental investigations are needed to test for the vaccine efficacy and efficiency.
Collapse
Affiliation(s)
- Rehab I Moustafa
- Department of Microbial Biotechnology, Biotechnology Research Institute, National Research Centre, Egypt
| | - Ahmed H I Faraag
- Botany and Microbiology Department, Faculty of Science, Helwan University, Egypt
- School of Biotechnology, Badr University in Cairo, Egypt
| | | | - Mona M Agwa
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Egypt
| | - Hassan Elsayed
- Department of Microbial Biotechnology, Biotechnology Research Institute, National Research Centre, Egypt
| |
Collapse
|
22
|
Rando HM, Lordan R, Lee AJ, Naik A, Wellhausen N, Sell E, Kolla L, Gitter A, Greene CS. Application of Traditional Vaccine Development Strategies to SARS-CoV-2. mSystems 2023; 8:e0092722. [PMID: 36861991 PMCID: PMC10134813 DOI: 10.1128/msystems.00927-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Over the past 150 years, vaccines have revolutionized the relationship between people and disease. During the COVID-19 pandemic, technologies such as mRNA vaccines have received attention due to their novelty and successes. However, more traditional vaccine development platforms have also yielded important tools in the worldwide fight against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A variety of approaches have been used to develop COVID-19 vaccines that are now authorized for use in countries around the world. In this review, we highlight strategies that focus on the viral capsid and outwards, rather than on the nucleic acids inside. These approaches fall into two broad categories: whole-virus vaccines and subunit vaccines. Whole-virus vaccines use the virus itself, in either an inactivated or an attenuated state. Subunit vaccines contain instead an isolated, immunogenic component of the virus. Here, we highlight vaccine candidates that apply these approaches against SARS-CoV-2 in different ways. In a companion article (H. M. Rando, R. Lordan, L. Kolla, E. Sell, et al., mSystems 8:e00928-22, 2023, https://doi.org/10.1128/mSystems.00928-22), we review the more recent and novel development of nucleic acid-based vaccine technologies. We further consider the role that these COVID-19 vaccine development programs have played in prophylaxis at the global scale. Well-established vaccine technologies have proved especially important to making vaccines accessible in low- and middle-income countries. Vaccine development programs that use established platforms have been undertaken in a much wider range of countries than those using nucleic acid-based technologies, which have been led by wealthy Western countries. Therefore, these vaccine platforms, though less novel from a biotechnological standpoint, have proven to be extremely important to the management of SARS-CoV-2. IMPORTANCE The development, production, and distribution of vaccines is imperative to saving lives, preventing illness, and reducing the economic and social burdens caused by the COVID-19 pandemic. Vaccines that use cutting-edge biotechnology have played an important role in mitigating the effects of SARS-CoV-2. However, more traditional methods of vaccine development that were refined throughout the 20th century have been especially critical to increasing vaccine access worldwide. Effective deployment is necessary to reducing the susceptibility of the world's population, which is especially important in light of emerging variants. In this review, we discuss the safety, immunogenicity, and distribution of vaccines developed using established technologies. In a separate review, we describe the vaccines developed using nucleic acid-based vaccine platforms. From the current literature, it is clear that the well-established vaccine technologies are also highly effective against SARS-CoV-2 and are being used to address the challenges of COVID-19 globally, including in low- and middle-income countries. This worldwide approach is critical for reducing the devastating impact of SARS-CoV-2.
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ronan Lordan
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amruta Naik
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Sell
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Likhitha Kolla
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - COVID-19 Review Consortium
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Affonso de Oliveira JF, Zhao Z, Xiang Y, Shin MD, Villaseñor KE, Deng X, Shukla S, Chen S, Steinmetz NF. COVID-19 vaccines based on viral nanoparticles displaying a conserved B-cell epitope show potent immunogenicity and a long-lasting antibody response. Front Microbiol 2023; 14:1117494. [PMID: 37152732 PMCID: PMC10157238 DOI: 10.3389/fmicb.2023.1117494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 05/09/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 sparked intensive research into the development of effective vaccines, 50 of which have been approved thus far, including the novel mRNA-based vaccines developed by Pfizer and Moderna. Although limiting the severity of the disease, the mRNA-based vaccines presented drawbacks, such as the cold chain requirement. Moreover, antibody levels generated by these vaccines decline significantly after 6 months. These vaccines deliver mRNA encoding the full-length spike (S) glycoprotein of SARS-CoV-2, but must be updated as new strains and variants of concern emerge, creating a demand for adjusted formulations and booster campaigns. To overcome these challenges, we have developed COVID-19 vaccine candidates based on the highly conserved SARS CoV-2, 809-826 B-cell peptide epitope (denoted 826) conjugated to cowpea mosaic virus (CPMV) nanoparticles and bacteriophage Qβ virus-like particles, both platforms have exceptional thermal stability and facilitate epitope delivery with inbuilt adjuvant activity. We evaluated two administration methods: subcutaneous injection and an implantable polymeric scaffold. Mice received a prime-boost regimen of 100 μg per dose (2 weeks apart) or a single dose of 200 μg administered as a liquid formulation, or a polymer implant. Antibody titers were evaluated longitudinally over 50 weeks. The vaccine candidates generally elicited an early Th2-biased immune response, which stimulates the production of SARS-CoV-2 neutralizing antibodies, followed by a switch to a Th1-biased response for most formulations. Exceptionally, vaccine candidate 826-CPMV (administered as prime-boost, soluble injection) elicited a balanced Th1/Th2 immune response, which is necessary to prevent pulmonary immunopathology associated with Th2 bias extremes. While the Qβ-based vaccine elicited overall higher antibody titers, the CPMV-induced antibodies had higher avidity. Regardless of the administration route and formulation, our vaccine candidates maintained high antibody titers for more than 50 weeks, confirming a potent and durable immune response against SARS-CoV-2 even after a single dose.
Collapse
Affiliation(s)
| | - Zhongchao Zhao
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Yi Xiang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Matthew D. Shin
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | | | - Xinyi Deng
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Sourabh Shukla
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Shaochen Chen
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
- Center for Engineering in Cancer, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
24
|
Rabaan AA, Al-Ahmed SH, Albayat H, Alwarthan S, Alhajri M, Najim MA, AlShehail BM, Al-Adsani W, Alghadeer A, Abduljabbar WA, Alotaibi N, Alsalman J, Gorab AH, Almaghrabi RS, Zaidan AA, Aldossary S, Alissa M, Alburaiky LM, Alsalim FM, Thakur N, Verma G, Dhawan M. Variants of SARS-CoV-2: Influences on the Vaccines' Effectiveness and Possible Strategies to Overcome Their Consequences. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:507. [PMID: 36984508 PMCID: PMC10051174 DOI: 10.3390/medicina59030507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
The immune response elicited by the current COVID-19 vaccinations declines with time, especially among the immunocompromised population. Furthermore, the emergence of novel SARS-CoV-2 variants, particularly the Omicron variant, has raised serious concerns about the efficacy of currently available vaccines in protecting the most vulnerable people. Several studies have reported that vaccinated people get breakthrough infections amid COVID-19 cases. So far, five variants of concern (VOCs) have been reported, resulting in successive waves of infection. These variants have shown a variable amount of resistance towards the neutralising antibodies (nAbs) elicited either through natural infection or the vaccination. The spike (S) protein, membrane (M) protein, and envelope (E) protein on the viral surface envelope and the N-nucleocapsid protein in the core of the ribonucleoprotein are the major structural vaccine target proteins against COVID-19. Among these targets, S Protein has been extensively exploited to generate effective vaccines against COVID-19. Hence, amid the emergence of novel variants of SARS-CoV-2, we have discussed their impact on currently available vaccines. We have also discussed the potential roles of S Protein in the development of novel vaccination approaches to contain the negative consequences of the variants' emergence and acquisition of mutations in the S Protein of SARS-CoV-2. Moreover, the implications of SARS-CoV-2's structural proteins were also discussed in terms of their variable potential to elicit an effective amount of immune response.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Shamsah H. Al-Ahmed
- Specialty Paediatric Medicine, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Hawra Albayat
- Infectious Disease Department, King Saud Medical City, Riyadh 7790, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mashael Alhajri
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mustafa A. Najim
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Taibah University, Madinah 41411, Saudi Arabia
| | - Bashayer M. AlShehail
- Pharmacy Practice Department, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Wasl Al-Adsani
- Department of Medicine, Infectious Diseases Hospital, Kuwait City 63537, Kuwait
- Department of Infectious Diseases, Hampton Veterans Administration Medical Center, Hampton, VA 23667, USA
| | - Ali Alghadeer
- Department of Anesthesia, Dammam Medical Complex, Dammam 32245, Saudi Arabia
| | - Wesam A. Abduljabbar
- Department of Medical Laboratory Sciences, Fakeeh College for Medical Science, Jeddah 21134, Saudi Arabia
| | - Nouf Alotaibi
- Clinical Pharmacy Department, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Jameela Alsalman
- Infection Disease Unit, Department of Internal Medicine, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 435, Bahrain
| | - Ali H. Gorab
- Al Kuzama Primary Health Care Center, Al Khobar Health Network, Eastern Health Cluster, Al Khobar 34446, Saudi Arabia
| | - Reem S. Almaghrabi
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Ali A. Zaidan
- Gastroenterology Department, King Fahad Armed Forces Hospital, Jeddah 23831, Saudi Arabia
| | - Sahar Aldossary
- Pediatric Infectious Diseases, Women and Children’s Health Institute, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Lamees M. Alburaiky
- Pediatric Department, Safwa General Hospital, Eastern Health Cluster, Safwa 31921, Saudi Arabia
| | - Fatimah Mustafa Alsalim
- Department of Family Medicine, Primary Health Care, Qatif Health Cluster, Qatif 32434, Saudi Arabia
| | - Nanamika Thakur
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Geetika Verma
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, India
- Trafford College, Altrincham, Manchester WA14 5PQ, UK
| |
Collapse
|
25
|
Rando HM, Lordan R, Lee AJ, Naik A, Wellhausen N, Sell E, Kolla L, Gitter A, Greene CS. Application of Traditional Vaccine Development Strategies to SARS-CoV-2. ARXIV 2023:arXiv:2208.08907v2. [PMID: 36034485 PMCID: PMC9413721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Over the past 150 years, vaccines have revolutionized the relationship between people and disease. During the COVID-19 pandemic, technologies such as mRNA vaccines have received attention due to their novelty and successes. However, more traditional vaccine development platforms have also yielded important tools in the worldwide fight against the SARS-CoV-2 virus. A variety of approaches have been used to develop COVID-19 vaccines that are now authorized for use in countries around the world. In this review, we highlight strategies that focus on the viral capsid and outwards, rather than on the nucleic acids inside. These approaches fall into two broad categories: whole-virus vaccines and subunit vaccines. Whole-virus vaccines use the virus itself, either in an inactivated or attenuated state. Subunit vaccines contain instead an isolated, immunogenic component of the virus. Here, we highlight vaccine candidates that apply these approaches against SARS-CoV-2 in different ways. In a companion manuscript, we review the more recent and novel development of nucleic-acid based vaccine technologies. We further consider the role that these COVID-19 vaccine development programs have played in prophylaxis at the global scale. Well-established vaccine technologies have proved especially important to making vaccines accessible in low- and middle-income countries. Vaccine development programs that use established platforms have been undertaken in a much wider range of countries than those using nucleic-acid-based technologies, which have been led by wealthy Western countries. Therefore, these vaccine platforms, though less novel from a biotechnological standpoint, have proven to be extremely important to the management of SARS-CoV-2.
Collapse
Affiliation(s)
- Halie M Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America; Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America · Funded by the Gordon and Betty Moore Foundation (GBMF 4552); the National Human Genome Research Institute (R01 HG010067)
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Alexandra J Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America · Funded by the Gordon and Betty Moore Foundation (GBMF 4552)
| | - Amruta Naik
- Children's Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elizabeth Sell
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Likhitha Kolla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America · Funded by NIH Medical Scientist Training Program T32 GM07170
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; Morgridge Institute for Research, Madison, Wisconsin, United States of America · Funded by John W. and Jeanne M. Rowe Center for Research in Virology
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America; Childhood Cancer Data Lab, Alex's Lemonade Stand Foundation, Philadelphia, Pennsylvania, United States of America; Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America · Funded by the Gordon and Betty Moore Foundation (GBMF 4552); the National Human Genome Research Institute (R01 HG010067)
| |
Collapse
|
26
|
Monteiro FP, Tavares VDS, Souza RDSO, Venâncio LPR, Fabres-Klein MH, do Carmo RF, Klein RC, Lima JB, Araújo-Santos T. Coagulopathy and the humoral response against viral proteins in patients at different stages of COVID-19. Mem Inst Oswaldo Cruz 2023; 117:e220072. [PMID: 36700578 PMCID: PMC9870257 DOI: 10.1590/0074-02760220072] [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: 04/01/2022] [Accepted: 11/17/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Patients with severe coronavirus disease 2019 (COVID-19) often present with coagulopathies and have high titres of circulating antibodies against viral proteins. OBJECTIVES Herein, we evaluated the association between D-dimer and circulating immunoglobulin levels against viral proteins in patients at different clinical stages of COVID-19. METHODS For this, we performed a cross-sectional study involving patients of the first wave of COVID-19 clinically classified as oligosymptomatic (n = 22), severe (n = 30), cured (n = 27) and non-infected (n = 9). Next, we measured in the plasma samples the total and fraction of immunoglobulins against the nucleoprotein (NP) and the receptor-binding domain (RBD) of the spike proteins by enzyme-linked immunosorbent assay (ELISA) assays. FINDINGS Patients with severe disease had a coagulation disorder with high levels of D-dimer as well as circulating IgG against the NP but not the RBD compared to other groups of patients. In addition, high levels of D-dimer and IgG against the NP and RBD were associated with disease severity among the patients in this study. MAIN CONCLUSIONS Our data suggest that IgG against NP and RBD participates in the worsening of COVID-19. Although the humoral response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is partially understood, and more efforts are needed to clarify gaps in the knowledge of this process.
Collapse
Affiliation(s)
- Fernanda Pereira Monteiro
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil
| | - Victor de Souza Tavares
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil
| | - Rayane da Silva Oliveira Souza
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil
| | - Larissa Paola Rodrigues Venâncio
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil
| | - Mary Hellen Fabres-Klein
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil
| | - Rodrigo Feliciano do Carmo
- Universidade Federal do Vale do São Francisco, Colegiado de Ciências Farmacêuticas, Petrolina, PE, Brasil
| | - Raphael Contelli Klein
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil
| | - Jonilson Berlink Lima
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil
| | - Théo Araújo-Santos
- Universidade Federal do Oeste da Bahia, Centro das Ciências Biológicas e da Saúde, Núcleo de Estudos de Agentes Infecciosos e Vetores, Barreiras, BA, Brasil,+ Corresponding author:
| |
Collapse
|
27
|
Kovalenko A, Ryabchevskaya E, Evtushenko E, Nikitin N, Karpova O. Recombinant Protein Vaccines against Human Betacoronaviruses: Strategies, Approaches and Progress. Int J Mol Sci 2023; 24:1701. [PMID: 36675218 PMCID: PMC9863728 DOI: 10.3390/ijms24021701] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Betacoronaviruses have already troubled humanity more than once. In 2002-2003 and 2012, the SARS-CoV and MERS-CoV, respectively, caused outbreaks of respiratory syndromes with a fatal outcome. The spread of the SARS-CoV-2 coronavirus has become a pandemic. These three coronaviruses belong to the genus Betacoronavirus and have a zoonotic origin. The emergence of new coronavirus infections in the future cannot be ruled out, and vaccination is the main way to prevent the spread of the infection. Previous experience in the development of vaccines against SARS and MERS has helped to develop a number of vaccines against SARS-CoV-2 in a fairly short time. Among them, there are quite a few recombinant protein vaccines, which seem to be very promising in terms of safety, minimization of side effects, storage and transportation conditions. The problem of developing a universal betacoronavirus vaccine is also still relevant. Here, we summarize the information on the designing of vaccines based on recombinant proteins against highly pathogenic human betacoronaviruses SARS-CoV, MERS-CoV and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | | | - Nikolai Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | | |
Collapse
|
28
|
Gong HR, Hu YF, Li X, Yau T, Zhang BZ, Huang JD. Non-Neutralizing Epitopes Shade Neutralizing Epitopes against Omicron in a Multiple Epitope-Based Vaccine. ACS Infect Dis 2022; 8:2586-2593. [PMID: 36357959 PMCID: PMC9662650 DOI: 10.1021/acsinfecdis.2c00488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Indexed: 11/13/2022]
Abstract
The ongoing coronavirus disease 2019 pandemic has raised concerns about the risk of re-infection. Non-neutralizing epitopes are one of the major reasons for antibody-dependent enhancement. Past studies on the ancestral severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have revealed an infectivity-enhancing site on the ancestral SARS-CoV-2 spike protein. However, infection enhancement associated with the SARS-CoV-2 Omicron strain remains elusive. In this study, we examined the antibodies induced by a multiple epitope-based vaccine, which showed infection enhancement for the Omicron strain but not for the ancestral SARS-CoV-2 or Delta strain. By examining the antibodies induced by single epitope-based vaccines, we identified a conserved epitope, IDf (450-469), with neutralizing activity against ancestral SARS-CoV-2, Delta, and Omicron. Although neutralizing epitopes are present in the multiple epitope-based vaccine, other immunodominant non-neutralizing epitopes such as IDg (480-499) can shade their neutralizing activity, leading to infection enhancement of Omicron. Our study provides up-to-date epitope information on SARS-CoV-2 variants to help design better vaccines or antibody-based therapeutics against future variants.
Collapse
Affiliation(s)
- Hua-Rui Gong
- School of Biomedical Sciences, Li Ka Shing Faculty of
Medicine, University of Hong Kong, 3/F, Laboratory Block, 21
Sassoon Road, Hong kong999077, China
| | - Ye-fan Hu
- School of Biomedical Sciences, Li Ka Shing Faculty of
Medicine, University of Hong Kong, 3/F, Laboratory Block, 21
Sassoon Road, Hong kong999077, China
- Department of Medicine, School of Clinical Medicine,
University of Hong Kong, 4/F Professional Block, Queen Mary
Hospital, 102 Pokfulam Road, Hong Kong999077, China
| | - Xuechen Li
- Department of Chemistry, University of Hong
Kong, Pokfulam Road, Hong Kong999077, China
| | - Thomas Yau
- Department of Medicine, School of Clinical Medicine,
University of Hong Kong, 4/F Professional Block, Queen Mary
Hospital, 102 Pokfulam Road, Hong Kong999077, China
| | - Bao-Zhong Zhang
- Chinese Academy of Sciences (CAS) Key
Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology,
Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences,
Shenzhen518055, China
| | - Jian-Dong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of
Medicine, University of Hong Kong, 3/F, Laboratory Block, 21
Sassoon Road, Hong kong999077, China
- Chinese Academy of Sciences (CAS) Key
Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology,
Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences,
Shenzhen518055, China
- Department of Clinical Oncology, Shenzhen Key Laboratory
for Cancer Metastasis and Personalized Therapy, The University of Hong
Kong-Shenzhen Hospital, Shenzhen518055, China
- Guangdong-Hong Kong Joint Laboratory for RNA Medicine,
Sun Yat-Sen University, Guangzhou510120,
China
| |
Collapse
|
29
|
Zolfaghari MA, Ghadiri Moghaddam F, Rajput S, Karimi A, Naghi Vishteh M, Mahmoodpoor A, Dolati S, Yousefi M. SARS-CoV-2 vaccines: A double-edged sword throughout rapid evolution of COVID-19. Cell Biol Int 2022; 46:2009-2017. [PMID: 36047303 PMCID: PMC9539123 DOI: 10.1002/cbin.11903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 07/12/2022] [Accepted: 08/24/2022] [Indexed: 11/11/2022]
Abstract
After more than 2 years of the coronavirus disease 2019 pandemic caused by severe acute respiratory syndrome coronavirus 2, several questions have remained unanswered that affected our daily lives. Although substantial vaccine development could resist this challenge, emerging new variants in different countries could be considered as potent concerns regarding the adverse effects of reinfection or postvaccination. Precisely, these concerns address some significant and probable outcomes in vaccinated or reinfected models, followed by some virus challenges, such as antibody-dependent enhancement and cytokine storm. Therefore, the importance of evaluating the effectiveness of neutralizing antibodies (nAbs) elicited by vaccination and the rise of new variants must be addressed.
Collapse
Affiliation(s)
- Mohammad Ali Zolfaghari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Department of Molecular Medicine, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | | | - Shabnam Rajput
- Department of Pediatrics, School of MedicineJahrom University of Medical SciencesJahromIran
| | - Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Biotechnology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohadeseh Naghi Vishteh
- Department of Genetics and Molecular Biology, School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Ata Mahmoodpoor
- Department of AnesthesiologyTabriz University of Medical SciencesTabrizIran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| | - Mehdi Yousefi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Immunology, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
30
|
Matveeva O, Nechipurenko Y, Lagutkin D, Yegorov YE, Kzhyshkowska J. SARS-CoV-2 infection of phagocytic immune cells and COVID-19 pathology: Antibody-dependent as well as independent cell entry. Front Immunol 2022; 13:1050478. [PMID: 36532011 PMCID: PMC9751203 DOI: 10.3389/fimmu.2022.1050478] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Our review summarizes the evidence that COVID-19 can be complicated by SARS-CoV-2 infection of immune cells. This evidence is widespread and accumulating at an increasing rate. Research teams from around the world, studying primary and established cell cultures, animal models, and analyzing autopsy material from COVID-19 deceased patients, are seeing the same thing, namely that some immune cells are infected or capable of being infected with the virus. Human cells most vulnerable to infection include both professional phagocytes, such as monocytes, macrophages, and dendritic cells, as well as nonprofessional phagocytes, such as B-cells. Convincing evidence has accumulated to suggest that the virus can infect monocytes and macrophages, while data on infection of dendritic cells and B-cells are still scarce. Viral infection of immune cells can occur directly through cell receptors, but it can also be mediated or enhanced by antibodies through the Fc gamma receptors of phagocytic cells. Antibody-dependent enhancement (ADE) most likely occurs during the primary encounter with the pathogen through the first COVID-19 infection rather than during the second encounter, which is characteristic of ADE caused by other viruses. Highly fucosylated antibodies of vaccinees seems to be incapable of causing ADE, whereas afucosylated antibodies of persons with acute primary infection or convalescents are capable. SARS-CoV-2 entry into immune cells can lead to an abortive infection followed by host cell pyroptosis, and a massive inflammatory cascade. This scenario has the most experimental evidence. Other scenarios are also possible, for which the evidence base is not yet as extensive, namely productive infection of immune cells or trans-infection of other non-immune permissive cells. The chance of a latent infection cannot be ruled out either.
Collapse
Affiliation(s)
- Olga Matveeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Denis Lagutkin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Center of Phthisiopulmonology and Infectious Diseases under the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yegor E. Yegorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| |
Collapse
|
31
|
Thomas S, Smatti MK, Ouhtit A, Cyprian FS, Almaslamani MA, Thani AA, Yassine HM. Antibody-Dependent Enhancement (ADE) and the role of complement system in disease pathogenesis. Mol Immunol 2022; 152:172-182. [PMID: 36371813 PMCID: PMC9647202 DOI: 10.1016/j.molimm.2022.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Antibody-dependent enhancement (ADE) has been associated with severe disease outcomes in several viral infections, including respiratory infections. In vitro and in vivo studies showed that antibody-response to SARS-CoV and MERS-CoV could exacerbate infection via ADE. Recently in SARS CoV-2, the in vitro studies and structural analysis shows a risk of disease severity via ADE. This phenomenon is partially attributed to non-neutralizing antibodies or antibodies at sub-neutralizing levels. These antibodies result in antigen-antibody complexes' deposition and propagation of a chronic inflammatory process that destroys affected tissues. Further, antigen-antibody complexes may enhance the internalization of the virus into cells through the Fc gamma receptor (FcγR) and lead to further virus replication. Thus, ADE occur via two mechanisms; 1. Antibody mediated replication and 2. Enhanced immune activation. Antibody-mediated effector functions are mainly driven by complement activation, and the first complement in the cascade is complement 1q (C1q) which binds to the virus-antibody complex. Reports say that deficiency in circulating plasma levels of C1q, an independent predictor of mortality in high-risk patients, including diabetes, is associated with severe viral infections. Complement mediated ADE is reported in several viral infections such as dengue, West Nile virus, measles, RSV, Human immunodeficiency virus (HIV), and Ebola virus. This review discusses ADE in viral infections and the in vitro evidence of ADE in coronaviruses. We outline the mechanisms of ADE, emphasizing the role of complements, especially C1q in the outcome of the enhanced disease.
Collapse
Affiliation(s)
- Swapna Thomas
- Biomedical Research Center, Qatar University, Qatar; Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Qatar.
| | | | - Allal Ouhtit
- Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Qatar.
| | - Farhan S Cyprian
- Basic Medical Science Department, College of Medicine-QU Health, Qatar University, Qatar.
| | | | - Asmaa Al Thani
- Biomedical Research Center, Qatar University, Qatar; Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Qatar.
| | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Qatar; Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Qatar.
| |
Collapse
|
32
|
Xu Y, He Y, Momben-Abolfath S, Vertrees D, Li X, Norton MG, Struble EB. Zika Virus Infection and Antibody Neutralization in FcRn Expressing Placenta and Engineered Cell Lines. Vaccines (Basel) 2022; 10:vaccines10122059. [PMID: 36560469 PMCID: PMC9781090 DOI: 10.3390/vaccines10122059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
As a developmental toxicant, Zika virus (ZIKV) attacks both the growing nervous system, causing congenital Zika syndrome, and the placenta, resulting in pathological changes and associated adverse fetal outcomes. There are no vaccines, antibodies, or other treatments for ZIKV, despite the potential for its re-emergence. Multiple studies have highlighted the risk of antibodies for enhancing ZIKV infection, including during pregnancy, but the mechanisms for such effects are not fully understood. We have focused on the ability of the neonatal Fc receptor (FcRn) to interact with ZIKV in the presence and absence of relevant antibodies. We found that ZIKV replication was higher in Marvin Darby Canine Kidney (MDCK) cells that overexpress FcRn compared to those that do not, and knocking down FcRn decreased ZIKV RNA production. In the placenta trophoblast BeWo cell line, ZIKV infection itself downregulated FcRn at the mRNA and protein levels. Addition of anti-ZIKV antibodies to MDCK/FcRn cells resulted in non-monotonous neutralization curves with neutralization attenuation and even enhancement of infection at higher concentrations. Non-monotonous neutralization was also seen in BeWo cells at intermediate antibody concentrations. Our studies highlight the underappreciated role FcRn plays in ZIKV infection and may have implications for anti-ZIKV prophylaxis and therapy in pregnant women.
Collapse
|
33
|
Wang S, Wang CY, Kuo HK, Peng WJ, Huang JH, Kuo BS, Lin F, Liu YJ, Liu Z, Wu HT, Ding S, Hou KL, Cheng J, Yang YT, Jiang MH, Wang MS, Chen T, Xia WG, Lin E, Hung CH, Chen HJ, Shih Z, Lin YL, Ryan V, Hu MM, Heppner DG, Malherbe DC, Periasamy S, Kuzmina N, Subramani C, Hellerstein M, Monath TP, Rumyantsev A, Bukreyev A, Guirakhoo F. A Novel RBD-Protein/Peptide Vaccine Elicits Broadly Neutralizing Antibodies and Protects Mice and Macaques against SARS-CoV-2. Emerg Microbes Infect 2022; 11:2724-2734. [DOI: 10.1080/22221751.2022.2140608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
| | - Chang Yi Wang
- United Biomedical Inc., Asia, Taipei, Taiwan
- United BioPharma, HuKo, Hsin Chu, Taiwan
- United Biomedical Inc., Hauppauge, NY, USA
| | - Hui-Kai Kuo
- United Biomedical Inc., Asia, Taipei, Taiwan
| | | | | | | | - Feng Lin
- United Biomedical Inc., Hauppauge, NY, USA
| | | | - Zhi Liu
- United Biomedical Inc., Hauppauge, NY, USA
| | | | | | | | | | | | | | | | - Tony Chen
- United Biomedical Inc., Asia, Taipei, Taiwan
| | | | - Ed Lin
- United Biomedical Inc., Hauppauge, NY, USA
| | | | | | | | | | | | - Mei Mei Hu
- Vaxxinity, Inc., Dallas, TX, USA
- United BioPharma, HuKo, Hsin Chu, Taiwan
- United Biomedical Inc., Hauppauge, NY, USA
| | | | - Delphine C. Malherbe
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Sivakumar Periasamy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Natalia Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Chandru Subramani
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | | | | | | | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | | |
Collapse
|
34
|
Çölkesen F, Kepenek Kurt E, Vatansev H, Korkmaz C, Çölkesen F, Yücel F, Yıldız E, Evcen R, Aykan FS, Kılınç M, Aytekin G, Feyzioğlu B, Doğan M, Arslan Ş, Teke T, Keleş S, Reisli İ. Memory B cells and serum immunoglobulins are associated with disease severity and mortality in patients with COVID-19. Postgrad Med J 2022; 98:765-771. [PMID: 37062997 PMCID: PMC8783971 DOI: 10.1136/postgradmedj-2021-140540] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE OF THE STUDY The aim of this study was to investigate the relationship of B cell-mediated immunity with disease severity and mortality in patients with COVID-19. STUDY DESIGN In this retrospective cohort and single-centre study, 208 patients with laboratory-confirmed COVID-19 were recruited. A COVID-19 severity score, ranging from 0 to 10, was used to evaluate associations between various factors. Serum immunoglobulin levels and the number of cells in B lymphocyte subsets were measured and their association with disease severity and mortality in patients with COVID-19 examined. RESULTS The median age of the patients was 50 (35-63) years and 88 (42%) were female. The number of deceased patients was 17. The median COVID-19 severity score was 8 (6-8) in deceased patients and 1 (0-2) in survivors. Deceased patients had significantly lower levels of total B lymphocytes, naive B cells, switched memory B cells, and serum IgA, IgG, IgG1 and IgG2 than recovered patients (all p<0.05). In addition, a significant negative correlation was found between the number of these parameters and COVID-19 severity scores. Decrease in the number of total B cells and switched memory B cells as well as lower serum IgA, IgG and IgG1 levels were independent risk factors for mortality in patients with COVID-19. CONCLUSION In the present study, the prognosis of patients with COVID-19 was shown to be associated with the B cell subset and serum immunoglobulin levels.
Collapse
Affiliation(s)
- Fatih Çölkesen
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Esma Kepenek Kurt
- Department of Infectious Diseases and Clinical Microbiology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Hülya Vatansev
- Department of Chest Diseases, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Celalettin Korkmaz
- Department of Chest Diseases, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Fatma Çölkesen
- Department of Infectious Diseases and Clinical Microbiology, Konya Training and Research Hospital, Konya, Turkey
| | - Fatih Yücel
- Department of General Intensive Care Unit, Konya Training and Research Hospital, Konya, Turkey
| | - Eray Yıldız
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Recep Evcen
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Filiz Sadi Aykan
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Mehmet Kılınç
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Gökhan Aytekin
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Konya Training and Research Hospital, Konya, Turkey
| | - Bahadır Feyzioğlu
- Division of Medical Virology, Department of Medical Microbiology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Metin Doğan
- Department of Medical Microbiology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Şevket Arslan
- Division of Clinical Immunology and Allergy, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Turgut Teke
- Department of Chest Diseases, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Sevgi Keleş
- Division of Pediatric Allergy and Immunology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - İsmail Reisli
- Division of Pediatric Allergy and Immunology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| |
Collapse
|
35
|
Bigay J, Le Grand R, Martinon F, Maisonnasse P. Vaccine-associated enhanced disease in humans and animal models: Lessons and challenges for vaccine development. Front Microbiol 2022; 13:932408. [PMID: 36033843 PMCID: PMC9399815 DOI: 10.3389/fmicb.2022.932408] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The fight against infectious diseases calls for the development of safe and effective vaccines that generate long-lasting protective immunity. In a few situations, vaccine-mediated immune responses may have led to exacerbated pathology upon subsequent infection with the pathogen targeted by the vaccine. Such vaccine-associated enhanced disease (VAED) has been reported, or at least suspected, in animal models, and in a few instances in humans, for vaccine candidates against the respiratory syncytial virus (RSV), measles virus (MV), dengue virus (DENV), HIV-1, simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and the Middle East respiratory syndrome coronavirus (MERS-CoV). Although alleviated by clinical and epidemiological evidence, a number of concerns were also initially raised concerning the short- and long-term safety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is causing the ongoing COVID-19 pandemic. Although the mechanisms leading to this phenomenon are not yet completely understood, the individual and/or collective role of antibody-dependent enhancement (ADE), complement-dependent enhancement, and cell-dependent enhancement have been highlighted. Here, we review mechanisms that may be associated with the risk of VAED, which are important to take into consideration, both in the assessment of vaccine safety and in finding ways to define models and immunization strategies that can alleviate such concerns.
Collapse
Affiliation(s)
| | | | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud-INSERM U1184, CEA, Fontenay-Aux-Roses, France
| | | |
Collapse
|
36
|
Ghafouri F, Ahangari Cohan R, Samimi H, Hosseini Rad S M A, Naderi M, Noorbakhsh F, Haghpanah V. Development of a Multiepitope Vaccine Against SARS-CoV-2: Immunoinformatics Study. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2022; 3:e36100. [PMID: 35891920 PMCID: PMC9302570 DOI: 10.2196/36100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 05/16/2022] [Accepted: 07/04/2022] [Indexed: 11/21/2022]
Abstract
Background Since the first appearance of SARS-CoV-2 in China in December 2019, the world witnessed the emergence of the SARS-CoV-2 outbreak. Due to the high transmissibility rate of the virus, there is an urgent need to design and develop vaccines against SARS-CoV-2 to prevent more cases affected by the virus. Objective A computational approach is proposed for vaccine design against the SARS-CoV-2 spike (S) protein, as the key target for neutralizing antibodies, and envelope (E) protein, which contains a conserved sequence feature. Methods We used previously reported epitopes of S protein detected experimentally and further identified a collection of predicted B-cell and major histocompatibility (MHC) class II–restricted T-cell epitopes derived from E proteins with an identical match to SARS-CoV-2 E protein. Results The in silico design of our candidate vaccine against the S and E proteins of SARS-CoV-2 demonstrated a high affinity to MHC class II molecules and effective results in immune response simulations. Conclusions Based on the results of this study, the multiepitope vaccine designed against the S and E proteins of SARS-CoV-2 may be considered as a new, safe, and efficient approach to combatting the COVID-19 pandemic.
Collapse
Affiliation(s)
- Fatemeh Ghafouri
- Department of Biotechnology Faculty of Life Sciences and Biotechnology Shahid Beheshti University Tehran Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology New Technologies Research Group Pasteur Institute of Iran Tehran Iran
| | - Hilda Samimi
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | | | - Mahmood Naderi
- Digestive Diseases Research Center Digestive Diseases Research Institute Tehran University of Medical Sciences Tehran Iran
| | - Farshid Noorbakhsh
- Department of Immunology School of Medicine Tehran University of Medical Sciences Tehran Iran
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
- Personalized Medicine Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
37
|
Chen J, Xu W, Li L, Yi L, Jiang Y, Hao P, Xu Z, Zou W, Li P, Gao Z, Tian M, Jin N, Ren L, Li C. Immunogenicity and protective potential of chimeric virus-like particles containing SARS-CoV-2 spike and H5N1 matrix 1 proteins. Front Cell Infect Microbiol 2022; 12:967493. [PMID: 35923799 PMCID: PMC9339902 DOI: 10.3389/fcimb.2022.967493] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/28/2022] [Indexed: 12/17/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), has posed a constant threat to human beings and the world economy for more than two years. Vaccination is the first choice to control and prevent the pandemic. However, an effective SARS-CoV-2 vaccine against the virus infection is still needed. This study designed and prepared four kinds of virus-like particles (VLPs) using an insect expression system. Two constructs encoded wild-type SARS-CoV-2 spike (S) fused with or without H5N1 matrix 1 (M1) (S and SM). The other two constructs contained a codon-optimized spike gene and/or M1 gene (mS and mSM) based on protein expression, stability, and ADE avoidance. The results showed that the VLP-based vaccine could induce high SARS-CoV-2 specific antibodies in mice, including specific IgG, IgG1, and IgG2a. Moreover, the mSM group has the most robust ability to stimulate humoral immunity and cellular immunity than the other VLPs, suggesting the mSM is the best immunogen. Further studies showed that the mSM combined with Al/CpG adjuvant could stimulate animals to produce sustained high-level antibodies and establish an effective protective barrier to protect mice from challenges with mouse-adapted strain. The vaccine based on mSM and Al/CpG adjuvant is a promising candidate vaccine to prevent the COVID-19 pandemic.
Collapse
Affiliation(s)
- Jing Chen
- College of Veterinary medicine, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, Changchun, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Wang Xu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Lichao Yi
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuhang Jiang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Pengfei Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhiqiang Xu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Wancheng Zou
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Peiheng Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zihan Gao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Mingyao Tian
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- *Correspondence: Chang Li, ; Linzhu Ren, ; Ningyi Jin,
| | - Linzhu Ren
- College of Animal Sciences, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, Changchun, China
- *Correspondence: Chang Li, ; Linzhu Ren, ; Ningyi Jin,
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- *Correspondence: Chang Li, ; Linzhu Ren, ; Ningyi Jin,
| |
Collapse
|
38
|
Tien CF, Tsai WT, Chen CH, Chou HJ, Zhang MM, Lin JJ, Lin EJ, Dai SS, Ping YH, Yu CY, Kuo YP, Tsai WH, Chen HW, Yu GY. Glycosylation and S-palmitoylation regulate SARS-CoV-2 spike protein intracellular trafficking. iScience 2022; 25:104709. [PMID: 35813875 PMCID: PMC9250814 DOI: 10.1016/j.isci.2022.104709] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/19/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Post-translational modifications (PTMs), such as glycosylation and palmitoylation, are critical to protein folding, stability, intracellular trafficking, and function. Understanding regulation of PTMs of SARS-CoV-2 spike (S) protein could help the therapeutic drug design. Herein, the VSV vector was used to produce SARS-CoV-2 S pseudoviruses to examine the roles of the 611LYQD614 and cysteine-rich motifs in S protein maturation and virus infectivity. Our results show that 611LY612 mutation alters S protein intracellular trafficking and reduces cell surface expression level. It also changes S protein glycosylation pattern and decreases pseudovirus infectivity. The S protein contains four cysteine-rich clusters with clusters I and II as the main palmitoylation sites. Mutations of clusters I and II disrupt S protein trafficking from ER-to-Golgi, suppress pseudovirus production, and reduce spike-mediated membrane fusion activity. Taken together, glycosylation and palmitoylation orchestrate the S protein maturation processing and are critical for S protein-mediated membrane fusion and infection. 611LY612 mutation alters the glycosylation pattern of the SARS-CoV-2 S protein 611LY612 mutation reduces S protein surface expression level Palmitoylation targets mature S protein to the Golgi and plasma membrane Palmitoylation is required for pseudovirus and SARS-CoV-2 production
Collapse
|
39
|
Shalash AO, Toth I, Skwarczynski M. The potential of developing a protective peptide-based vaccines against SARS-CoV-2. Drug Dev Res 2022; 83:1251-1256. [PMID: 35751566 PMCID: PMC9349783 DOI: 10.1002/ddr.21969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/20/2022] [Accepted: 06/14/2022] [Indexed: 12/11/2022]
Abstract
COVID‐19 pandemic has been the deadliest infectious disease outbreak since Spanish flu. The emerging variant lineages, decay of neutralizing antibodies, and occur of reinfections require the development of highly protective and safe vaccines. As currently approved COVID‐19 vaccines that utilize virus‐related genetic material are less than ideal, other vaccine types have been also widely investigated. Among them, peptide‐based vaccines hold great promise in countering COVID‐19 as they may overcome most of the shortcomings of RNA/DNA and protein vaccines. Two basic types of potential peptide vaccines can be developed. The first type are those which rely on cytotoxic T‐cell (CTL) responses to kill infected host cells and stop the replication via employing CTL‐epitopes as vaccine antigens. The second type of peptide vaccines are those that rely on B‐cell peptide epitopes to trigger humoral response via generating SARS‐CoV‐2‐specific antibodies to neutralize and/or opsonize the virus. We propose that combining both cellular and humoral immune responses would be highly protective. Here we discuss opportunities and challenges in the development of an effective and safe peptide‐based vaccine against COVID‐19.
Collapse
Affiliation(s)
- Ahmed O Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
40
|
Guérin P, Yahi N, Azzaz F, Chahinian H, Sabatier JM, Fantini J. Structural Dynamics of the SARS-CoV-2 Spike Protein: A 2-Year Retrospective Analysis of SARS-CoV-2 Variants (from Alpha to Omicron) Reveals an Early Divergence between Conserved and Variable Epitopes. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123851. [PMID: 35744971 PMCID: PMC9230616 DOI: 10.3390/molecules27123851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 12/17/2022]
Abstract
We analyzed the epitope evolution of the spike protein in 1,860,489 SARS-CoV-2 genomes. The structural dynamics of these epitopes was determined by molecular modeling approaches. The D614G mutation, selected in the first months of the pandemic, is still present in currently circulating SARS-CoV-2 strains. This mutation facilitates the conformational change leading to the demasking of the ACE2 binding domain. D614G also abrogated the binding of facilitating antibodies to a linear epitope common to SARS-CoV-1 and SARS-CoV-2. The main neutralizing epitope of the N-terminal domain (NTD) of the spike protein showed extensive structural variability in SARS-CoV-2 variants, especially Delta and Omicron. This epitope is located on the flat surface of the NTD, a large electropositive area which binds to electronegatively charged lipid rafts of host cells. A facilitating epitope located on the lower part of the NTD appeared to be highly conserved among most SARS-CoV-2 variants, which may represent a risk of antibody-dependent enhancement (ADE). Overall, this retrospective analysis revealed an early divergence between conserved (facilitating) and variable (neutralizing) epitopes of the spike protein. These data aid in the designing of new antiviral strategies that could help to control COVID-19 infection by mimicking neutralizing antibodies or by blocking facilitating antibodies.
Collapse
Affiliation(s)
| | - Nouara Yahi
- INSERM UMR_S 1072, Aix-Marseille University, CEDEX, 13015 Marseille, France; (N.Y.); (F.A.); (H.C.)
| | - Fodil Azzaz
- INSERM UMR_S 1072, Aix-Marseille University, CEDEX, 13015 Marseille, France; (N.Y.); (F.A.); (H.C.)
| | - Henri Chahinian
- INSERM UMR_S 1072, Aix-Marseille University, CEDEX, 13015 Marseille, France; (N.Y.); (F.A.); (H.C.)
| | - Jean-Marc Sabatier
- Inst Neurophysiopathol, Aix-Marseille University, CNRS, INP, CEDEX, 13005 Marseille, France;
| | - Jacques Fantini
- INSERM UMR_S 1072, Aix-Marseille University, CEDEX, 13015 Marseille, France; (N.Y.); (F.A.); (H.C.)
- Correspondence:
| |
Collapse
|
41
|
Zhou W, Xu C, Luo M, Wang P, Xu Z, Xue G, Jin X, Huang Y, Li Y, Nie H, Jiang Q, Anashkina AA. MutCov: A pipeline for evaluating the effect of mutations in spike protein on infectivity and antigenicity of SARS-CoV-2. Comput Biol Med 2022; 145:105509. [PMID: 35421792 PMCID: PMC8993498 DOI: 10.1016/j.compbiomed.2022.105509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/16/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing an outbreak of coronavirus disease 2019 (COVID-19), is a major threat to public health worldwide. Previous studies have shown that the spike protein of SARS-CoV-2 determines viral infectivity and major antigenicity. However, the spike protein has been undergoing various mutations, which bring a great challenge to the prevention and treatment of COVID-19. Here we present the MutCov, a pipeline for evaluating the effect of mutations in spike protein on infectivity and antigenicity of SARS-CoV-2 by calculating the binding free energy between spike protein and angiotensin-converting enzyme 2 (ACE2) or neutralizing monoclonal antibody (mAb). The predicted infectivity and antigenicity were highly consistent with biologically experimental results, and demonstrated that the MutCov achieved good prediction performance. In conclusion, the MutCov is of high importance for systematically evaluating the effect of novel mutations and improving the prevention and treatment of COVID-19. The source code and installation instruction of MutCov are freely available at http://jianglab.org.cn/MutCov.
Collapse
Affiliation(s)
- Wenyang Zhou
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Chang Xu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Meng Luo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Pingping Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Zhaochun Xu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Guangfu Xue
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Xiyun Jin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Yan Huang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Yiqun Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Huan Nie
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Qinghua Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China.
| | - Anastasia A Anashkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
42
|
Farouq MAH, Acevedo R, Ferro VA, Mulheran PA, Al Qaraghuli MM. The Role of Antibodies in the Treatment of SARS-CoV-2 Virus Infection, and Evaluating Their Contribution to Antibody-Dependent Enhancement of Infection. Int J Mol Sci 2022; 23:6078. [PMID: 35682757 PMCID: PMC9181534 DOI: 10.3390/ijms23116078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023] Open
Abstract
Antibodies play a crucial role in the immune response, in fighting off pathogens as well as helping create strong immunological memory. Antibody-dependent enhancement (ADE) occurs when non-neutralising antibodies recognise and bind to a pathogen, but are unable to prevent infection, and is widely known and is reported as occurring in infection caused by several viruses. This narrative review explores the ADE phenomenon, its occurrence in viral infections and evaluates its role in infection by SARS-CoV-2 virus, which causes coronavirus disease 2019 (COVID-19). As of yet, there is no clear evidence of ADE in SARS-CoV-2, though this area is still subject to further study.
Collapse
Affiliation(s)
- Mohammed A. H. Farouq
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
| | - Reinaldo Acevedo
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| | - Valerie A. Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
| | - Paul A. Mulheran
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
| | - Mohammed M. Al Qaraghuli
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
- EPSRC Future Manufacturing Research Hub for Continuous Manufacturing and Advanced Crystallisation (CMAC), University of Strathclyde, 99 George Street, Glasgow G1 1RD, UK
| |
Collapse
|
43
|
Luan N, Li T, Wang Y, Cao H, Yin X, Lin K, Liu C. Th2-Oriented Immune Serum After SARS-CoV-2 Vaccination Does Not Enhance Infection In Vitro. Front Immunol 2022; 13:882856. [PMID: 35464483 PMCID: PMC9024142 DOI: 10.3389/fimmu.2022.882856] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
The relatively lower protection rate of the alum-adjuvanted inactivated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines reminds us of the antibody-dependent enhancement (ADE) phenomenon observed in preclinical studies during the development of vaccines for Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1). In this study, using the S1 segment of the SARS-CoV-2 spike protein or inactivated whole SARS-CoV-2 virus as an antigen and aluminum as an adjuvant, the risk of ADE of infection with T helper 2 (Th2)-oriented immune serum from mice (N=6) and humans (N=5) was examined in immune cell lines, which show different expression patterns of Fc receptors. Neither the immune serum from alum-adjuvanted S1 subunit vaccines nor inactivated SARS-CoV-2 vaccination enhanced SARS-CoV-2 S pseudotyped virus infection in any of the tested cell lines in vitro. Because both of these Th2-oriented immune sera could block SARS-CoV-2 infection without ADE of infection, we speculate that the lower protection rate of the inactivated SARS-CoV-2 vaccine may be attributed to the lower neutralizing antibody titers induced or the pulmonary eosinophilic immunopathology accompanied by eosinophilic infiltration in the lungs upon virus exposure. Adjustment of the immunization schedule to elevate the neutralizing antibody levels and skew adjuvants toward Th1-oriented responses may be considered to increase the efficacies of both inactivated and spike protein-based subunit SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Ning Luan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Tao Li
- Institute for Biological Product Control, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Yunfei Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Xingxiao Yin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Kangyang Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| |
Collapse
|
44
|
Abstract
Antibody-dependent enhancement (ADE) of infection is generally known for many viruses. A potential risk of ADE in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has also been discussed since the beginning of the coronavirus disease 2019 (COVID-19) pandemic; however, clinical evidence of the presence of antibodies with ADE potential is limited. Here, we show that ADE antibodies are produced by SARS-CoV-2 infection and the ADE process can be mediated by at least two different host factors, Fcγ receptor (FcγR) and complement component C1q. Of 89 serum samples collected from acute or convalescent COVID-19 patients, 62.9% were found to be positive for SARS-CoV-2-specific IgG. FcγR- and/or C1q-mediated ADE were detected in 50% of the IgG-positive sera, whereas most of them showed neutralizing activity in the absence of FcγR and C1q. Importantly, ADE antibodies were found in 41.4% of the acute COVID-19 patients. Neutralizing activity was also detected in most of the IgG-positive sera, but it was counteracted by ADE in subneutralizing conditions in the presence of FcγR or C1q. Although the clinical importance of ADE needs to be further investigated with larger numbers of COVID-19 patient samples, our data suggest that SARS-CoV-2 utilizes multiple mechanisms of ADE. C1q-mediated ADE may particularly have a clinical impact since C1q is present at high concentrations in plasma and its receptors are ubiquitously expressed on the surfaces of many types of cells, including respiratory epithelial cells, which SARS-CoV-2 primarily infects. IMPORTANCE Potential risks of antibody-dependent enhancement (ADE) in the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been discussed and the proposed mechanism mostly depends on the Fc gamma receptor (FcγR). However, since FcγRs are exclusively expressed on immune cells, which are not primary targets of SARS-CoV-2, the clinical importance of ADE of SARS-CoV-2 infection remains controversial. Our study demonstrates that SARS-CoV-2 infection induces antibodies that increase SARS-CoV-2 infection through another ADE mechanism in which complement component C1q mediates the enhancement. Although neutralizing activity was also detected in the serum samples, it was counteracted by ADE in the presence of FcγR or C1q. Considering the ubiquity of C1q and its cellular receptors, C1q-mediated ADE may more likely occur in respiratory epithelial cells, which SARS-CoV-2 primarily infects. Our data highlight the importance of careful monitoring of the antibody properties in COVID-19 convalescent and vaccinated individuals.
Collapse
|
45
|
Chattopadhyay A, Saigal Kalra K, Saikia D, Yadav V, Chouksey J, Jajoo M, Sherwal BL. Severe Multi-inflammatory Syndrome in Children Temporally Related to COVID 19-Clinical Course, Laboratory Profile and Outcomes from a North Indian PICU. J Intensive Care Med 2022; 37:1229-1237. [PMID: 35469487 DOI: 10.1177/08850666221092302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective: We describe the trajectory of clinical course, laboratory markers and outcomes in children with severe multi-inflammatory syndrome temporally related to COVID-19 (MIS-C) admitted to our pediatric intensive care unit (PICU). Methods: This was a prospective case series of children admitted to PICU between May 1, 2020 and January 31, 2021, fulfilling the case definition of MIS-C published by World Health Organization (WHO) or Centers for Disease Control and Prevention (CDC). We analyzed demographic, clinical, laboratory data and echocardiographic findings. We also plotted the variation in trends between survivors and nonsurvivors. Results: Of the 34 critically ill children referred to PICU with diagnosis of MIS-C only 17 fulfilled the WHO/CDC classification of MIS-C, rest were MISC mimickers albeit other tropical infections. Median age at admission was 4 years (range 1y 6 mo-8 years). Fever, rash and conjunctival redness were most prominent symptoms. Myocardial involvement was seen in 70.5% while 76.4% developed shock; Invasive mechanical ventilation was required in 64.7% cases. Inflammatory markers showed a downward trend such as-median C- reactive protein (mg/L) had a serial reduction in levels-from (median/IQR) 210 (132.60, 246.90) at admission to 52.3 (42, 120) on Day 3. Median Ferritin (ng/ml) (n = 12) was 690 (203, 1324), serum LDH (IU/L) (n = 12) was 505 (229.5, 1032) and Mean D-dimer (ng/ml) (n = 7) was 5093.85 (1991.65), suggestive of hyperinflammatory syndrome. Twelve patients received intravenous immune globulin, with adjunctive steroid therapy used in two third of the cases. Six children died, 4 of them were under-5 years of age. Tocilizumab was prescribed in two children with high vasotrope inotrope score (VIS), cardiogenic shock and oxygenation index more than 15, both survived. Conclusions: Severe MIS-C has a heterogenous presentation, local or regional outbreaks of prevalent infectious diseases often lead to confusion and overdiagnosis. Higher proportion of mortality was seen in Under -5 children with MISC. Shock-like presentation, presence of myocardial dysfunction or nonsurvivor status is associated with higher trend of inflammatory markers and more profound multi-organ dysfunction. If disease progresses rapidly despite first line therapy (IvIg and steroids), use of Tocilizumab should be considered-as a rescue therapy under resource limitations in the absence of extracorporeal support.
Collapse
Affiliation(s)
- Arpita Chattopadhyay
- Department of Pediatrics, 75299Chacha Nehru Bal Chikitsalaya, Geeta Colony, New Delhi, India
| | - Karnika Saigal Kalra
- Department of Microbiology, 75299Chacha Nehru Bal Chikitsalaya, Geeta Colony, New Delhi, India
| | - Diganta Saikia
- Department of Pediatrics, 75299Chacha Nehru Bal Chikitsalaya, Geeta Colony, New Delhi, India
| | - Varshanjali Yadav
- Department of Pediatrics, 75299Chacha Nehru Bal Chikitsalaya, Geeta Colony, New Delhi, India
| | - Juhi Chouksey
- Department of Pediatrics, 75299Chacha Nehru Bal Chikitsalaya, Geeta Colony, New Delhi, India
| | - Mamta Jajoo
- Department of Pediatrics, 75299Chacha Nehru Bal Chikitsalaya, Geeta Colony, New Delhi, India
| | - B L Sherwal
- Director, Chacha Nehru Bal Chikitsalaya, Geeta Colony, New Delhi, India
| |
Collapse
|
46
|
Long Q, Yang Y, Yang M, Bai H, Sun W, Yang X, Huang W, Li D, Ma Y. Recombinant VLPs empower RBM peptides showing no immunogenicity in native SARS-COV-2 protein to elicit a robust neutralizing antibody response. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 41:102527. [PMID: 35104670 PMCID: PMC8800570 DOI: 10.1016/j.nano.2022.102527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/11/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022]
Abstract
New SARS-COV-2 vaccine strategies are still urgently needed, especially for emerging virus mutations and variants. In this study, we focused on analyzing the antigenicity and vaccine potency of linear peptide epitopes located in receptor binding motif (RBM) of spike (S) protein. Nine 12 to 16-mer overlapping peptides (P1-P9) were synthesized chemically and coupled to carrier protein KLH for the immunization in mice. Four of identified peptides were further engineered to present on the surface of recombinant Hepatitis B core antigen (HBcAg) virus-like particles (VLPs) respectively. Antisera obtained from VLPs -immunized mice demonstrated strong reactivity and affinity to S1 protein or inactivated virus and neutralizing activity against virus infection in vitro. This study indicates that recombinant VLPs empower peptides which display underprivileged antigenicity in native protein to elicit high levels of neutralizing antibody, providing potential epitope candidates and an effective delivery strategy for the development of a multi-epitope vaccine.
Collapse
Affiliation(s)
- Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Mengli Yang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Wenjia Sun
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Center for Diseases Control and Prevention, Kunming, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China.
| |
Collapse
|
47
|
Kovalenko AO, Ryabchevskaya EM, Evtushenko EA, Manukhova TI, Kondakova OA, Ivanov PA, Arkhipenko MV, Gushchin VA, Nikitin NA, Karpova OV. Vaccine Candidate Against COVID-19 Based on Structurally Modified Plant Virus as an Adjuvant. Front Microbiol 2022; 13:845316. [PMID: 35295298 PMCID: PMC8919459 DOI: 10.3389/fmicb.2022.845316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
A recombinant vaccine candidate has been developed based on the major coronaviruses’ antigen (S protein) fragments and a novel adjuvant—spherical particles (SPs) formed during tobacco mosaic virus thermal remodeling. The receptor-binding domain and the highly conserved antigenic fragments of the S2 protein subunit were chosen for the design of recombinant coronavirus antigens. The set of three antigens (Co1, CoF, and PE) was developed and used to create a vaccine candidate composed of antigens and SPs (SPs + 3AG). Recognition of SPs + 3AG compositions by commercially available antibodies against spike proteins of SARS-CoV and SARS-CoV-2 was confirmed. The immunogenicity testing of these compositions in a mouse model showed that SPs improved immune response to the CoF and PE antigens. Total IgG titers against both proteins were 9–16 times higher than those to SPs. Neutralizing activity against SARS-CoV-2 in serum samples collected from hamsters immunized with the SPs + 3AG was demonstrated.
Collapse
Affiliation(s)
- Angelina O Kovalenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Ekaterina A Evtushenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana I Manukhova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga A Kondakova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Peter A Ivanov
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Marina V Arkhipenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir A Gushchin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nikolai A Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga V Karpova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
48
|
Crowley AR, Natarajan H, Hederman AP, Bobak CA, Weiner JA, Wieland-Alter W, Lee J, Bloch EM, Tobian AAR, Redd AD, Blankson JN, Wolf D, Goetghebuer T, Marchant A, Connor RI, Wright PF, Ackerman ME. Boosting of cross-reactive antibodies to endemic coronaviruses by SARS-CoV-2 infection but not vaccination with stabilized spike. eLife 2022; 11:e75228. [PMID: 35289271 PMCID: PMC8923670 DOI: 10.7554/elife.75228] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Preexisting antibodies to endemic coronaviruses (CoV) that cross-react with SARS-CoV-2 have the potential to influence the antibody response to COVID-19 vaccination and infection for better or worse. In this observational study of mucosal and systemic humoral immunity in acutely infected, convalescent, and vaccinated subjects, we tested for cross-reactivity against endemic CoV spike (S) protein at subdomain resolution. Elevated responses, particularly to the β-CoV OC43, were observed in all natural infection cohorts tested and were correlated with the response to SARS-CoV-2. The kinetics of this response and isotypes involved suggest that infection boosts preexisting antibody lineages raised against prior endemic CoV exposure that cross-react. While further research is needed to discern whether this recalled response is desirable or detrimental, the boosted antibodies principally targeted the better-conserved S2 subdomain of the viral spike and were not associated with neutralization activity. In contrast, vaccination with a stabilized spike mRNA vaccine did not robustly boost cross-reactive antibodies, suggesting differing antigenicity and immunogenicity. In sum, this study provides evidence that antibodies targeting endemic CoV are robustly boosted in response to SARS-CoV-2 infection but not to vaccination with stabilized S, and that depending on conformation or other factors, the S2 subdomain of the spike protein triggers a rapidly recalled, IgG-dominated response that lacks neutralization activity.
Collapse
Affiliation(s)
- Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth CollegeHanoverUnited States
| | - Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth CollegeHanoverUnited States
| | | | - Carly A Bobak
- Biomedical Data Science, Dartmouth CollegeHanoverUnited States
| | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth CollegeHanoverUnited States
| | - Wendy Wieland-Alter
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical CenterLebanonUnited States
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth CollegeHanoverUnited States
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Aaron AR Tobian
- Department of Pathology, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Andrew D Redd
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of MedicineBaltimoreUnited States
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaUnited States
| | - Joel N Blankson
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Dana Wolf
- Hadassah University Medical CenterJerusalemIsrael
| | - Tessa Goetghebuer
- Institute for Medical Immunology, Université libre de BruxellesCharleroiBelgium
- Pediatric Department, CHU St PierreBrusselsBelgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de BruxellesCharleroiBelgium
| | - Ruth I Connor
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical CenterLebanonUnited States
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical CenterLebanonUnited States
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth CollegeHanoverUnited States
- Thayer School of Engineering, Dartmouth CollegeHanoverUnited States
- Biomedical Data Science, Dartmouth CollegeHanoverUnited States
| |
Collapse
|
49
|
Pieri M, Nicolai E, Ciotti M, Nuccetelli M, Sarubbi S, Pelagalli M, Bernardini S. Antibody response to COVID-19 vaccine: A point of view that can help to optimize dose distribution. Int Immunopharmacol 2022; 102:108406. [PMID: 34862126 PMCID: PMC8626226 DOI: 10.1016/j.intimp.2021.108406] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 11/05/2022]
Abstract
The global strategy to control coronavirus disease is based on the availability of COVID-19 vaccines. More information about response to a single dose vaccine could help to better understand and optimize the management of the vaccine campaign. Workers from the University of Rome "Tor Vergata" and the University Hospital of University of Rome "Tor Vergata," were monitored during their vaccination program. Serum samples were collected between the first and second dose and after the second dose. University personnel has been vaccinated with two doses of Vaxzevria vaccine 12 weeks apart, while hospital personnel has been vaccinated with two doses of Comirnaty 3 weeks apart. IgG antibodies (Abs) against the Receptor Binding Domain (RBD) of the virus spike surface glycoprotein and neutralizing antibodies (NT) anti-SARS-CoV-2 that block the interaction between RBD and the surface receptor cellular angiotensin converting enzyme (ACE2) were measured using the CL-series Mindray chemiluminescent assays, respectively. Different amounts of antibodies produced after the two doses of vaccine were found. Individuals with a previous natural infection developed a higher Abs titer. Among the individuals with no history of past SARS-CoV-2 infection, 5% had an Abs level of the same order of magnitude of infected people, suggesting that they acquired the infection in an asymptomatic way. In such individuals, one dose of vaccine may be sufficient to obtain a protective immune response.
Collapse
Affiliation(s)
- Massimo Pieri
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; Department of Laboratory Medicine, Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Eleonora Nicolai
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| | - Marco Ciotti
- Virology Unit, Laboratory of Clinical Microbiology and Virology, Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Marzia Nuccetelli
- Department of Laboratory Medicine, Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Serena Sarubbi
- Department of Laboratory Medicine, Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Martina Pelagalli
- Department of Laboratory Medicine, Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; Department of Laboratory Medicine, Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy; IFCC Emerging Technologies Division, Via Carlo Farini 81, 20159 Milan, Italy
| |
Collapse
|
50
|
Qiang X, Zhu S, Li J, Chen W, Yang H, Wang P, Tracey KJ, Wang H. Monoclonal antibodies capable of binding SARS-CoV-2 spike protein receptor-binding motif specifically prevent GM-CSF induction. J Leukoc Biol 2022; 111:261-267. [PMID: 33759207 PMCID: PMC8251270 DOI: 10.1002/jlb.3covcra0920-628rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/21/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
A severe acute respiratory syndrome (SARS)-like coronavirus 2 (SARS-CoV-2) has recently caused a pandemic COVID-19 disease that infected approximately 94 million and killed more than 2,000,000 people worldwide. Like the SARS-CoV, SARS-CoV-2 also employs a receptor-binding motif (RBM) of its envelope spike protein for binding the host angiotensin-converting enzyme 2 (ACE2) to gain viral entry. Currently, extensive efforts are being made to produce vaccines against a surface fragment of a SARS-CoV-2, such as the spike protein, in order to boost protective antibodies that can inhibit virus-ACE2 interaction to prevent viral entry. It was previously unknown how spike protein-targeting antibodies would affect innate inflammatory responses to SARS-CoV-2 infections. Here we generated a highly purified recombinant protein corresponding to the RBM of SARS-CoV-2, and used it to screen for cross-reactive monoclonal antibodies (mAbs). We found two RBM-binding mAbs that competitively inhibited its interaction with human ACE2, and specifically blocked the RBM-induced GM-CSF secretion in both human peripheral blood mononuclear cells and murine macrophage cultures. Our findings have suggested a possible strategy to prevent SARS-CoV-2-elicited "cytokine storm," and revealed a potentially anti-inflammatory and protective mechanism for SARS-CoV-2 spike-based vaccines.
Collapse
Affiliation(s)
- Xiaoling Qiang
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNew YorkUSA
| | - Shu Zhu
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNew YorkUSA
| | - Jianhua Li
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
| | - Weiqiang Chen
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNew YorkUSA
| | - Huan Yang
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
| | - Ping Wang
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNew YorkUSA
| | - Kevin J. Tracey
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNew YorkUSA
| | - Haichao Wang
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNew YorkUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNew YorkUSA
| |
Collapse
|