1
|
Ying B, Liang CY, Desai P, Scheaffer SM, Elbashir SM, Edwards DK, Thackray LB, Diamond MS. Ipsilateral or contralateral boosting of mice with mRNA vaccines confers equivalent immunity and protection against a SARS-CoV-2 Omicron strain. J Virol 2024; 98:e0057424. [PMID: 39194250 PMCID: PMC11406931 DOI: 10.1128/jvi.00574-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Boosting with mRNA vaccines encoding variant-matched spike proteins has been implemented to mitigate their reduced efficacy against emerging SARS-CoV-2 variants. Nonetheless, in humans, it remains unclear whether boosting in the ipsilateral or contralateral arm with respect to the priming doses impacts immunity and protection. Here, we boosted K18-hACE2 mice with either monovalent mRNA-1273 (Wuhan-1 spike) or bivalent mRNA-1273.214 (Wuhan-1 + BA.1 spike) vaccine in the ipsilateral or contralateral leg after a two-dose priming series with mRNA-1273. Boosting in the ipsilateral or contralateral leg elicited equivalent levels of serum IgG and neutralizing antibody responses against Wuhan-1 and BA.1. While contralateral boosting with mRNA vaccines resulted in the expansion of spike-specific B and T cells beyond the ipsilateral draining lymph node (DLN) to the contralateral DLN, administration of a third mRNA vaccine dose at either site resulted in similar levels of antigen-specific germinal center B cells, plasmablasts/plasma cells, T follicular helper cells, and CD8+ T cells in the DLNs and the spleen. Furthermore, ipsilateral and contralateral boosting with mRNA-1273 or mRNA-1273.214 vaccines conferred similar homologous or heterologous immune protection against SARS-CoV-2 BA.1 virus challenge with equivalent reductions in viral RNA and infectious virus in the nasal turbinates and lungs. Collectively, our data show limited differences in B and T cell immune responses after ipsilateral and contralateral site boosting by mRNA vaccines that do not substantively impact protection against an Omicron strain.IMPORTANCESequential boosting with mRNA vaccines has been an effective strategy to overcome waning immunity and neutralization escape by emerging SARS-CoV-2 variants. However, it remains unclear how the site of boosting relative to the primary vaccination series shapes optimal immune responses or breadth of protection against variants. In K18-hACE2 transgenic mice, we observed that intramuscular boosting with historical monovalent or variant-matched bivalent vaccines in the ipsilateral or contralateral limb elicited comparable levels of serum spike-specific antibody and antigen-specific B and T cell responses. Moreover, boosting on either side conferred equivalent protection against a SARS-CoV-2 Omicron challenge strain. Our data in mice suggest that the site of intramuscular boosting with an mRNA vaccine does not substantially impact immunity or protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Baoling Ying
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chieh-Yu Liang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Yang CH, Shen KY, Ho HM, Huang CY, Cheng YJ, Pu CC, Chiu FF, Huang WC, Liao HC, Chen HW, Liao CL, Liu SJ, Huang MH. Boosting DNA vaccine power by lipid nanoparticles surface engineered with amphiphilic bioresorbable copolymer. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102261. [PMID: 39071950 PMCID: PMC11278320 DOI: 10.1016/j.omtn.2024.102261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/14/2024] [Indexed: 07/30/2024]
Abstract
Successful DNA vaccination generally requires the aid of either a viral vector within vaccine components or an electroporation device into the muscle or skin of the host. However, these systems come with certain obstacles, including limited transgene capacity, broad preexisting immunity in humans, and substantial cell death caused by high voltage pulses, respectively. In this study, we repurposed the use of an amphiphilic bioresorbable copolymer (ABC), called PLA-PEG, as a surface engineering agent that conciliates lipid nanoparticles (LNPs) between stability during preparation and biocompatibility post-vaccination. The LNP carrier can be loaded with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike-specific DNA; in this form, the DNA-LNP is immunogenic in hamsters and elicits protective immunity following DNA-LNP vaccination against heterologous virus challenge or as a hybrid-type vaccine booster against SARS-CoV-2 variants. The data provide comprehensive information on the relationships between LNP composition, manufacturing process, and vaccine efficacy. The outcomes of this study offer new insights into designing next-generation LNP formulations and pave the way for boosting vaccine power to combat existing and possible emerging infectious diseases/pathogens.
Collapse
Affiliation(s)
- Chung-Hsiang Yang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Kuan-Yin Shen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hui-Min Ho
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chiung-Yi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yu-Jhen Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chih-Chun Pu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Fang-Feng Chiu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Wan-Chun Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Hsi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
3
|
Kumari M, Liang KH, Su SC, Lin HT, Lu YF, Wu MJ, Chen WY, Wu HC. Multivalent mRNA Vaccine Elicits Broad Protection against SARS-CoV-2 Variants of Concern. Vaccines (Basel) 2024; 12:714. [PMID: 39066352 PMCID: PMC11281580 DOI: 10.3390/vaccines12070714] [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: 05/10/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
SARS-CoV-2 new waves are primarily caused by changes to the spike protein (S), which can substantially decrease the efficacy of vaccines. Therefore, we tested several multivalent mRNA-LNP vaccines, targeting the full-length S proteins of different variants, and identified an optimal combination for protection against VOCs in BALB/c mice. The tested formulations included trivalent (WT + BA.5 + XBB.1.5), pentavalent (WT + BA.5 + XBB.1.5 + BQ.1.1 + CH.1.1), and octavalent (WT + BA.5 + XBB.1.5 + BQ.1.1 + CH.1.1 + Alpha + Delta + BA.2) vaccines. Among these multivalent vaccines, the pentavalent vaccine showed superior protection for almost all tested variants. Despite this, each multivalent vaccine elicited greater broad-spectrum neutralizing antibodies than the previously evaluated bivalent vaccine (WT + BA.5). Subsequently, we redesigned the multivalent vaccine to efficiently generate neutralizing antibodies against recent VOCs, including EG.5.1. Immunization with the redesigned pentavalent vaccine (WT + EG.5.1 + XBB.1.16 + Delta + BA.5) showed moderate levels of protection against recent Omicron VOCs. Results suggest that the neutralization activity of multivalent vaccines is better than those of the tested bivalent vaccines against WT + BA.5 and WT + EG.5.1. Moreover, the pentavalent vaccine we developed may be highly useful for neutralizing new Omicron VOCs.
Collapse
Affiliation(s)
- Monika Kumari
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (M.K.); (S.-C.S.); (H.-T.L.); (Y.-F.L.); (M.-J.W.); (W.-Y.C.)
| | - Kang-Hao Liang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei 11529, Taiwan;
| | - Shih-Chieh Su
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (M.K.); (S.-C.S.); (H.-T.L.); (Y.-F.L.); (M.-J.W.); (W.-Y.C.)
| | - Hsiu-Ting Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (M.K.); (S.-C.S.); (H.-T.L.); (Y.-F.L.); (M.-J.W.); (W.-Y.C.)
| | - Yu-Feng Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (M.K.); (S.-C.S.); (H.-T.L.); (Y.-F.L.); (M.-J.W.); (W.-Y.C.)
| | - Ming-Jane Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (M.K.); (S.-C.S.); (H.-T.L.); (Y.-F.L.); (M.-J.W.); (W.-Y.C.)
| | - Wan-Yu Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (M.K.); (S.-C.S.); (H.-T.L.); (Y.-F.L.); (M.-J.W.); (W.-Y.C.)
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan; (M.K.); (S.-C.S.); (H.-T.L.); (Y.-F.L.); (M.-J.W.); (W.-Y.C.)
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei 11529, Taiwan;
| |
Collapse
|
4
|
Rojas Chávez RA, Fili M, Han C, Rahman SA, Bicar IGL, Gregory S, Helverson A, Hu G, Darbro BW, Das J, Brown GD, Haim H. Mapping the Evolutionary Space of SARS-CoV-2 Variants to Anticipate Emergence of Subvariants Resistant to COVID-19 Therapeutics. PLoS Comput Biol 2024; 20:e1012215. [PMID: 38857308 PMCID: PMC11192331 DOI: 10.1371/journal.pcbi.1012215] [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: 07/06/2023] [Revised: 06/21/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024] Open
Abstract
New sublineages of SARS-CoV-2 variants-of-concern (VOCs) continuously emerge with mutations in the spike glycoprotein. In most cases, the sublineage-defining mutations vary between the VOCs. It is unclear whether these differences reflect lineage-specific likelihoods for mutations at each spike position or the stochastic nature of their appearance. Here we show that SARS-CoV-2 lineages have distinct evolutionary spaces (a probabilistic definition of the sequence states that can be occupied by expanding virus subpopulations). This space can be accurately inferred from the patterns of amino acid variability at the whole-protein level. Robust networks of co-variable sites identify the highest-likelihood mutations in new VOC sublineages and predict remarkably well the emergence of subvariants with resistance mutations to COVID-19 therapeutics. Our studies reveal the contribution of low frequency variant patterns at heterologous sites across the protein to accurate prediction of the changes at each position of interest.
Collapse
Affiliation(s)
| | - Mohammad Fili
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Changze Han
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Syed A. Rahman
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Isaiah G. L. Bicar
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Sullivan Gregory
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Annika Helverson
- Department of Biostatistics, College of Public Health, The University of Iowa, Iowa City, Iowa, United States of America
| | - Guiping Hu
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Benjamin W. Darbro
- Department of Pediatrics, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Jishnu Das
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Grant D. Brown
- Department of Biostatistics, College of Public Health, The University of Iowa, Iowa City, Iowa, United States of America
| | - Hillel Haim
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
5
|
Faraji N, Zeinali T, Joukar F, Aleali MS, Eslami N, Shenagari M, Mansour-Ghanaei F. Mutational dynamics of SARS-CoV-2: Impact on future COVID-19 vaccine strategies. Heliyon 2024; 10:e30208. [PMID: 38707429 PMCID: PMC11066641 DOI: 10.1016/j.heliyon.2024.e30208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
The rapid emergence of multiple strains of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has sparked profound concerns regarding the ongoing evolution of the virus and its potential impact on global health. Classified by the World Health Organization (WHO) as variants of concern (VOC), these strains exhibit heightened transmissibility and pathogenicity, posing significant challenges to existing vaccine strategies. Despite widespread vaccination efforts, the continual evolution of SARS-CoV-2 variants presents a formidable obstacle to achieving herd immunity. Of particular concern is the coronavirus spike (S) protein, a pivotal viral surface protein crucial for host cell entry and infectivity. Mutations within the S protein have been shown to enhance transmissibility and confer resistance to antibody-mediated neutralization, undermining the efficacy of traditional vaccine platforms. Moreover, the S protein undergoes rapid molecular evolution under selective immune pressure, leading to the emergence of diverse variants with distinct mutation profiles. This review underscores the urgent need for vigilance and adaptation in vaccine development efforts to combat the evolving landscape of SARS-CoV-2 mutations and ensure the long-term effectiveness of global immunization campaigns.
Collapse
Affiliation(s)
- Niloofar Faraji
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Tahereh Zeinali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Maryam Sadat Aleali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Narges Eslami
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shenagari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
6
|
Jalil S, Keskinen T, Juutila J, Sartori Maldonado R, Euro L, Suomalainen A, Lapatto R, Kuuluvainen E, Hietakangas V, Otonkoski T, Hyvönen ME, Wartiovaara K. Genetic and functional correction of argininosuccinate lyase deficiency using CRISPR adenine base editors. Am J Hum Genet 2024; 111:714-728. [PMID: 38579669 PMCID: PMC11023919 DOI: 10.1016/j.ajhg.2024.03.004] [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: 08/23/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
Argininosuccinate lyase deficiency (ASLD) is a recessive metabolic disorder caused by variants in ASL. In an essential step in urea synthesis, ASL breaks down argininosuccinate (ASA), a pathognomonic ASLD biomarker. The severe disease forms lead to hyperammonemia, neurological injury, and even early death. The current treatments are unsatisfactory, involving a strict low-protein diet, arginine supplementation, nitrogen scavenging, and in some cases, liver transplantation. An unmet need exists for improved, efficient therapies. Here, we show the potential of a lipid nanoparticle-mediated CRISPR approach using adenine base editors (ABEs) for ASLD treatment. To model ASLD, we first generated human-induced pluripotent stem cells (hiPSCs) from biopsies of individuals homozygous for the Finnish founder variant (c.1153C>T [p.Arg385Cys]) and edited this variant using the ABE. We then differentiated the hiPSCs into hepatocyte-like cells that showed a 1,000-fold decrease in ASA levels compared to those of isogenic non-edited cells. Lastly, we tested three different FDA-approved lipid nanoparticle formulations to deliver the ABE-encoding RNA and the sgRNA targeting the ASL variant. This approach efficiently edited the ASL variant in fibroblasts with no apparent cell toxicity and minimal off-target effects. Further, the treatment resulted in a significant decrease in ASA, to levels of healthy donors, indicating restoration of the urea cycle. Our work describes a highly efficient approach to editing the disease-causing ASL variant and restoring the function of the urea cycle. This method relies on RNA delivered by lipid nanoparticles, which is compatible with clinical applications, improves its safety profile, and allows for scalable production.
Collapse
Affiliation(s)
- Sami Jalil
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Keskinen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juhana Juutila
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Rocio Sartori Maldonado
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Liliya Euro
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Lapatto
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Emilia Kuuluvainen
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ville Hietakangas
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mervi E Hyvönen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kirmo Wartiovaara
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinical Genetics, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
7
|
Clever S, Limpinsel L, Meyer zu Natrup C, Schünemann LM, Beythien G, Rosiak M, Hülskötter K, Gregor KM, Tuchel T, Kalodimou G, Freudenstein A, Kumar S, Baumgärtner W, Sutter G, Tscherne A, Volz A. Single MVA-SARS-2-ST/N Vaccination Rapidly Protects K18-hACE2 Mice against a Lethal SARS-CoV-2 Challenge Infection. Viruses 2024; 16:417. [PMID: 38543782 PMCID: PMC10974247 DOI: 10.3390/v16030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
The sudden emergence of SARS-CoV-2 demonstrates the need for new vaccines that rapidly protect in the case of an emergency. In this study, we developed a recombinant MVA vaccine co-expressing SARS-CoV-2 prefusion-stabilized spike protein (ST) and SARS-CoV-2 nucleoprotein (N, MVA-SARS-2-ST/N) as an approach to further improve vaccine-induced immunogenicity and efficacy. Single MVA-SARS-2-ST/N vaccination in K18-hACE2 mice induced robust protection against lethal respiratory SARS-CoV-2 challenge infection 28 days later. The protective outcome of MVA-SARS-2-ST/N vaccination correlated with the activation of SARS-CoV-2-neutralizing antibodies (nABs) and substantial amounts of SARS-CoV-2-specific T cells especially in the lung of MVA-SARS-2-ST/N-vaccinated mice. Emergency vaccination with MVA-SARS-2-ST/N just 2 days before lethal SARS-CoV-2 challenge infection resulted in a delayed onset of clinical disease outcome in these mice and increased titers of nAB or SARS-CoV-2-specific T cells in the spleen and lung. These data highlight the potential of a multivalent COVID-19 vaccine co-expressing S- and N-protein, which further contributes to the development of rapidly protective vaccination strategies against emerging pathogens.
Collapse
Affiliation(s)
- Sabrina Clever
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Leonard Limpinsel
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Christian Meyer zu Natrup
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Lisa-Marie Schünemann
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Malgorzata Rosiak
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Katharina Manuela Gregor
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Tamara Tuchel
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Astrid Freudenstein
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Satendra Kumar
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| |
Collapse
|
8
|
Muñoz-Alía MÁ, Nace RA, Balakrishnan B, Zhang L, Packiriswamy N, Singh G, Warang P, Mena I, Narjari R, Vandergaast R, Peng KW, García-Sastre A, Schotsaert M, Russell SJ. Surface-modified measles vaccines encoding oligomeric, prefusion-stabilized SARS-CoV-2 spike glycoproteins boost neutralizing antibody responses to Omicron and historical variants, independent of measles seropositivity. mBio 2024; 15:e0292823. [PMID: 38193729 PMCID: PMC10865805 DOI: 10.1128/mbio.02928-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Serum titers of SARS-CoV-2-neutralizing antibodies (nAbs) correlate well with protection from symptomatic COVID-19 but decay rapidly in the months following vaccination or infection. In contrast, measles-protective nAb titers are lifelong after measles vaccination, possibly due to persistence of the live-attenuated virus in lymphoid tissues. We, therefore, sought to generate a live recombinant measles vaccine capable of driving high SARS-CoV-2 nAb responses. Since previous clinical testing of a live measles vaccine encoding a SARS-CoV-2 spike glycoprotein resulted in suboptimal anti-spike antibody titers, our new vectors were designed to encode prefusion-stabilized SARS-CoV-2 spike glycoproteins, trimerized via an inserted peptide domain, and displayed on a dodecahedral miniferritin scaffold. Additionally, to circumvent the blunting of vaccine efficacy by preformed anti-measles antibodies, we extensively modified the measles surface glycoproteins. Comprehensive in vivo mouse testing demonstrated the potent induction of high titer nAbs in measles-immune mice and confirmed the significant contributions to overall potency afforded by prefusion stabilization, trimerization, and miniferritin display of the SARS-CoV-2 spike glycoprotein. In animals primed and boosted with a measles virus (MeV) vaccine encoding the ancestral SARS-CoV-2 spike, high-titer nAb responses against ancestral virus strains were only weakly cross-reactive with the Omicron variant. However, in primed animals that were boosted with a MeV vaccine encoding the Omicron BA.1 spike, antibody titers to both ancestral and Omicron strains were robustly elevated, and the passive transfer of serum from these animals protected K18-ACE2 mice from infection and morbidity after exposure to BA.1 and WA1/2020 strains. Our results demonstrate that by engineering the antigen, we can develop potent measles-based vaccine candidates against SARS-CoV-2.IMPORTANCEAlthough the live-attenuated measles virus (MeV) is one of the safest and most efficacious human vaccines, a measles-vectored COVID-19 vaccine candidate expressing the SARS-CoV-2 spike failed to elicit neutralizing antibody (nAb) responses in a phase-1 clinical trial, especially in measles-immune individuals. Here, we constructed a comprehensive panel of MeV-based COVID-19 vaccine candidates using a MeV with extensive modifications on the envelope glycoproteins (MeV-MR). We show that artificial trimerization of the spike is critical for the induction of nAbs and that their magnitude can be significantly augmented when the spike protein is synchronously fused to a dodecahedral scaffold. Furthermore, preexisting measles immunity did not abolish heterologous immunity elicited by our vector. Our results highlight the importance of antigen optimization in the development of spike-based COVID-19 vaccines and therapies.
Collapse
Affiliation(s)
- Miguel Á. Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Vyriad Inc, Rochester, Minnesota, USA
| | - Rebecca A. Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Vyriad Inc, Rochester, Minnesota, USA
- Imanis Life Sciences, Rochester, Minnesota, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Vyriad Inc, Rochester, Minnesota, USA
- Imanis Life Sciences, Rochester, Minnesota, USA
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Wu L, Li X, Qian X, Wang S, Liu J, Yan J. Lipid Nanoparticle (LNP) Delivery Carrier-Assisted Targeted Controlled Release mRNA Vaccines in Tumor Immunity. Vaccines (Basel) 2024; 12:186. [PMID: 38400169 PMCID: PMC10891594 DOI: 10.3390/vaccines12020186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, lipid nanoparticles (LNPs) have attracted extensive attention in tumor immunotherapy. Targeting immune cells in cancer therapy has become a strategy of great research interest. mRNA vaccines are a potential choice for tumor immunotherapy, due to their ability to directly encode antigen proteins and stimulate a strong immune response. However, the mode of delivery and lack of stability of mRNA are key issues limiting its application. LNPs are an excellent mRNA delivery carrier, and their structural stability and biocompatibility make them an effective means for delivering mRNA to specific targets. This study summarizes the research progress in LNP delivery carrier-assisted targeted controlled release mRNA vaccines in tumor immunity. The role of LNPs in improving mRNA stability, immunogenicity, and targeting is discussed. This review aims to systematically summarize the latest research progress in LNP delivery carrier-assisted targeted controlled release mRNA vaccines in tumor immunity to provide new ideas and strategies for tumor immunotherapy, as well as to provide more effective treatment plans for patients.
Collapse
Affiliation(s)
- Liusheng Wu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Xiaoqiang Li
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China;
| | - Xinye Qian
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| | - Shuang Wang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China;
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| |
Collapse
|
10
|
Abdoli A, Jamshidi H, Taqavian M, Baghal ML, Jalili H. Omicron-specific and bivalent omicron-containing vaccine candidates elicit potent virus neutralisation in the animal model. Sci Rep 2024; 14:268. [PMID: 38168473 PMCID: PMC10762194 DOI: 10.1038/s41598-023-50822-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024] Open
Abstract
Omicron variant (B.1.1.529) is able to escape from naturally acquired and vaccine-induced immunity, which mandates updating the current COVID-19 vaccines. Here, we investigated and compared the neutralising antibody induction of the ancestral variant-based BIV1-CovIran vaccine, the Omicron variant-based BIV1-CovIran Plus vaccine, and the novel bivalent vaccine candidate, BBIV1-CovIran, against the Omicron and ancestral Wuhan variants on the rat model. After inactivating the viral particles, the viruses were purified and formulated. Bivalent vaccines were a composition of 2.5 µg (5 µg total) or 5 µg (10 µg total) doses of each ansectral-based and Omicron-based monovalent vaccine. Subsequently, the potency of the monovalent and bivalent vaccines was investigated using the virus neutralisation test (VNT). The group that received three doses of the Omicron-specific vaccine demonstrated neutralisation activity against the Omicron variant with a geometric mean titer of 337.8. However, three doses of the Wuhan variant-specific vaccine could neutralise the Omicron variant at a maximum of 1/32 serum dilution. The neutralisation activity of the Omicron-specific vaccine, when administered as the booster dose after two doses of the Wuhan variant-specific vaccine, was 100% against the Omicron variant and the Wuhan variant at 1/64 and 1/128 serum dilution, respectively. Three doses of 5 µg bivalent vaccine could effectively neutralise both variants at the minimum of 1/128 serum dilution. The 10 µg bivalent vaccine at three doses showed even higher neutralisation titers: the geometric mean of 388 (95% CI 242.2-621.7) against Omicron and 445.7 (95% CI 303.3-655.0) against Wuhan. It is shown that the candidate bivalent and Omicron-specific vaccines could elicit a potent immune response against both Wuhan-Hu-1 and Omicron BA.1 variants.
Collapse
Affiliation(s)
- Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
- Amirabad Virology Laboratory, Vaccine Unit, Tehran, Iran
| | - Hamidreza Jamshidi
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Hasan Jalili
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.
| |
Collapse
|
11
|
Ye Z, Bonam SR, McKay LGA, Plante JA, Walker J, Zhao Y, Huang C, Chen J, Xu C, Li Y, Liu L, Harmon J, Gao S, Song D, Zhang Z, Plante KS, Griffiths A, Chen J, Hu H, Xu Q. Monovalent SARS-COV-2 mRNA vaccine using optimal UTRs and LNPs is highly immunogenic and broadly protective against Omicron variants. Proc Natl Acad Sci U S A 2023; 120:e2311752120. [PMID: 38134199 PMCID: PMC10756290 DOI: 10.1073/pnas.2311752120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
The emergence of highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) that are resistant to the current COVID-19 vaccines highlights the need for continued development of broadly protective vaccines for the future. Here, we developed two messenger RNA (mRNA)-lipid nanoparticle (LNP) vaccines, TU88mCSA and ALCmCSA, using the ancestral SARS-CoV-2 spike sequence, optimized 5' and 3' untranslated regions (UTRs), and LNP combinations. Our data showed that these nanocomplexes effectively activate CD4+ and CD8+ T cell responses and humoral immune response and provide complete protection against WA1/2020, Omicron BA.1 and BQ.1 infection in hamsters. Critically, in Omicron BQ.1 challenge hamster models, TU88mCSA and ALCmCSA not only induced robust control of virus load in the lungs but also enhanced protective efficacy in the upper respiratory airways. Antigen-specific immune analysis in mice revealed that the observed cross-protection is associated with superior UTRs [Carboxylesterase 1d (Ces1d)/adaptor protein-3β (AP3B1)] and LNP formulations that elicit robust lung tissue-resident memory T cells. Strong protective effects of TU88mCSA or ALCmCSA against both WA1/2020 and VOCs suggest that this mRNA-LNP combination can be a broadly protective vaccine platform in which mRNA cargo uses the ancestral antigen sequence regardless of the antigenic drift. This approach could be rapidly adapted for clinical use and timely deployment of vaccines against emerging and reemerging VOCs.
Collapse
Affiliation(s)
- Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX77555
| | - Lindsay G. A. McKay
- National Emerging Infectious Diseases Laboratories and Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA02215
| | - Jessica A. Plante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX77555
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX77555
| | - Jordyn Walker
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX77555
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX77555
| | - Yu Zhao
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Changfeng Huang
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Chutian Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Yamin Li
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY13210
| | - Lihan Liu
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Joseph Harmon
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Shuliang Gao
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Donghui Song
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Zhibo Zhang
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| | - Kenneth S. Plante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX77555
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX77555
| | - Anthony Griffiths
- National Emerging Infectious Diseases Laboratories and Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA02215
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX77555
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA02155
| |
Collapse
|
12
|
Wu Y, Shi J, He X, Lu J, Gao X, Zhu X, Chen X, Zhang M, Fang L, Zhang J, Yuan Z, Xiao G, Zhou P, Pan X. Protection of the receptor binding domain (RBD) dimer against SARS-CoV-2 and its variants. J Virol 2023; 97:e0127923. [PMID: 37843372 PMCID: PMC10688353 DOI: 10.1128/jvi.01279-23] [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: 08/18/2023] [Accepted: 09/16/2023] [Indexed: 10/17/2023] Open
Abstract
IMPORTANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants achieved immune escape and became less virulent and easily transmissible through rapid mutation in the spike protein, thus the efficacy of vaccines on the market or in development continues to be challenged. Updating the vaccine, exploring compromise vaccination strategies, and evaluating the efficacy of candidate vaccines for the emerging variants in a timely manner are important to combat complex and volatile SARS-CoV-2. This study reports that vaccines prepared from the dimeric receptor-binding domain (RBD) recombinant protein, which can be quickly produced using a mature and stable process platform, had both good immunogenicity and protection in vivo and could completely protect rodents from lethal challenge by SARS-CoV-2 and its variants, including the emerging Omicron XBB.1.16, highlighting the value of dimeric recombinant vaccines in the post-COVID-19 era.
Collapse
Affiliation(s)
- Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jian Shi
- Wuhan YZY Biopharma Co., Ltd., Wuhan, China
| | - Xiaoxue He
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jia Lu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xiao Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xuerui Zhu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xinlan Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Man Zhang
- Wuhan YZY Biopharma Co., Ltd., Wuhan, China
| | | | - Jing Zhang
- Wuhan YZY Biopharma Co., Ltd., Wuhan, China
| | - Zhiming Yuan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | | | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
13
|
Hayashi H, Sun J, Yanagida Y, Otera T, Tai JA, Nishikawa T, Yamashita K, Sakaguchi N, Yoshida S, Baba S, Chang CY, Shimamura M, Okamoto S, Amaishi Y, Chono H, Mineno J, Rakugi H, Morishita R, Nakagami H. Intradermal administration of DNA vaccine targeting Omicron SARS-CoV-2 via pyro-drive jet injector provides the prolonged neutralizing antibody production via germinal center reaction. Sci Rep 2023; 13:13033. [PMID: 37563266 PMCID: PMC10415318 DOI: 10.1038/s41598-023-40172-y] [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: 03/08/2023] [Accepted: 08/06/2023] [Indexed: 08/12/2023] Open
Abstract
Emerging SARS-CoV-2 Omicron variants are highly contagious with enhanced immune escape mechanisms against the initially approved COVID-19 vaccines. Therefore, we require stable alternative-platform vaccines that confer protection against newer variants of SARS-CoV-2. We designed an Omicron B.1.1.529 specific DNA vaccine using our DNA vaccine platform and evaluated the humoral and cellular immune responses. SD rats intradermally administered with Omicron-specific DNA vaccine via pyro-drive jet injector (PJI) thrice at 2-week intervals elicited high antibody titers against the Omicron subvariants as well as the ancestral strain. Indeed, the Omicron B.1.1.529-specific antibody titer and neutralizing antibody were higher than that of other strains. Longitudinal monitoring indicated that anti-spike (ancestral and Omicron) antibody titers decreased toward 30 weeks after the first vaccination dose. However, neutralization activity remained unaltered. Germinal center formation was histologically detected in lymph nodes in rats immunized with Omicron DNA vaccine. Ancestral spike-specific immune cell response was slightly weaker than Omicron spike-specific response in splenocytes with Omicron-adapted DNA vaccine, evaluated by ELISpot assay. Collectively, our findings suggest that Omicron targeting DNA vaccines via PJI can elicit robust durable antibody production mediated by germinal center reaction against this new variant as well as partially against the spike protein of other SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Hiroki Hayashi
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Jiao Sun
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuka Yanagida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takako Otera
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Anges Inc., Tokyo, Japan
| | - Jiayu A Tai
- Department of Device Application for Molecular Therapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomoyuki Nishikawa
- Department of Device Application for Molecular Therapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kunihiko Yamashita
- Department of Device Application for Molecular Therapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Daicel Co, Osaka, Japan
| | | | - Shota Yoshida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Satoshi Baba
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Chin Yang Chang
- Department of Gene and Stem Cell Regenerative Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Munehisa Shimamura
- Department of Gene and Stem Cell Regenerative Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | - Hiromi Rakugi
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
14
|
Narita K, Ikeda D, Seki M, Fukumoto A, Tabata R, Uesugi Y, Miura D, Takeuchi M, Doi M, Umezawa Y, Otsuka Y, Matsue K. Prevalence and Clinical Outcome of Omicron Breakthrough Infection in Patients With Hematologic Disease: A Prospective Observational Cohort Study. Hemasphere 2023; 7:e905. [PMID: 37292116 PMCID: PMC10247214 DOI: 10.1097/hs9.0000000000000905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Affiliation(s)
- Kentaro Narita
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| | - Daisuke Ikeda
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| | - Mizuki Seki
- Postgraduate Education Center, Kameda Medical Center, Kamogawa-shi, Japan
| | - Ami Fukumoto
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| | - Rikako Tabata
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| | - Yuka Uesugi
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| | - Daisuke Miura
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| | - Masami Takeuchi
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| | - Masahiro Doi
- Central laboratory, Kameda Medical Center, Kamogawa-shi, Japan
| | - Yuka Umezawa
- Central laboratory, Kameda Medical Center, Kamogawa-shi, Japan
| | | | - Kosei Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa-shi, Japan
| |
Collapse
|
15
|
Ma Q, Li M, Ma L, Zhang C, Zhang H, Zhong H, Wen J, Wang Y, Yan Z, Xiong W, Wu L, Guo J, Yang W, Yang Z, Zhang B. SARS-CoV-2 bivalent mRNA vaccine with broad protection against variants of concern. Front Immunol 2023; 14:1195299. [PMID: 37292197 PMCID: PMC10244545 DOI: 10.3389/fimmu.2023.1195299] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/04/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant has rapidly spread around the globe. With a substantial number of mutations in its Spike protein, the SARS-CoV-2 Omicron variant is prone to immune evasion and led to the reduced efficacy of approved vaccines. Thus, emerging variants have brought new challenges to the prevention of COVID-19 and updated vaccines are urgently needed to provide better protection against the Omicron variant or other highly mutated variants. Materials and methods Here, we developed a novel bivalent mRNA vaccine, RBMRNA-405, comprising a 1:1 mix of mRNAs encoding both Delta-derived and Omicron-derived Spike proteins. We evaluated the immunogenicity of RBMRNA-405 in BALB/c mice and compared the antibody response and prophylactic efficacy induced by monovalent Delta or Omicron-specific vaccine with the bivalent RBMRNA-405 vaccine in the SARSCoV-2 variant challenge. Results Results showed that the RBMRNA-405 vaccine could generate broader neutralizing antibody responses against both Wuhan-Hu-1 and other SARS-CoV-2 variants, including Delta, Omicron, Alpha, Beta, and Gamma. RBMRNA-405 efficiently blocked infectious viral replication and lung injury in both Omicron- and Delta-challenged K18-ACE2 mice. Conclusion Our data suggest that RBMRNA-405 is a promising bivalent SARS-CoV-2 vaccine with broad-spectrum efficacy for further clinical development.
Collapse
Affiliation(s)
- Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Man Li
- Department of Drug Discovery and Development, Argorna Pharmaceuticals Co., Ltd, Guangzhou, China
| | - Lin Ma
- Department of Manufacturing, Guangzhou RiboBio Co., Ltd, Guangzhou, China
| | - Caroline Zhang
- Department of Manufacturing, Guangzhou RiboBio Co., Ltd, Guangzhou, China
| | - Hong Zhang
- Department of Drug Discovery and Development, Argorna Pharmaceuticals Co., Ltd, Guangzhou, China
| | - Huiling Zhong
- Department of Drug Discovery and Development, Argorna Pharmaceuticals Co., Ltd, Guangzhou, China
| | - Jian Wen
- Department of Manufacturing, Guangzhou RiboBio Co., Ltd, Guangzhou, China
| | - Yongsheng Wang
- Department of Drug Discovery and Development, Argorna Pharmaceuticals Co., Ltd, Guangzhou, China
| | - Zewei Yan
- Department of Drug Discovery and Development, Argorna Pharmaceuticals Co., Ltd, Guangzhou, China
| | - Wei Xiong
- Department of Manufacturing, Guangzhou RiboBio Co., Ltd, Guangzhou, China
| | - Linping Wu
- Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jianmin Guo
- Guangdong Provincial Key Laboratory of Drug Non-clinical Evaluation and Research, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangzhou, China
| | - Wei Yang
- Guangdong Provincial Key Laboratory of Drug Non-clinical Evaluation and Research, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangzhou, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Biliang Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Drug Discovery and Development, Argorna Pharmaceuticals Co., Ltd, Guangzhou, China
- Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
16
|
Li X, Liu J, Li W, Peng Q, Li M, Ying Z, Zhang Z, Liu X, Wu X, Zhao D, Yang L, Cao S, Huang Y, Shi L, Xu H, Wang Y, Yue G, Suo Y, Nie J, Huang W, Li J, Li Y. Heterologous prime-boost immunisation with mRNA- and AdC68-based 2019-nCoV variant vaccines induces broad-spectrum immune responses in mice. Front Immunol 2023; 14:1142394. [PMID: 37006275 PMCID: PMC10050358 DOI: 10.3389/fimmu.2023.1142394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
The ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 or 2019-nCoV) variants has been associated with the transmission and pathogenicity of COVID-19. Therefore, exploring the optimal immunisation strategy to improve the broad-spectrum cross-protection ability of COVID-19 vaccines is of great significance. Herein, we assessed different heterologous prime-boost strategies with chimpanzee adenovirus vector-based COVID-19 vaccines plus Wuhan-Hu-1 (WH-1) strain (AdW) and Beta variant (AdB) and mRNA-based COVID-19 vaccines plus WH-1 strain (ARW) and Omicron (B.1.1.529) variant (ARO) in 6-week-old female BALB/c mice. AdW and AdB were administered intramuscularly or intranasally, while ARW and ARO were administered intramuscularly. Intranasal or intramuscular vaccination with AdB followed by ARO booster exhibited the highest levels of cross-reactive IgG, pseudovirus-neutralising antibody (PNAb) responses, and angiotensin-converting enzyme-2 (ACE2)-binding inhibition rates against different 2019-nCoV variants among all vaccination groups. Moreover, intranasal AdB vaccination followed by ARO induced higher levels of IgA and neutralising antibody responses against live 2019-nCoV than intramuscular AdB vaccination followed by ARO. A single dose of AdB administered intranasally or intramuscularly induced broader cross-NAb responses than AdW. Th1-biased cellular immune response was induced in all vaccination groups. Intramuscular vaccination-only groups exhibited higher levels of Th1 cytokines than intranasal vaccination-only and intranasal vaccination-containing groups. However, no obvious differences were found in the levels of Th2 cytokines between the control and all vaccination groups. Our findings provide a basis for exploring vaccination strategies against different 2019-nCoV variants to achieve high broad-spectrum immune efficacy.
Collapse
Affiliation(s)
- Xingxing Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Jingjing Liu
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenjuan Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Qinhua Peng
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Miao Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Zhifang Ying
- Department of Respiratory Virus Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Zelun Zhang
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Xinyu Liu
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Xiaohong Wu
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Danhua Zhao
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Lihong Yang
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Shouchun Cao
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yanqiu Huang
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Leitai Shi
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Hongshan Xu
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yunpeng Wang
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Guangzhi Yue
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yue Suo
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Jianhui Nie
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Weijin Huang
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Jia Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yuhua Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
17
|
Fang Z, Monteiro VS, Renauer PA, Shang X, Suzuki K, Ling X, Bai M, Xiang Y, Levchenko A, Booth CJ, Lucas C, Chen S. Polyvalent mRNA vaccination elicited potent immune response to monkeypox virus surface antigens. Cell Res 2023; 33:407-410. [PMID: 36879038 PMCID: PMC9988199 DOI: 10.1038/s41422-023-00792-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Affiliation(s)
- Zhenhao Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | | | - Paul A Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Xingbo Shang
- System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Kazushi Suzuki
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Xinyu Ling
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Meizhu Bai
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Yan Xiang
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Andre Levchenko
- System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Carmen J Booth
- Department of Comparative Medicine, Yale University, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale University, New Haven, CT, USA. .,Center for Infection and Immunity, Yale University, New Haven, CT, USA.
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA. .,System Biology Institute, Yale University, West Haven, CT, USA. .,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA. .,Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA. .,Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA. .,Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA. .,Wu-Tsai Institute, Yale University, New Haven, CT, USA. .,Center for RNA Science and Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
Carabelli AM, Peacock TP, Thorne LG, Harvey WT, Hughes J, Peacock SJ, Barclay WS, de Silva TI, Towers GJ, Robertson DL. SARS-CoV-2 variant biology: immune escape, transmission and fitness. Nat Rev Microbiol 2023; 21:162-177. [PMID: 36653446 PMCID: PMC9847462 DOI: 10.1038/s41579-022-00841-7] [Citation(s) in RCA: 298] [Impact Index Per Article: 298.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 01/19/2023]
Abstract
In late 2020, after circulating for almost a year in the human population, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exhibited a major step change in its adaptation to humans. These highly mutated forms of SARS-CoV-2 had enhanced rates of transmission relative to previous variants and were termed 'variants of concern' (VOCs). Designated Alpha, Beta, Gamma, Delta and Omicron, the VOCs emerged independently from one another, and in turn each rapidly became dominant, regionally or globally, outcompeting previous variants. The success of each VOC relative to the previously dominant variant was enabled by altered intrinsic functional properties of the virus and, to various degrees, changes to virus antigenicity conferring the ability to evade a primed immune response. The increased virus fitness associated with VOCs is the result of a complex interplay of virus biology in the context of changing human immunity due to both vaccination and prior infection. In this Review, we summarize the literature on the relative transmissibility and antigenicity of SARS-CoV-2 variants, the role of mutations at the furin spike cleavage site and of non-spike proteins, the potential importance of recombination to virus success, and SARS-CoV-2 evolution in the context of T cells, innate immunity and population immunity. SARS-CoV-2 shows a complicated relationship among virus antigenicity, transmission and virulence, which has unpredictable implications for the future trajectory and disease burden of COVID-19.
Collapse
Affiliation(s)
| | - Thomas P Peacock
- Department of Infectious Disease, St Mary's Medical School, Imperial College London, London, UK
| | - Lucy G Thorne
- Division of Infection and Immunity, University College London, London, UK
| | - William T Harvey
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
- Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Sharon J Peacock
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
| | - Wendy S Barclay
- Department of Infectious Disease, St Mary's Medical School, Imperial College London, London, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
| | - Greg J Towers
- Division of Infection and Immunity, University College London, London, UK
| | - David L Robertson
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK.
| |
Collapse
|
19
|
Zhao G, Zhang Z, Ding Y, Hou J, Liu Y, Zhang M, Sui C, Wang L, Xu X, Gao X, Kou Z. A DNA Vaccine Encoding the Full-Length Spike Protein of Beta Variant (B.1.351) Elicited Broader Cross-Reactive Immune Responses against Other SARS-CoV-2 Variants. Vaccines (Basel) 2023; 11:513. [PMID: 36992097 PMCID: PMC10054764 DOI: 10.3390/vaccines11030513] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The SARS-CoV-2 pandemic remains an ongoing threat to global health with emerging variants, especially the Omicron variant and its sub-lineages. Although large-scale vaccination worldwide has delivered outstanding achievements for COVID-19 prevention, a declining effectiveness to a different extent in emerging SARS-CoV-2 variants was observed in the vaccinated population. Vaccines eliciting broader spectrum neutralizing antibodies and cellular immune responses are urgently needed and important. To achieve this goal, rational vaccine design, including antigen modeling, screening and combination, vaccine pipelines, and delivery, are keys to developing a next-generation COVID-19 vaccine. In this study, we designed several DNA constructs based on codon-optimized spike coding regions of several SARS-CoV-2 variants and analyzed their cross-reactive antibodies, including neutralizing antibodies, and cellular immune responses against several VOCs in C57BL/6 mice. The results revealed that different SARS-CoV-2 VOCs induced different cross-reactivity; pBeta, a DNA vaccine encoding the spike protein of the Beta variant, elicited broader cross-reactive neutralizing antibodies against other variants including the Omicron variants BA.1 and BA.4/5. This result demonstrates that the spike antigen from the Beta variant potentially serves as one of the antigens for multivalent vaccine design and development against variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Gan Zhao
- Advaccine Biopharmaceutics (Suzhou) Co., Ltd., Suzhou 215000, China
| | | | | | | | | | | | | | | | | | | | - Zhihua Kou
- Advaccine Biopharmaceutics (Suzhou) Co., Ltd., Suzhou 215000, China
| |
Collapse
|
20
|
Ren P, Peng L, Yang L, Suzuki K, Fang Z, Renauer PA, Lin Q, Bai M, Li T, Clark P, Klein D, Chen S. RAMIHM generates fully human monoclonal antibodies by rapid mRNA immunization of humanized mice and BCR-seq. Cell Chem Biol 2023; 30:85-96.e6. [PMID: 36640761 PMCID: PMC9868106 DOI: 10.1016/j.chembiol.2022.12.005] [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: 03/04/2022] [Revised: 10/17/2022] [Accepted: 12/19/2022] [Indexed: 01/15/2023]
Abstract
As a clinical vaccine, lipid nanoparticle (LNP) mRNA has demonstrated potent and broad antibody responses, leading to speculation about its potential for antibody discovery. Here, we developed RAMIHM, a highly efficient strategy for developing fully human monoclonal antibodies that employs rapid mRNA immunization of humanized mice followed by single B cell sequencing (scBCR-seq). We immunized humanized transgenic mice with RAMIHM and generated 15 top-ranked clones from peripheral blood, plasma B, and memory B cell populations, demonstrating a high rate of antigen-specificity (93.3%). Two Omicron-specific neutralizing antibodies with high potency and one broad-spectrum neutralizing antibody were discovered. Furthermore, we extended the application of RAMIHM to cancer immunotherapy targets, including a single transmembrane protein CD22 and a multi-transmembrane G protein-coupled receptor target, GPRC5D, which is difficult for traditional protein immunization methods. RAMIHM-scBCR-seq is a broadly applicable platform for the rapid and efficient development of fully human monoclonal antibodies against an assortment of targets.
Collapse
Affiliation(s)
- Ping Ren
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Kazushi Suzuki
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Zhenhao Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Paul A Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA; Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT 06520, USA
| | - Qianqian Lin
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Meizhu Bai
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Tongqing Li
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University, New Haven, CT 06520, USA
| | - Paul Clark
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA
| | - Daryl Klein
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University, New Haven, CT 06520, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; System Biology Institute, Yale University, West Haven, CT 06520, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06520, USA; Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT 06520, USA; Immunobiology Program, Yale University, New Haven, CT 06520, USA; Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA; Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA; Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
21
|
Muñoz-Alía MÁ, Nace RA, Balakrishnan B, Zhang L, Packiriswamy N, Singh G, Warang P, Mena I, Narjari R, Vandergaast R, García-Sastre A, Schotsaert M, Russell SJ. Surface-modified measles vaccines encoding oligomeric, fusion-stabilized SARS-CoV-2 spike glycoproteins bypass measles seropositivity, boosting neutralizing antibody responses to omicron and historical variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.12.16.520799. [PMID: 36561187 PMCID: PMC9774211 DOI: 10.1101/2022.12.16.520799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Serum titers of SARS-CoV-2 neutralizing antibodies (nAb) correlate well with protection from symptomatic COVID-19, but decay rapidly in the months following vaccination or infection. In contrast, measles-protective nAb titers are life-long after measles vaccination, possibly due to persistence of the live-attenuated virus in lymphoid tissues. We therefore sought to generate a live recombinant measles vaccine capable of driving high SARS-CoV-2 nAb responses. Since previous clinical testing of a live measles vaccine encoding a SARS-CoV-2 spike glycoprotein resulted in suboptimal anti-spike antibody titers, our new vectors were designed to encode prefusion-stabilized SARS-CoV-2 spike glycoproteins, trimerized via an inserted peptide domain and displayed on a dodecahedral miniferritin scaffold. Additionally, to circumvent the blunting of vaccine efficacy by preformed anti-measles antibodies, we extensively modified the measles surface glycoproteins. Comprehensive in vivo mouse testing demonstrated potent induction of high titer nAb in measles-immune mice and confirmed the significant incremental contributions to overall potency afforded by prefusion stabilization, trimerization, and miniferritin-display of the SARS-CoV-2 spike glycoprotein, and vaccine resurfacing. In animals primed and boosted with a MeV vaccine encoding the ancestral SARS-CoV-2 spike, high titer nAb responses against ancestral virus strains were only weakly cross-reactive with the omicron variant. However, in primed animals that were boosted with a MeV vaccine encoding the omicron BA.1 spike, antibody titers to both ancestral and omicron strains were robustly elevated and the passive transfer of serum from these animals protected K18-ACE2 mice from infection and morbidity after exposure to BA.1 and WA1/2020 strains. Our results demonstrate that antigen engineering can enable the development of potent measles-based SARS-CoV-2 vaccine candidates.
Collapse
Affiliation(s)
- Miguel Á. Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
- Vyriad Inc, Rochester, MN, USA
| | - Rebecca A. Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
- Vyriad Inc, Rochester, MN, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Imanis Life Sciences, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
22
|
Peng P, Feng C, Hu J, He C, Deng H, Fan Q, Xiang J, Tang G, Jiang ML, Hu F, Li F, Wang K, Tang N, Tang XP, Huang A. Extensive neutralization against SARS-CoV-2 variants elicited by Omicron-specific subunit vaccine as a heterologous booster. iScience 2022; 25:105465. [PMID: 36338432 PMCID: PMC9621588 DOI: 10.1016/j.isci.2022.105465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/09/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
To overcome the increased risk of SARS-CoV-2 reinfection or post-vaccination infection caused by the Omicron variant, Omicron-specific vaccines were considered a potential strategy. We reported the increased magnitude and breadth of antibody response against VOCs elicited by post-vaccination Delta and Omicron infection, compared to WT infection without vaccination. Then, in mouse models, three doses of Omicron-RBD immunization elicited comparable neutralizing antibody (NAb) titers with three doses of WT-RBD immunization, but the neutralizing activity was not cross-active. By contrast, a heterologous Omicron-RBD booster following two doses of WT-RBD immunization increased the NAb titers against Omicron by 9-folds than the homologous WT-RBD booster. Moreover, it retains neutralization against both WT and current VOCs. Results suggest that Omicron-specific subunit booster shows its advantages in the immune protection from both WT and current VOCs and that SARS-CoV-2 vaccines including two or more virus lineages might improve the NAb response.
Collapse
Affiliation(s)
- Pai Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
| | - Chengqian Feng
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jie Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
| | - Changlong He
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
- Department of Laboratory Medicine, People’s Hospital of Jiulongpo District, Chongqing, China
| | - Haijun Deng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
| | - Qinghong Fan
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jin Xiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
| | - Guofang Tang
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Meng-ling Jiang
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fengyu Hu
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Feng Li
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Kai Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
| | - Xiao-ping Tang
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Yixue Yuan Road No.1, Chongqing 400016, China
| |
Collapse
|
23
|
Khong KW, Zhang R, Hung IFN. The Four Ws of the Fourth Dose COVID-19 Vaccines: Why, Who, When and What. Vaccines (Basel) 2022; 10:1924. [PMID: 36423020 PMCID: PMC9694140 DOI: 10.3390/vaccines10111924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 10/13/2023] Open
Abstract
With the emergence of SARS-CoV-2 variants, vaccine breakthrough is a major public health concern. With evidence of reduced neutralizing antibody activity against Omicron variants and fading antibody level after the third-dose booster vaccine, there are suggestions of a fourth-dose booster vaccine. In this review, the benefits of a fourth-dose booster is evaluated from four perspectives, including the effectiveness of the booster dose against virus variants (Why), susceptible groups of individuals who may benefit from additional booster dose (Who), selection of vaccine platforms to better enhance immunity (What) and appropriate intervals between the third and fourth booster dose (When). In summary, a fourth dose can temporarily boost the immune response against SARS-CoV-2 variants and can be considered for specific groups of individuals. A heterologous vaccine strategy using mRNA vaccine in individuals primed with inactivated vaccine may boost immunity against variants. The timing of the fourth dose should be individualized but an interval of 4 months after the third-dose booster is appropriate. A universal fourth booster dose is not necessary.
Collapse
Affiliation(s)
- Ka-Wa Khong
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ruiqi Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- State Key Laboratory for Emerging Infectious Disease, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| |
Collapse
|
24
|
Focosi D, Maggi F. Do We Really Need Omicron Spike-Based Updated COVID-19 Vaccines? Evidence and Pipeline. Viruses 2022; 14:2488. [PMID: 36366586 PMCID: PMC9692555 DOI: 10.3390/v14112488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
The wild-type SARS-CoV-2 Spike-based vaccines authorized so far have reduced COVID-19 severity, but periodic boosts are required to counteract the decline in immunity. An accelerated rate of immune escape to vaccine-elicited immunity has been associated with Spike protein antigenic shifts, as seen in the Omicron variant of concern and its sublineages, demanding the development of Omicron Spike-based vaccines. Herein, we review the evidence in animal models and topline results from ongoing clinical trials with such updated vaccines, discussing the pros and cons for their deployment.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| | - Fabrizio Maggi
- National Institute for Infectious Diseases “L. Spallanzani”, 00161 Rome, Italy
| |
Collapse
|
25
|
Han Y, An Y, Chen Q, Xu K, Liu X, Xu S, Duan H, Vogel AB, Şahin U, Wang Q, Dai L, Gao GF. mRNA vaccines expressing homo-prototype/Omicron and hetero-chimeric RBD-dimers against SARS-CoV-2. Cell Res 2022; 32:1022-1025. [PMID: 36104506 PMCID: PMC9472730 DOI: 10.1038/s41422-022-00720-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/24/2022] [Indexed: 01/31/2023] Open
Affiliation(s)
- Yuxuan Han
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yaling An
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qian Chen
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Xueyuan Liu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Senyu Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Huixin Duan
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Qihui Wang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Lianpan Dai
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China.
| | - George F Gao
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
26
|
Fang Z, Monteiro VS, Hahn AM, Grubaugh ND, Lucas C, Chen S. Bivalent mRNA vaccine booster induces robust antibody immunity against Omicron lineages BA.2, BA.2.12.1, BA.2.75 and BA.5. Cell Discov 2022; 8:108. [PMID: 36220819 PMCID: PMC9552143 DOI: 10.1038/s41421-022-00473-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/17/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Zhenhao Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,System Biology Institute, Yale University, West Haven, CT, USA.,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | | | - Anne M Hahn
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.,Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA. .,System Biology Institute, Yale University, West Haven, CT, USA. .,Center for Cancer Systems Biology, Yale University, West Haven, CT, USA. .,Immunobiology Program, Yale University, New Haven, CT, USA. .,Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA. .,MD-PhD Program, Yale University, New Haven, CT, USA. .,Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA. .,Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA. .,Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
27
|
Peng L, Fang Z, Renauer PA, McNamara A, Park JJ, Lin Q, Zhou X, Dong MB, Zhu B, Zhao H, Wilen CB, Chen S. Multiplexed LNP-mRNA vaccination against pathogenic coronavirus species. Cell Rep 2022; 40:111160. [PMID: 35921835 PMCID: PMC9294034 DOI: 10.1016/j.celrep.2022.111160] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/07/2022] [Accepted: 07/13/2022] [Indexed: 12/02/2022] Open
Abstract
Although COVID-19 vaccines have been developed, multiple pathogenic coronavirus species exist, urging on development of multispecies coronavirus vaccines. Here we develop prototype lipid nanoparticle (LNP)-mRNA vaccine candidates against SARS-CoV-2 Delta, SARS-CoV, and MERS-CoV, and we test how multiplexing LNP-mRNAs can induce effective immune responses in animal models. Triplex and duplex LNP-mRNA vaccinations induce antigen-specific antibody responses against SARS-CoV-2, SARS-CoV, and MERS-CoV. Single-cell RNA sequencing profiles the global systemic immune repertoires and respective transcriptome signatures of vaccinated animals, revealing a systemic increase in activated B cells and differential gene expression across major adaptive immune cells. Sequential vaccination shows potent antibody responses against all three species, significantly stronger than simultaneous vaccination in mixture. These data demonstrate the feasibility, antibody responses, and single-cell immune profiles of multispecies coronavirus vaccination. The direct comparison between simultaneous and sequential vaccination offers insights into optimization of vaccination schedules to provide broad and potent antibody immunity against three major pathogenic coronavirus species.
Collapse
Affiliation(s)
- Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Zhenhao Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Paul A Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; Molecular Cell Biology, Genetics and Development Program, Yale University, New Haven, CT 06516, USA
| | - Andrew McNamara
- Department of Immunobiology, Yale University, New Haven, CT 06510, USA; Department of Laboratory Medicine, Yale University, New Haven, CT 06510, USA
| | - Jonathan J Park
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; M.D.-Ph.D. Program, Yale University, West Haven, CT 06516, USA
| | - Qianqian Lin
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Xiaoyu Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA
| | - Matthew B Dong
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; Department of Immunobiology, Yale University, New Haven, CT 06510, USA; M.D.-Ph.D. Program, Yale University, West Haven, CT 06516, USA; Immunobiology Program, Yale University, New Haven, CT 06510, USA
| | - Biqing Zhu
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA
| | - Hongyu Zhao
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA; Department of Biostatistics, Yale University School of Public Health, New Haven, CT 06510, USA; Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Craig B Wilen
- Department of Immunobiology, Yale University, New Haven, CT 06510, USA; Department of Laboratory Medicine, Yale University, New Haven, CT 06510, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; System Biology Institute, Yale University, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT 06516, USA; Molecular Cell Biology, Genetics and Development Program, Yale University, New Haven, CT 06516, USA; M.D.-Ph.D. Program, Yale University, West Haven, CT 06516, USA; Immunobiology Program, Yale University, New Haven, CT 06510, USA; Computational Biology and Bioinformatics Program, Yale University, New Haven, CT 06510, USA; Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
28
|
Rauf A, Abu-Izneid T, Khalil AA, Hafeez N, Olatunde A, Rahman M, Semwal P, Al-Awthan YS, Bahattab OS, Khan IN, Khan MA, Sharma R. Nanoparticles in clinical trials of COVID-19: An update. Int J Surg 2022; 104:106818. [PMID: 35953020 PMCID: PMC9359769 DOI: 10.1016/j.ijsu.2022.106818] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 11/01/2022]
Abstract
Once the World Health Organization (WHO) declared the COVID-19 (Coronavirus Infectious Disease-19) outbreak to be pandemic, massive efforts have been launched by researchers around the globe to combat this emerging infectious disease. Strategies that must be investigated such as expanding testing capabilities, developing effective medicines, as well as developing safe and effective vaccines for COVID-19 disease that produce long-lasting immunity to human system. Now-a-days, bio-sensing, medication delivery, imaging, and antimicrobial treatment are just a few of the medical applications for nanoparticles (NPs). Since the early 1990s, nanoparticle drug delivery methods have been employed in clinical trials. Since then, the discipline of nanomedicine has evolved in tandem with expanding technological demands to better medicinal delivery. Newer generations of NPs have emerged in recent decades that are capable of performing additional delivery tasks, allowing for therapy via novel therapeutic modalities. Many of these next generation NPs and associated products have entered clinical trials and have been approved for diverse indications in the present clinical environment. For systemic applications, NPs or nanomedicine-based drug delivery systems have substantial benefits over their non-formulated and free drug counterparts. Nanoparticle systems, for example, are capable of delivering medicines and treating parts of the body that are inaccessible to existing delivery systems. As a result, NPs medication delivery is one of the most studied preclinical and clinical systems. NPs-based vaccines delivering SARS-CoV-2 antigens will play an increasingly important role in prolonging or improving COVID-19 vaccination outcomes. This review provides insights about employing NPs-based drug delivery systems for the treatment of COVID-19 to increase the bioavailability of current drugs, reducing their toxicity, and to increase their efficiency. This article also exhibits their capability and efficacy, and highlighting the future aspects and challenges on nanoparticle products in clinical trials of COVID-19.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, 23430, Khyber Pakhtunkhwa (KP), Pakistan.
| | - Tareq Abu-Izneid
- Pharmaceutical Sciences Department, College of Pharmacy, Al Ain University for Science and Technology, Al Ain, United Arab Emirates
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, 54000, Pakistan
| | - Nabia Hafeez
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar-KPK, 25120, KPK, Pakistan
| | - Ahmed Olatunde
- Department of Medical Biochemistry, Abubakar Tafawa Balewa University, Bauchi, 740272, Nigeria
| | - Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Prabhakar Semwal
- Department of Life Sciences, Graphic Era Deemed to be University, Dehradun, 248002, Uttarakhand, India
| | | | - Omar Salem Bahattab
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Ishaq N Khan
- Institute of Basic Medical Sciences Khyber Medical University, Peshawar, 25100, Pakistan
| | - Muhammad Arslan Khan
- Department of Pharmacy, Faculty of Pharmacy, The University of Lahore, 54000, Pakistan
| | - Rohit Sharma
- Department of Rasa Shastra &Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
29
|
Fang Z, Peng L, Lucas C, Lin Q, Zhou L, Yang L, Feng Y, Ren P, Renauer PA, Monteiro VS, Hahn AM, Park JJ, Zhou X, Grubaugh ND, Wilen CB, Chen S. Heterotypic vaccination responses against SARS-CoV-2 Omicron BA.2. Cell Discov 2022; 8:69. [PMID: 35853867 PMCID: PMC9295082 DOI: 10.1038/s41421-022-00435-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/09/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Zhenhao Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Qianqian Lin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Liqun Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Yanzhi Feng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
| | - Ping Ren
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Paul A Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Valter S Monteiro
- Department of Immunobiology, Yale University, New Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
| | - Anne M Hahn
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Jonathan J Park
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- MD-PhD Program, Yale University, New Haven, CT, USA
| | - Xiaoyu Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Craig B Wilen
- Department of Immunobiology, Yale University, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University, New Haven, CT, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA.
- Immunobiology Program, Yale University, New Haven, CT, USA.
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA.
- MD-PhD Program, Yale University, New Haven, CT, USA.
- Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
- Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
30
|
da Silva ES, Kohnen M, Gilson G, Staub T, Arendt V, Hilger C, Servais JY, Charpentier E, Domingues O, Snoeck CJ, Ollert M, Seguin-Devaux C, Perez-Bercoff D. Pre-Omicron Vaccine Breakthrough Infection Induces Superior Cross-Neutralization against SARS-CoV-2 Omicron BA.1 Compared to Infection Alone. Int J Mol Sci 2022; 23:7675. [PMID: 35887023 PMCID: PMC9320437 DOI: 10.3390/ijms23147675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 02/05/2023] Open
Abstract
SARS-CoV-2 variants raise concern because of their high transmissibility and their ability to evade neutralizing antibodies elicited by prior infection or by vaccination. Here, we compared the neutralizing abilities of sera from 70 unvaccinated COVID-19 patients infected before the emergence of variants of concern (VOCs) and of 16 vaccine breakthrough infection (BTI) cases infected with Gamma or Delta against the ancestral B.1 strain, the Gamma, Delta and Omicron BA.1 VOCs using live virus. We further determined antibody levels against the Nucleocapsid (N) and full Spike proteins, the receptor-binding domain (RBD) and the N-terminal domain (NTD) of the Spike protein. Convalescent sera featured considerable variability in the neutralization of B.1 and in the cross-neutralization of different strains. Their neutralizing capacity moderately correlated with antibody levels against the Spike protein and the RBD. All but one convalescent serum failed to neutralize Omicron BA.1. Overall, convalescent sera from patients with moderate disease had higher antibody levels and displayed a higher neutralizing ability against all strains than patients with mild or severe forms of the disease. The sera from BTI cases fell into one of two categories: half the sera had a high neutralizing activity against the ancestral B.1 strain as well as against the infecting strain, while the other half had no or a very low neutralizing activity against all strains. Although antibody levels against the spike protein and the RBD were lower in BTI sera than in unvaccinated convalescent sera, most neutralizing sera also retained partial neutralizing activity against Omicron BA.1, suggestive of a better cross-neutralization and higher affinity of vaccine-elicited antibodies over virus-induced antibodies. Accordingly, the IC50: antibody level ratios were comparable for BTI and convalescent sera, but remained lower in the neutralizing convalescent sera from patients with moderate disease than in BTI sera. The neutralizing activity of BTI sera was strongly correlated with antibodies against the Spike protein and the RBD. Together, these findings highlight qualitative differences in antibody responses elicited by infection in vaccinated and unvaccinated individuals. They further indicate that breakthrough infection with a pre-Omicron variant boosts immunity and induces cross-neutralizing antibodies against different strains, including Omicron BA.1.
Collapse
Affiliation(s)
- Eveline Santos da Silva
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| | - Michel Kohnen
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Georges Gilson
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Therese Staub
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Victor Arendt
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Christiane Hilger
- Molecular and Translational Allergology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg;
| | - Jean-Yves Servais
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| | - Emilie Charpentier
- Clinical and Applied Virology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.C.); (C.J.S.)
| | - Olivia Domingues
- Allergy and Clinical Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (O.D.); (M.O.)
| | - Chantal J. Snoeck
- Clinical and Applied Virology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.C.); (C.J.S.)
| | - Markus Ollert
- Allergy and Clinical Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (O.D.); (M.O.)
| | - Carole Seguin-Devaux
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| | - Danielle Perez-Bercoff
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| |
Collapse
|
31
|
Dangi T, Sanchez S, Lew MH, Visvabharathy L, Richner J, Koralnik IJ, Penaloza-MacMaster P. Pre-existing immunity modulates responses to mRNA boosters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.06.27.497248. [PMID: 35794898 PMCID: PMC9258286 DOI: 10.1101/2022.06.27.497248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
mRNA vaccines have shown high efficacy in preventing severe COVID-19, but breakthrough infections, emerging variants and waning antibody levels have warranted the use of boosters. Although mRNA boosters have been widely implemented, the extent to which pre-existing immunity influences the efficacy of boosters remains unclear. In a cohort of individuals primed with the mRNA-1273 or BNT162b2 vaccines, we observed that lower antibody levels before boost were associated with higher fold-increase in antibody levels after boost, suggesting that pre-existing antibody modulates the boosting capacity of mRNA vaccines. Mechanistic studies in mice show that pre-existing antibodies significantly limit antigen expression and priming of B cell responses after mRNA vaccination. Furthermore, we demonstrate that the relative superiority of an updated Omicron vaccine over the original vaccine is critically dependent on the serostatus of the host. These data demonstrate that pre-existing immunity dictates responses to mRNA vaccination, elucidating specific circumstances when updated SARS-CoV-2 vaccines confer superior protection to original vaccines.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Min Han Lew
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lavanya Visvabharathy
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Justin Richner
- Department of Microbiology & Immunology, University of Illinois Chicago College of Medicine, Chicago, IL 60612
| | - Igor J. Koralnik
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
32
|
Rojas Chávez RA, Fili M, Han C, Rahman SA, Bicar IGL, Gregory S, Hu G, Das J, Brown GD, Haim H. Mutability Patterns Across the Spike Glycoprotein Reveal the Diverging and Lineage-specific Evolutionary Space of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.02.01.478697. [PMID: 35132415 PMCID: PMC8820662 DOI: 10.1101/2022.02.01.478697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Mutations in the spike glycoprotein of SARS-CoV-2 allow the virus to probe the sequence space in search of higher-fitness states. New sublineages of SARS-CoV-2 variants-of-concern (VOCs) continuously emerge with such mutations. Interestingly, the sites of mutation in these sublineages vary between the VOCs. Whether such differences reflect the random nature of mutation appearance or distinct evolutionary spaces of spike in the VOCs is unclear. Here we show that each position of spike has a lineage-specific likelihood for mutations to appear and dominate descendent sublineages. This likelihood can be accurately estimated from the lineage-specific mutational profile of spike at a protein-wide level. The mutability environment of each position, including adjacent sites on the protein structure and neighboring sites on the network of comutability, accurately forecast changes in descendent sublineages. Mapping of imminent changes within the VOCs can contribute to the design of immunogens and therapeutics that address future forms of SARS-CoV-2.
Collapse
|
33
|
Beavis AC, Li Z, Briggs K, Huertas-Díaz MC, Wrobel ER, Najera M, An D, Orr-Burks N, Murray J, Patil P, Huang J, Mousa J, Hao L, Hsiang TY, Gale M, Harvey SB, Tompkins SM, Hogan RJ, Lafontaine ER, Jin H, He B. Efficacy of Parainfluenza Virus 5 (PIV5)-vectored Intranasal COVID-19 Vaccine as a Single Dose Vaccine and as a Booster against SARS-CoV-2 Variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.06.07.495215. [PMID: 35702147 PMCID: PMC9196109 DOI: 10.1101/2022.06.07.495215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Immunization with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines has greatly reduced coronavirus disease 2019 (COVID-19)-related deaths and hospitalizations, but waning immunity and the emergence of variants capable of immune escape indicate the need for novel SARS-CoV-2 vaccines. An intranasal parainfluenza virus 5 (PIV5)-vectored COVID-19 vaccine CVXGA1 has been proven efficacious in animal models and blocks contact transmission of SARS-CoV-2 in ferrets. CVXGA1 vaccine is currently in human clinical trials in the United States. This work investigates the immunogenicity and efficacy of CVXGA1 and other PIV5-vectored vaccines expressing additional antigen SARS-CoV-2 nucleoprotein (N) or SARS-CoV-2 variant spike (S) proteins of beta, delta, gamma, and omicron variants against homologous and heterologous challenges in hamsters. A single intranasal dose of CVXGA1 induces neutralizing antibodies against SARS-CoV-2 WA1 (ancestral), delta variant, and omicron variant and protects against both homologous and heterologous virus challenges. Compared to mRNA COVID-19 vaccine, neutralizing antibody titers induced by CVXGA1 were well-maintained over time. When administered as a boost following two doses of a mRNA COVID-19 vaccine, PIV5-vectored vaccines expressing the S protein from WA1 (CVXGA1), delta, or omicron variants generate higher levels of cross-reactive neutralizing antibodies compared to three doses of a mRNA vaccine. In addition to the S protein, the N protein provides added protection as assessed by the highest body weight gain post-challenge infection. Our data indicates that PIV5-vectored COVID-19 vaccines, such as CVXGA1, can serve as booster vaccines against emerging variants.
Collapse
Affiliation(s)
- Ashley C. Beavis
- CyanVac LLC, Athens, Georgia, 30602
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Zhuo Li
- CyanVac LLC, Athens, Georgia, 30602
| | - Kelsey Briggs
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - María Cristina Huertas-Díaz
- CyanVac LLC, Athens, Georgia, 30602
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Elizabeth R. Wrobel
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | | | - Dong An
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Nichole Orr-Burks
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | | | - Jiachen Huang
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Jarrod Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Linhui Hao
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington
| | - Tien-Ying Hsiang
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington
| | - Stephen B. Harvey
- Animal Resources, University of Georgia, Athens, Georgia; Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - S. Mark Tompkins
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Robert Jeffrey Hogan
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Eric R. Lafontaine
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Hong Jin
- CyanVac LLC, Athens, Georgia, 30602
| | - Biao He
- CyanVac LLC, Athens, Georgia, 30602
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| |
Collapse
|
34
|
Nantanee R, Jantarabenjakul W, Jaru-Ampornpan P, Sodsai P, Himananto O, Athipunjapong J, Sophonphan J, Nanthapisal S, Hirankarn N, Puthanakit T. A Randomized Clinical Trial of a Fractional Low Dose of BNT162b2 Booster in Adults Following AZD1222. Vaccines (Basel) 2022; 10:914. [PMID: 35746522 PMCID: PMC9230769 DOI: 10.3390/vaccines10060914] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022] Open
Abstract
In the era of globally predominant omicron strains, a COVID-19 booster vaccine is needed. Our study aimed to evaluate the immunogenicity of a half-dose BNT162b2 booster after AZD1222 in healthy adults. A randomized trial of volunteers aged 18-69 years who received two-dose AZD1222 was conducted. The participants were randomized to receive the BNT162b2 vaccine intramuscularly-half (15 µg) vs. standard dose (30 µg). The immunogenicity was evaluated by a surrogate virus neutralization test (sVNT) against omicron variants and anti-spike-receptor-binding-domain IgG (anti-S-RBD IgG). From November-December 2021, 100 adults with a median age of 59.3 years (IQR 33.4-65.5) were enrolled. A booster dose was given at median of 98 days (IQR 92-128) after AZD1222. At day 14, the geometric means (GMs) of anti-S-RBD IgG in half- vs. standard-dose group were 2329.8 vs. 2574.7 BAU/mL, with a geometric mean ratio (GMR) of 0.90 (0.77-1.06). The GMs of sVNT against the omicron variant in the half- and standard-dose groups were 74.4% inhibition (95% CI 68.8-80.5) and 67.3% inhibition (57.9-78.1), respectively, with GMR of 0.95 (0.69-1.30). At day 90, the sVNT indicated 22.3% inhibition (95% CI 14.9-33.4) and 20.4% inhibition (13.1-32.0), respectively, with GMR of 1.09 (0.60-1.98). The fractional low-dose BNT162b2 mRNA booster vaccine provided non-inferior immunogenicity responses. During a shortage of vaccine supply, a fractional low dose should be considered for a booster vaccination program.
Collapse
Affiliation(s)
- Rapisa Nantanee
- Center of Excellence in Pediatric Infectious Diseases and Vaccines, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (R.N.); (W.J.); (J.A.)
- Pediatric Allergy and Clinical Immunology Research Unit, Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watsamon Jantarabenjakul
- Center of Excellence in Pediatric Infectious Diseases and Vaccines, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (R.N.); (W.J.); (J.A.)
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Peera Jaru-Ampornpan
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani 12120, Thailand;
| | - Pimpayao Sodsai
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Orawan Himananto
- Monoclonal Antibody Production and Application Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani 12120, Thailand;
| | - Jitthiwa Athipunjapong
- Center of Excellence in Pediatric Infectious Diseases and Vaccines, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (R.N.); (W.J.); (J.A.)
| | - Jiratchaya Sophonphan
- The HIV Netherlands Australia Thailand Research Collaboration (HIV-NAT), The Thai Red Cross AIDS Research Centre, Bangkok 10330, Thailand;
| | - Sira Nanthapisal
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand;
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Thanyawee Puthanakit
- Center of Excellence in Pediatric Infectious Diseases and Vaccines, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (R.N.); (W.J.); (J.A.)
| |
Collapse
|
35
|
Aparicio B, Ruiz M, Casares N, Silva L, Egea J, Pérez P, Albericio G, Esteban M, García-Arriaza J, Lasarte JJ, Sarobe P. Enhanced cross-recognition of SARS-CoV-2 Omicron variant by peptide vaccine-induced antibodies. Front Immunol 2022; 13:1044025. [PMID: 36761163 PMCID: PMC9902865 DOI: 10.3389/fimmu.2022.1044025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/29/2022] [Indexed: 01/26/2023] Open
Abstract
Current vaccines against SARS-CoV-2, based on the original Wuhan sequence, induce antibodies with different degrees of cross-recognition of new viral variants of concern. Despite potent responses generated in vaccinated and infected individuals, the Omicron (B.1.1.529) variant causes breakthrough infections, facilitating viral transmission. We previously reported a vaccine based on a cyclic peptide containing the 446-488 S1 sequence (446-488cc) of the SARS-CoV-2 spike (S) protein from Wuhan isolate. To provide the best immunity against Omicron, here we compared Omicron-specific immunity induced by a Wuhan-based 446-488cc peptide, by a Wuhan-based recombinant receptor-binding domain (RBD) vaccine and by a new 446-488cc peptide vaccine based on the Omicron sequence. Antibodies induced by Wuhan peptide 446-488cc in three murine strains not only recognized the Wuhan and Omicron 446-488 peptides similarly, but also Wuhan and Omicron RBD protein variants. By contrast, antibodies induced by the Wuhan recombinant RBD vaccine showed a much poorer cross-reactivity for the Omicron RBD despite similar recognition of Wuhan and Omicron peptide variants. Finally, although the Omicron-based 446-488cc peptide vaccine was poorly immunogenic in mice due to the loss of T cell epitopes, co-immunization with Omicron peptide 446-488cc and exogenous T cell epitopes induced strong cross-reactive antibodies that neutralized Omicron SARS-CoV-2 virus. Since mutations occurring within this sequence do not alter T cell epitopes in humans, these results indicate the robust immunogenicity of 446-488cc-based peptide vaccines that induce antibodies with a high cross-recognition capacity against Omicron, and suggest that this sequence could be included in future vaccines targeting the Omicron variant.
Collapse
Affiliation(s)
- Belén Aparicio
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - Marta Ruiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - Noelia Casares
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - Leyre Silva
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - Josune Egea
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - Patricia Pérez
- Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Guillermo Albericio
- Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Juan J Lasarte
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - Pablo Sarobe
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|