1
|
Kousar R, Akhtar T, Lin CJ, Lebedev T, Li YC, Yang CC, Wang WJ, Chen HF, Su WC, Biswas PK, Saqib NU, Belay SA, Chang TC, Guo DW, Li Q, Patrick B, Usama M, Wu CS, Ma WL, Sher YP, Huang CC, Hung MC, Li XG. Anti-SARS-CoV-2 and anticancer properties of triptolide and its derived carbonized nanomaterials. Cancer Lett 2025; 619:217677. [PMID: 40147583 DOI: 10.1016/j.canlet.2025.217677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
The COVID-19 pandemic remains an ongoing global health threat, yet effective treatments are still lacking. This has led to a high demand for complementary/alternative medicine, such as Chinese herbal medicines for curbing the COVID-19 pandemic. Given the dual anticancer and antiviral activities of many herbal drugs, they may hold a multifaceted potential to tackle both cancer and SARS-CoV-2. Triptolide is the major bioactive compound isolated from Tripterygium wilfordii Hook F (TwHF), a traditional Chinese medicinal herb recognized for its beneficial pharmacological properties in many diseases, including cancer and viral infection. However, its application in the clinic has been greatly limited due to its toxicity and poor water solubility. Here, from a screen of a natural compound library of Chinese Pharmacopoeia, we identified triptolide as a top candidate to inhibit cell entry of SARS-CoV-2. We demonstrated that triptolide robustly blocked viral entry at nanomolar concentrations in cellular models, with broad range activity against emerging Omicron variants of SARS-CoV-2. Mechanistically, triptolide disrupted the interaction of SARS-CoV-2 spike protein with its receptor ACE2. Furthermore, we synthesized water-soluble, triptolide-derived carbon quantum dots. Compared to triptolide, these highly biocompatible nanomaterials exhibited prominent antiviral capabilities against Omicron variants of SARS-CoV-2 with less cytotoxicity. Finally, we showed that triptolide-derived carbonized materials excelled in their anticancer properties compared to triptolide and Minnelide, a water-soluble analog of triptolide. Together, our results provide a rationale for the potential development of triptolide-carbonized derivatives as a promising antiviral candidate for the current pandemic and future outbreaks, as well as anticancer agents.
Collapse
Affiliation(s)
- Rubina Kousar
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung, 406040, Taiwan
| | - Tahira Akhtar
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, 406040, Taiwan
| | - Chin-Jung Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 20224, Taiwan
| | - Timofey Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Yi-Chuan Li
- Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung, 406040, Taiwan
| | - Chih-Chao Yang
- Department of Biological Science and Technology, China Medical University, Taichung, 406040, Taiwan
| | - Wei-Jan Wang
- Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung, 406040, Taiwan
| | - Hsiao-Fan Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Cell Biology, China Medical University, Taichung, 406040, Taiwan
| | - Wen-Chi Su
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; International Master's Program of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Pulak Kumar Biswas
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan
| | - Najm Us Saqib
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan
| | - Sefealem Assefa Belay
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung, 406040, Taiwan
| | - Tzu-Chi Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan
| | - Da-Wei Guo
- Department of Post-Baccalaureate Veterinary Medicine, Asia University, Taichung, 413305, Taiwan
| | - Qiangdu Li
- Department of Psychiatry, The Third Municipal Hospital of Weihai, Shandong Province, China
| | - Bbumba Patrick
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; International Master's Program of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan
| | - Muhammad Usama
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, 406040, Taiwan
| | - Chen-Shiou Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Department of Medical Research, Taichung Veterans General Hospital, Taichung, 407219, Taiwan
| | - Wen-Lung Ma
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan
| | - Yuh-Pyng Sher
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Center for Molecular Medicine, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Chih-Ching Huang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 20224, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan; Center for Molecular Medicine, China Medical University Hospital, Taichung, 404327, Taiwan.
| | - Xing-Guo Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
2
|
Baum HE, Santopaolo M, Francis O, Milodowski EJ, Entwistle K, Oliver E, Hitchings B, Diamond D, Thomas AC, Mitchell RE, Kibble M, Gupta K, Di Bartolo N, Klenerman P, Brown A, Morales-Aza B, Oliver J, Berger I, Toye AM, Finn A, Goenka A, Davidson AD, Ring S, Molloy L, Lewcock M, Northstone K, Roth F, Timpson NJ, Wooldridge L, Halliday A, Rivino L. Hybrid B- and T-Cell Immunity Associates With Protection Against Breakthrough Infection After Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination in Avon Longitudinal Study of Parents and Children (ALSPAC) Participants. J Infect Dis 2025:jiaf246. [PMID: 40392230 DOI: 10.1093/infdis/jiaf246] [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: 01/06/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Immunological memory to vaccination and viral infection involves the coordinated action of B and T cells; thus, integrated analysis of these 2 components is critical for understanding their respective contributions to protection against breakthrough infections (BIs) after vaccination. METHODS We investigated cellular and humoral immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and/or vaccination in 300 adult participants from the Avon Longitudinal Study of Parents and Children (ALSPAC). Participants were grouped by those with (cases) and without (controls) a history of SARS-CoV-2 infection. To provide a quantitative correlate for protection against BI in the 8-month period after the study, Youden index thresholds were calculated for all immune measures analyzed. RESULTS The magnitude of antibody and T-cell responses following the second vaccine dose was associated with protection against BI in participants with a history of SARS-CoV-2 infection (cases), but not in infection-naive controls. Over 8 months of follow-up, 2 threshold combinations provided the best performance for protection against BI in cases: (i) anti-spike immunoglobulin G (IgG) (≥666.4 binding antibody units [BAU]/mL) combined with anti-nucleocapsid pan-immunoglobulin (pan-Ig) (≥0.1332 BAU/mL) and (ii) spike 1-specific T cells (≥195.6 spot-forming units/106 peripheral blood mononuclear cells) combined with anti-N pan-Ig (≥0.1332 BAU/mL). Both combinations offered 100% specificity for detecting cases without BI, with sensitivities of 83.3% and 72.2%, respectively. CONCLUSIONS Collectively, these results suggest that hybrid B- and T-cell immunity offers superior protection from BI after coronavirus disease 2019 (COVID-19) vaccination, and this finding has implications for designing next-generation COVID-19 vaccines that are capable of eliciting immunity to a broader repertoire of SARS-CoV-2 proteins.
Collapse
Affiliation(s)
- Holly E Baum
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Marianna Santopaolo
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Ore Francis
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Emily J Milodowski
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Katrina Entwistle
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Elizabeth Oliver
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Benjamin Hitchings
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Divya Diamond
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Amy C Thomas
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ruth E Mitchell
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Milla Kibble
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Kapil Gupta
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Natalie Di Bartolo
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Paul Klenerman
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Begonia Morales-Aza
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Jennifer Oliver
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Imre Berger
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Ash M Toye
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Adam Finn
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Paediatric Immunology and Infectious Diseases, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Anu Goenka
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Paediatric Immunology and Infectious Diseases, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Andrew D Davidson
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Susan Ring
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Lynn Molloy
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Melanie Lewcock
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kate Northstone
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Firona Roth
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Nicholas J Timpson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Linda Wooldridge
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Alice Halliday
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Laura Rivino
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
3
|
Lino MM, Mather S, Trani M, Chen Y, Caubel P, De Bernardi B. Challenges and Innovations in Pharmacovigilance and Signal Management During the COVID-19 Pandemic: An Industry Perspective. Vaccines (Basel) 2025; 13:481. [PMID: 40432093 DOI: 10.3390/vaccines13050481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/29/2025] Open
Abstract
Vaccine marketing authorization holders (MAHs) are responsible for the conduction of global vaccine pharmacovigilance on their vaccine products. A safety signal is detected when a new adverse event (AE) or aspect of an AE occurs after exposure to the vaccine and warrants further investigation to determine whether a causal association may exist. Signal detection and evaluation (signal management) begins at the start of vaccine development, before an MAH submits an application for authorization to regulatory authorities, continues through the course of all clinical trials, and carries on beyond development into the post-marketing phase. As long as the vaccine remains authorized anywhere in the world, pharmacovigilance continues. During the time that the COVID-19 vaccine became widely available after authorization and approval, clinical trials were also ongoing, and therefore all clinical development and post-authorization safety information was closely monitored for safety by the MAH. MAH pharmacovigilance activities were adapted to manage the unprecedented volume of safety information that became available within a very short timeframe following worldwide vaccination campaigns. No vaccine had previously been administered to such a large number of individuals in such a short time, nor had there previously been a public health vaccine experience that was the subject of so many medical and non-medical writings. The MAH's COVID-19 vaccine signal detection methods included the continuous review of accruing clinical trial data and the quantitative and qualitative analyses of spontaneously reported experiences. Review of published and unpublished medical literature and epidemiology-based analyses such as observed vs. expected analysis based on reported adverse events following immunization (AEFIs) played key roles in pharmacovigilance and signal management. All methods of signal detection and evaluation have caveats, but when considered in totality, can advance our understanding of a vaccine's safety profile and therefore the risk-benefit considerations for vaccinating both individuals and large populations of people. All COVID-19 vaccines authorized for use were subject to an unprecedented level of pharmacovigilance by their individual MAHs, national regulatory authorities, public health organizations, and others during the years immediately following regulatory authorization and full approval. The intense worldwide focus on pharmacovigilance and the need for MAHs and regulatory/health authorities to quickly evaluate incoming safety information, spurred frequent and timely communications between national and regional health authorities and between MAHs and regulatory/health authorities, spotlighting a unique opportunity for individuals committed to patient safety to share important accruing safety information in a collegial and less traditionally formal manner than usual. The global pandemic precipitated by the SARS-CoV-2 virus created a significant impetus for MAHs to develop innovative vaccines to change the course of the COVID-19 pandemic. Pharmacovigilance also had to meet unprecedented needs. In this article, unique aspects of COVID-19 vaccine pharmacovigilance encountered by one MAH will be summarized.
Collapse
Affiliation(s)
- Maria Maddalena Lino
- Vaccine Research and Development and Worldwide Safety, Safety Surveillance and Risk Management, Pfizer, 20152 Milan, Italy
| | - Susan Mather
- Vaccine Research and Development and Worldwide Safety, Safety Surveillance and Risk Management, Pfizer, Collegeville, PA 10965, USA
| | - Marianna Trani
- Vaccine Research and Development and Worldwide Safety, Safety Surveillance and Risk Management, Pfizer, 20152 Milan, Italy
| | - Yan Chen
- Vaccine Research and Development and Worldwide Safety, Safety Surveillance and Risk Management, Pfizer, Collegeville, PA 10965, USA
| | - Patrick Caubel
- Vaccine Research and Development, Pfizer, Pearl River, New York, NY 19426, USA
| | - Barbara De Bernardi
- Vaccine Research and Development and Worldwide Safety, Safety Surveillance and Risk Management, Pfizer, 20152 Milan, Italy
| |
Collapse
|
4
|
Saha A, Ghosh Roy S, Dwivedi R, Tripathi P, Kumar K, Nambiar SM, Pathak R. Beyond the Pandemic Era: Recent Advances and Efficacy of SARS-CoV-2 Vaccines Against Emerging Variants of Concern. Vaccines (Basel) 2025; 13:424. [PMID: 40333293 PMCID: PMC12031379 DOI: 10.3390/vaccines13040424] [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: 02/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccination has been instrumental in curbing the transmission of SARS-CoV-2 and mitigating the severity of clinical manifestations associated with COVID-19. Numerous COVID-19 vaccines have been developed to this effect, including BioNTech-Pfizer and Moderna's mRNA vaccines, as well as adenovirus vector-based vaccines such as Oxford-AstraZeneca. However, the emergence of new variants and subvariants of SARS-CoV-2, characterized by enhanced transmissibility and immune evasion, poses significant challenges to the efficacy of current vaccination strategies. In this review, we aim to comprehensively outline the landscape of emerging SARS-CoV-2 variants of concern (VOCs) and sub-lineages that have recently surfaced in the post-pandemic years. We assess the effectiveness of existing vaccines, including their booster doses, against these emerging variants and subvariants, such as BA.2-derived sub-lineages, XBB sub-lineages, and BA.2.86 (Pirola). Furthermore, we discuss the latest advancements in vaccine technology, including multivalent and pan-coronavirus approaches, along with the development of several next-generation coronavirus vaccines, such as exosome-based, virus-like particle (VLP), mucosal, and nanomaterial-based vaccines. Finally, we highlight the key challenges and critical areas for future research to address the evolving threat of SARS-CoV-2 subvariants and to develop strategies for combating the emergence of new viral threats, thereby improving preparedness for future pandemics.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Sounak Ghosh Roy
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Naval Medical Research Command, Silver Spring, MD 20910, USA;
| | - Richa Dwivedi
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Kamal Kumar
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA;
| | - Shashank Manohar Nambiar
- Division of Hepatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
5
|
Almabhouh S, Cecon E, Basubas F, Molina-Fernandez R, Maciej Stepniewski T, Selent J, Jockers R, Rahmeh A, Oliva B, Fernandez-Fuentes N. Computational Design and Evaluation of Peptides to Target SARS-CoV-2 Spike-ACE2 Interaction. Molecules 2025; 30:1750. [PMID: 40333723 PMCID: PMC12029774 DOI: 10.3390/molecules30081750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 05/09/2025] Open
Abstract
The receptor-binding domain (RBD) of SARS-CoV-2 spike protein is responsible for the recognition of the Angiotensin-Converting Enzyme 2 (ACE2) receptor in human cells and, thus, plays a critical role in viral infection. The therapeutic value of targeting this interaction has been proven by a sizable body of research investigating antibodies, small proteins, aptamers, and peptides. This study presents a novel peptide that impinges the interaction between RBD and ACE2. Starting from a very large pool of structurally designed peptides extracted from our database, PepI-Covid19, a diverse set of peptides were studied using molecular dynamics simulations. Ten of the most promising were chemically synthesized and validated both in vitro and in a cell-based assay. Our results indicate that one of the peptides (PEP10) exhibited the highest disruption of the RBD/ACE2 complex, effectively blocking the binding of two molecules and consequently inhibiting the SARS-CoV-2 spike-mediated cell entry of viruses pseudotyped with the spike of the D614G, Delta, and Omicron variants. PEP10 can potentially serve as a scaffold that can be further optimized for improved affinity and efficacy.
Collapse
Affiliation(s)
- Saja Almabhouh
- Structural Bioinformatics Laboratory (GRIB-IMIM), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (S.A.); (R.M.-F.); (B.O.)
| | - Erika Cecon
- Institute Cochin, INSERM, CNRS, Université Paris Cité, F-75014 Paris, France; (E.C.); (R.J.)
| | - Florence Basubas
- Synthetic Biology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (F.B.); (A.R.)
| | - Ruben Molina-Fernandez
- Structural Bioinformatics Laboratory (GRIB-IMIM), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (S.A.); (R.M.-F.); (B.O.)
| | | | - Jana Selent
- GPCR Drug Discovery, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (T.M.S.); (J.S.)
| | - Ralf Jockers
- Institute Cochin, INSERM, CNRS, Université Paris Cité, F-75014 Paris, France; (E.C.); (R.J.)
| | - Amal Rahmeh
- Synthetic Biology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (F.B.); (A.R.)
| | - Baldo Oliva
- Structural Bioinformatics Laboratory (GRIB-IMIM), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (S.A.); (R.M.-F.); (B.O.)
| | - Narcis Fernandez-Fuentes
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth SY23 3EE, UK
| |
Collapse
|
6
|
Dhawan M, Thakur N, Sharma M, Rabaan AA. The comprehensive insights into the B-cells-mediated immune response against COVID-19 infection amid the ongoing evolution of SARS-CoV-2. Biomed Pharmacother 2025; 185:117936. [PMID: 40056829 DOI: 10.1016/j.biopha.2025.117936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025] Open
Abstract
The antibody-mediated immune response is crucial for the development of protective immunity against SARS-CoV-2, the virus responsible for the COVID-19 pandemic. Understanding the interaction between SARS-CoV-2 and the immune system is critical because new variants emerge as a result of the virus's ongoing evolution. Understanding the function of B cells in the SARS-CoV-2 infection process is critical for developing effective and long-lasting vaccines against this virus. Triggered by the innate immune response, B cells transform into memory B cells (MBCs). It is fascinating to observe how MBCs provide enduring immune defence, not only eradicating the infection but also safeguarding against future reinfection. If there is a lack of B cell activation or if the B cells are not functioning properly, it can lead to a serious manifestation of the disease and make immunisation less effective. Individuals with disruptions in the B cells have shown increased production of cytokines and chemokines, resulting in a poor prognosis for the disease. Therefore, we have developed an updated review article to gain insight into the involvement of B cells in SARS-CoV-2 infection. The discussion has covered the generation, functioning, and dynamics of neutralising antibodies (nAbs). Furthermore, we have emphasised immunotherapeutics that rely on nAbs.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, Punjab 141004, India; Trafford College, Altrincham, Altrincham, Manchester WA14 5PQ, UK.
| | - Nanamika Thakur
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Ali A Rabaan
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh 13328, Saudi Arabia; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan.
| |
Collapse
|
7
|
Karisola P, Kanerva M, Vuokko A, Liira H, Wang S, Kvarnström K, Varonen M, Suojalehto H, Alenius H. Patients with post-COVID-19 condition show minor blood transcriptomic changes, with altered erythrocyte gene expression in a male subgroup. Front Immunol 2025; 16:1500997. [PMID: 40191210 PMCID: PMC11968430 DOI: 10.3389/fimmu.2025.1500997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background The mechanisms underlying persistent symptoms after non-severe COVID-19 remain unclear. This study aimed to investigate transcriptomic changes in peripheral blood cells of patients with post-COVID-19 condition (PCC) and assess if distinct clinical subtypes with specific gene signatures could be identified. Methods The cohort included 111 PCC patients from the SARS-CoV-2 Omicron variant era, with 57 recovered (Recov) and 54 having prolonged symptoms indicative of PCC. The results were compared to 63 healthy controls (Ctrl) without known SARS-CoV-2 infection. Clinical data included patient assessments, laboratory results, comorbidities, and questionnaires on quality of life and functioning. Transcriptomic analysis and cellular deconvolution methods were used on total RNA from peripheral blood mononuclear cells (PBMCs). Results PCC patients had more comorbidities (mean 1.3) and more frequently (59%) at least one comorbidity than recovered patients (31%) and controls (24%). Overall, past COVID-19 illness or current PCC symptoms caused minimal changes in the blood cell transcriptome, with only 3-6 differentially expressed genes (DEGs) identified across comparisons. However, a subset of male PCC patients exhibited an increased fraction of deconvoluted erythroblasts and significant genome-wide gene expression changes, with 399 DEGs compared to recovered and control males. These genes were enriched in pathways related to heme metabolism and gas exchange in erythrocytes. Conclusions Persistent symptoms in PCC are multifactorial and not directly linked to peripheral blood cell gene expression changes. However, a subgroup of male PCC patients shows distinct erythrocyte responses that may contribute to long-term symptoms.
Collapse
Affiliation(s)
- Piia Karisola
- Human Microbiome (HUMI) Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mari Kanerva
- Department of Infection Control, TYKS Turku University Hospital, The Wellbeing Services County of Southwest Finland, Turku, Finland
- Outpatient Clinic for Long-Term Effects of COVID-19, Helsinki University Central Hospital, Helsinki, Finland
| | - Aki Vuokko
- Occupational Medicine, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Helena Liira
- Outpatient Clinic for Long-Term Effects of COVID-19, Helsinki University Central Hospital, Helsinki, Finland
| | - Shuyuan Wang
- Human Microbiome (HUMI) Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi Kvarnström
- Outpatient Clinic for Long-Term Effects of COVID-19, Helsinki University Central Hospital, Helsinki, Finland
| | - Mikko Varonen
- Outpatient Clinic for Long-Term Effects of COVID-19, Helsinki University Central Hospital, Helsinki, Finland
| | - Hille Suojalehto
- Occupational Medicine, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Harri Alenius
- Human Microbiome (HUMI) Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Garcia Lopez V, Plate L. Comparative Interactome Profiling of Nonstructural Protein 3 Across SARS-CoV-2 Variants Emerged During the COVID-19 Pandemic. Viruses 2025; 17:447. [PMID: 40143373 PMCID: PMC11946765 DOI: 10.3390/v17030447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
SARS-CoV-2 virus and its variants remain a global health threat, due to their capacity for rapid evolution. Variants throughout the COVID-19 pandemic exhibited variations in virulence, impacting vaccine protection and disease severity. Investigating nonstructural protein variants is critical to understanding viral evolution and manipulation of host protein interactions. We focus on nonstructural protein 3 (nsp3), with multiple domains with different activities, including viral polyprotein cleavage, host deubiquitylation, de-ISGylation, and double-membrane vesicle formation. Using affinity purification-mass spectrometry (AP-MS), we identify differential protein interactions in nsp3 caused by mutations found in variants identified between 2019 and 2024: Alpha 20I, Beta 20H, Delta 21I, Delta 21J, Gamma 20J, Kappa 21B, Lambda 21G, Omicron 21K, and Omicron 21L. A small set of amino acid substitutions in the N-terminal region of nsp3 (nsp3.1) could be traced to increased interactions with RNA-binding proteins, which are vital in viral replication. Meanwhile, variants of the central region of nsp3 (nsp3.2) were found to share interactions with protein quality control machinery, including ER-associated degradation. In this construct, shared trends in interactor enrichment are observed between Omicron 21K and Delta 21I. These results underscore how minor mutations reshape host interactions, emphasizing the evolutionary arms race between the host and virus. We provide a roadmap to track the interaction changes driven by SARS-CoV-2 variant evolution.
Collapse
Affiliation(s)
- Valeria Garcia Lopez
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37240, USA;
| | - Lars Plate
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37240, USA;
- Department of Chemistry, Vanderbilt University, Nashville, TN 37240, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
9
|
Müller S, Schmetz A, Knaul JK, Wilke T, Yang J, Dornig S, Lehmann C, Spinner CD. COVID-19 Disease Burden in the Omicron Variant-Dominated Endemic Phase: Insights from the ROUTINE-COV19 Study Using Real-World German Statutory Health Insurance Data. Viruses 2025; 17:424. [PMID: 40143351 PMCID: PMC11945757 DOI: 10.3390/v17030424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
The ROUTINE-COV19 study explores the burden of COVID-19 in Germany during the early endemic phase, assessing disease patterns and their impact on the healthcare system from 1 July 2022 to 30 June 2023. Using anonymized statutory health insurance data from over 3 million individuals in Thuringia and Saxony, COVID-19 cases were identified through diagnostic codes, with severe and critical cases defined by hospitalization and intensive care criteria. The study focused on high-risk populations as identified by the German Immunization Technical Advisory Group. During the study period, 414,648 new COVID-19 cases were documented, with peaks in October 2022 and March 2023. Severe cases occurred at a rate of 241.6 per 100,000 persons, with in-hospital mortality exceeding 12%. Critical cases requiring intensive care had an in-hospital mortality rate of 32.2%. COVID-19-related hospitalizations averaged 9.94 days, generating direct costs of EUR 64.9 million, while indirect costs from work absenteeism amounted to EUR 454.3 million, representing 7.5% of all-cause absenteeism costs. Despite entering an endemic phase, COVID-19 continues to pose a substantial burden, particularly among older adults and those with pre-existing cardiovascular conditions.
Collapse
Affiliation(s)
| | - Andrea Schmetz
- BioNTech Europe GmbH, 10179 Berlin, Germany; (A.S.); (J.K.K.)
| | - Julia K. Knaul
- BioNTech Europe GmbH, 10179 Berlin, Germany; (A.S.); (J.K.K.)
| | - Thomas Wilke
- Institut für Pharmakoökonomie und Arzneimittellogistik (IPAM), 23966 Wismar, Germany;
| | | | | | - Clara Lehmann
- Department of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Christoph D. Spinner
- Department of Clinical Medicine, University Medical Center, Technical University of Munich, 81675 Munich, Germany;
| |
Collapse
|
10
|
Shoshi HR, Basher AK, Pyash AS, Hossain MK, Chowdhury F, Hassan MZ. Hesitancy towards COVID-19 booster vaccine among healthcare workers in Bangladesh. BMC Health Serv Res 2025; 25:346. [PMID: 40050921 PMCID: PMC11884019 DOI: 10.1186/s12913-025-12482-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/25/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Despite completing the COVID-19 vaccination series, healthcare workers (HCWs) remain at an elevated risk of re-infection. Booster uptake, though essential for this group, remains poorly characterized among Bangladeshi HCWs. This study identified the prevalence and driving factors behind booster hesitancy among Bangladeshi HCWs, providing valuable insights for targeted interventions. METHOD From December 2022 to June 2023, we conducted a cross-sectional survey among 1772 HCWs enrolled from 20 healthcare facilities of all tiers purposively selected across four administrative divisions of Bangladesh. We collected information through face-to-face interviews regarding their sociodemographic, pre-existing, and currently existing medical conditions, COVID-19 vaccination status, and their intention, hesitancy, and willingness to receive future booster doses. We used a multivariable logistic regression model to analyze factors associated with booster hesitancy. Odd's ratio with 95% confidence intervals (CIs) was calculated for each factor, with p < 0.05 considered statistically significant. RESULT Of the 1772 HCWs interviewed in our study, 49% (879) were nurses [median age 36 years (IQR: 30.0-46.0)]; 69% were female. Among the respondents, 94% (1667) were willing to take a booster, and 6% (105) showed hesitancy. Safety concerns, especially regarding potential side effects post-booster administration (86%), emerged as the leading cause of booster hesitancy among healthcare workers. Our multivariable logistic regression analysis revealed that support staff, compared to physicians, were the most hesitant to receive any additional booster dose (aOR 4.68, 95% CI: 1.56-9.03; p=0.006). Compared to rural residency, HCWs with an urban residency type were also more reluctant to receive booster doses (aOR 4.45, 95% CI: 2.03-9.73; p < 0.001). CONCLUSION Concerns about side effects following booster administration were the primary driver of hesitancy in our study. Targeted interventions focusing on education and addressing these anxieties-supported by evidence-based communication strategies-could play a crucial role in improving booster acceptance and safeguarding this vulnerable workforce.
Collapse
Affiliation(s)
- Homayra Rahman Shoshi
- Programme for Respiratory Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Mohakhali, Dhaka, Bangladesh
| | - Ahamed Khairul Basher
- Programme for Respiratory Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Mohakhali, Dhaka, Bangladesh
| | - Ashrak Shad Pyash
- Programme for Respiratory Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Mohakhali, Dhaka, Bangladesh
| | - Md Kamal Hossain
- Programme for Respiratory Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Mohakhali, Dhaka, Bangladesh
| | - Fahmida Chowdhury
- Programme for Respiratory Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Mohakhali, Dhaka, Bangladesh
| | - Md Zakiul Hassan
- Programme for Respiratory Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Mohakhali, Dhaka, Bangladesh.
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
11
|
Algaissi A, Khan E, Tabassum H, Samreen S, Khamjan NA, Lohani M, Khan S, Kameli N, Madkhali F, Ahmad IZ. Campesterol and dithymoquinone as a potent inhibitors of SARS cov-2 main proteases-promising drug candidates for targeting its novel variants. J Biomol Struct Dyn 2025; 43:2534-2548. [PMID: 38288958 DOI: 10.1080/07391102.2023.2301684] [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: 02/06/2023] [Accepted: 09/13/2023] [Indexed: 02/16/2024]
Abstract
The sudden outbreak of the COVID-19 pandemic has currently taken approximately 2.4 million lives, with no specific medication and fast-tracked tested vaccines for prevention. These vaccines have their own adverse effects, which have severely affected the global healthcare system. The discovery of the main protease structure of coronavirus (Mpro/Clpro) has resulted in the identification of compounds having antiviral potential, especially from the herbal system. In this study, the computer-associated drug design tools were utilised to analyze the reported phytoconstituents of Nigella sativa for their antiviral activity against the main protease. Fifty-eight compounds were subjected to pharmacological parameter analysis to determine their lead likeness in comparison to the standard drugs (chloroquine and nirmatrelvir) used in the treatment of SARS-CoV-2. Nearly 31 compounds were docked against five different SARS-CoV-2 main proteases, and all compounds showed better binding affinity and inhibition constant against the proteases. However, dithymoquinone and campesterol displayed the best binding scores and hence were further subjected to dynamics and MMPBSA study for 100 ns. The stability analysis shows that dithymoquinone and campesterol show less variation in fluctuation in residues compared to standard complexes. Moreover, dithymoquinone exhibited higher binding affinity and favorable interaction followed by campesterol as compared to the standard drug. The in silico computational analysis provides a promising hit for regulating the main proteases activity.
Collapse
Affiliation(s)
- Abdullah Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Emerging and Epidemic Infectious Diseases Research Unit, Medical Research Center, Jazan University, Jazan, Saudi Arabia
| | - Elhan Khan
- Natural Products Laboratory, Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, India
| | - Heena Tabassum
- Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Sadiyah Samreen
- Natural Products Laboratory, Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, India
| | - Nizar A Khamjan
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Mohtashim Lohani
- Medical Research Centre, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, Ha'il University, Ha'il, Saudi Arabia
| | - Nader Kameli
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Faisal Madkhali
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Iffat Zareen Ahmad
- Natural Products Laboratory, Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
12
|
Zhang X, Reinsmoen NL, Kobashigawa JA. HLA Mismatches Identified by a Novel Algorithm Predict Risk of Antibody-mediated Rejection From De Novo Donor-specific Antibodies. Transplantation 2025; 109:519-526. [PMID: 39049137 DOI: 10.1097/tp.0000000000005140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
BACKGROUND The development of de novo donor-specific antibodies (dnDSA) and antibody-mediated rejection (AMR) remains a barrier to long-term graft and patient survival. Most dnDSA are directed against mismatched donor HLA-DQ antigens. Here, we describe a novel algorithm, which we have termed categorical amino acid mismatched epitope, to evaluate HLA-DQ mismatches. METHODS In this algorithm, amino acid residues of HLA-DQ protein were categorized into 4 groups based on their chemical characteristics. The likelihood of categorically mismatched peptides presented by the recipient's HLA-DRB1 was expressed as a normalized value, %Rank score. Categorical HLA-DQ mismatches were analyzed in 386 heart transplant recipients who were mismatched with their donors at the HLA-DQB1 locus. RESULTS We found that the presence of DQB1 mismatches with %Rank score ≤1 was associated with the development of dnDSA ( P = 0.002). Furthermore, dnDSA increased the risk of AMR only in recipients who had DQ mismatches with %Rank score ≤1 (hazard ratio = 5.8), but the freedom from AMR was comparable between recipients with dnDSA and those without dnDSA if %Rank scores of DQ mismatching were >1. CONCLUSIONS These results suggest that HLA-DQ mismatches evaluated by the categorical amino acid mismatched epitope algorithm can stratify the risk of development of dnDSA and AMR in heart transplant recipients.
Collapse
Affiliation(s)
- Xiaohai Zhang
- HLA and Immunogenetics Laboratory, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Nancy L Reinsmoen
- Independent HLA Consultant, Cedars-Sinai Medical Center, Scottsdale, AZ
| | | |
Collapse
|
13
|
Arora P, Zhang L, Nehlmeier I, Kempf A, Graichen L, Kreitz E, Sidarovich A, Rocha C, Gärtner S, Winkler M, Schulz S, Jäck HM, Hoffmann M, Pöhlmann S. Host cell lectins ASGR1 and DC-SIGN jointly with TMEM106B confer ACE2 independence and imdevimab resistance to SARS-CoV-2 pseudovirus with spike mutation E484D. J Virol 2025; 99:e0123024. [PMID: 39791910 PMCID: PMC11852847 DOI: 10.1128/jvi.01230-24] [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: 07/16/2024] [Accepted: 11/28/2024] [Indexed: 01/12/2025] Open
Abstract
The naturally occurring mutation E484D in the spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can render viral entry ACE2 independent and imdevimab resistant. Here, we investigated whether the cellular proteins ASGR1, DC-SIGN, and TMEM106B, which interact with the viral S protein, can contribute to these processes. Employing S protein-pseudotyped particles, we found that expression of ASGR1 or DC-SIGN jointly with TMEM106B allowed for robust entry of mutant E484D into otherwise non-susceptible cells, while this effect was not observed upon separate expression of the single proteins and upon infection with SARS-CoV-2 wild type (WT). Furthermore, expression of ASGR1 or DC-SIGN conferred ACE2 independence and imdevimab resistance to entry of mutant E484D but not WT, and entry under those conditions was dependent on endogenous TMEM106B. These results suggest that engagement of certain cellular lectins can direct SARS-CoV-2 mutant E484D to an ACE2-independent, TMEM106B-dependent entry pathway that is not inhibited by imdevimab.IMPORTANCEThe interaction of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein with the ACE2 receptor determines the viral cell tropism and is the key target of the neutralizing antibody response. Here, we show that SARS-CoV-2 with a single, naturally occurring mutation in the spike protein, E484D, can use the cellular lectins ASGR1 and DC-SIGN in conjunction with TMEM106B for ACE2-independent entry and evasion of therapeutic antibodies. These results suggest that engagement of cellular lectins might modulate target cell choice of SARS-CoV-2 and might allow evasion of certain neutralizing antibodies.
Collapse
Affiliation(s)
- Prerna Arora
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Lu Zhang
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Luise Graichen
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Eike Kreitz
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Anzhalika Sidarovich
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Cheila Rocha
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Sabine Gärtner
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Michael Winkler
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Sebastian Schulz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
14
|
Kamelian K, Sievers B, Chen-Xu M, Turner S, Cheng MTK, Altaf M, Kemp SA, Abdullahi A, Csiba K, Collier DA, Mlcochova P, Meng B, Jones RB, The CITIID-NIHR BioResource COVID-19 Collaboration, Smith D, Bradley J, Smith KGC, Doffinger R, Smith RM, Gupta RK. Humoral responses to SARS-CoV-2 vaccine in vasculitis-related immune suppression. SCIENCE ADVANCES 2025; 11:eadq3342. [PMID: 39937891 PMCID: PMC11817922 DOI: 10.1126/sciadv.adq3342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025]
Abstract
Immune suppression poses a challenge to vaccine immunogenicity. We show that serum antibody neutralization against SARS-CoV-2 Omicron descendants was largely absent post-doses 1 and 2 in individuals with vasculitis treated with rituximab. Detectable and increasing neutralizing titers were observed post-doses 3 and 4, except for XBB. Rituximab in vasculitis exacerbates neutralization deficits over standard immunosuppressive therapy, although impairment resolves over time since dosing. We observed discordance between detectable IgG binding and neutralizing activity specifically in the context of rituximab use, with high proportions of individuals showing reasonable IgG titer but no neutralization. ADCC response was more frequently detectable compared to neutralization in the context of rituximab, indicating that a notable proportion of binding antibodies are non-neutralizing. Therefore, use of rituximab is associated with severe impairment in neutralization against Omicron descendants despite repeated vaccinations, with better preservation of non-neutralizing antibody activity.
Collapse
Affiliation(s)
- Kimia Kamelian
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Benjamin Sievers
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Michael Chen-Xu
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, Cambridgeshire, UK
| | - Sam Turner
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Mark Tsz Kin Cheng
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Mazharul Altaf
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Steven A. Kemp
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Kata Csiba
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Dami A. Collier
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Bo Meng
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Rachel B. Jones
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, Cambridgeshire, UK
| | | | - Derek Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - John Bradley
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Department of Renal Medicine, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Kenneth G. C. Smith
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia
- University of Melbourne, Melbourne, VIC 3010, Australia
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrooke’s Hospital, Cambridge, UK
| | - Rona M. Smith
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, Cambridgeshire, UK
- Department of Renal Medicine, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Ravindra K. Gupta
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
- Africa Health Research Institute, Durban, South Africa
| |
Collapse
|
15
|
Batlle D, Hassler L, Wysocki J. ACE2, From the Kidney to SARS-CoV-2: Donald Seldin Award Lecture 2023. Hypertension 2025; 82:166-180. [PMID: 39624896 DOI: 10.1161/hypertensionaha.124.22064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
ACE2 (angiotensin-converting enzyme 2) is a monocarboxypeptidase that cleaves Ang II (angiotensin II) among other substrates. ACE2 is present in the cell membrane of many organs, most abundantly in epithelial cells of kidney proximal tubules and the small intestine, and also exists in soluble forms in plasma and body fluids. Membrane-bound ACE2 exerts a renoprotective action by metabolizing Ang II and therefore attenuating the undesirable actions of excess Ang II. Therefore, soluble ACE2, by downregulating this peptide, may exert a therapeutic action. Our laboratory has designed ACE2 truncates that pass the glomerular filtration barrier to target the kidney renin-angiotensin system directly and, therefore, compensate for loss of kidney membrane-bound ACE2. Membrane-bound ACE2 is also the essential receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Soluble ACE2 proteins have been studied as a way to intercept SARS-CoV-2 from binding to membrane-bound ACE2 and prevent cell entry of SARS-CoV-2 altogether. We bioengineered a soluble ACE2 protein, termed ACE2 618-DDC-ABD, with increased binding affinity for SARS-CoV-2 and prolonged duration of action, which, when administered intranasally, provides near-complete protection from lethality in k18hACE2 mice infected with different SARS-CoV-2 variants. The main advantage of soluble ACE2 proteins for the neutralization of SARS-CoV-2 is their immediate onset of action and universality for current and future emerging SARS-CoV-2 variants. It is notable that ACE2 is critically involved in 2 dissimilar functions: as a receptor for cell entry of many coronaviruses and as an enzyme in the metabolism of Ang II, and yet in both cases, it is a therapeutic target.
Collapse
Affiliation(s)
- Daniel Batlle
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Luise Hassler
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jan Wysocki
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
16
|
Jiang H, Xia H, Wang Z, Xiong F. Discovery of Severe Acute Respiratory Syndrome Coronavirus 2 Main Protease Inhibitors through Rational Design of Novel Fluorinated 1,3,4-oxadiazole Amide Derivatives: An In-Silico Study. Chem Biodivers 2025:e202403179. [PMID: 39853882 DOI: 10.1002/cbdv.202403179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/23/2024] [Accepted: 01/24/2025] [Indexed: 01/26/2025]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) variants continue to emerge, there is an urgent need to develop more effective antiviral drugs capable of combating the COVID-19 pandemic. The main protease (Mpro) of SARS-CoV-2 is an evolutionarily conserved drug discovery target. The present study mainly focused on chemoinformatics computational methods to investigate the efficacy of our newly designed trifluoromethyl-1,3,4-oxadiazole amide derivatives as SARS-CoV-2 Mpro inhibitors. Drug-likeness absorption, distribution, metabolism, excretion, and toxicity analysis, molecular docking simulation, density functional theory (DFT), and molecular dynamics simulation methods were included. A comprehensive drug-likeness analysis was performed on the 14 newly designed compounds (1a-1n), and this series of small molecule inhibitors showed potential anti-SARS-CoV-2 activity. In order to reveal the mechanism of drug interaction, these novel compounds were classified by structure, and molecular docking simulations were performed. The results showed good interactions and identified the key amino acid residue GLY-143. Further DFT analysis using B3LYP-D3BJ functional and 6-311 + + G (d, p) basis set was performed to optimize the optimal configuration of the Mpro inhibitors, and the infrared spectrum of the vibration frequency was analyzed to clearly understand the structure and stability of the drug. The electrostatic potential map was analyzed to predict the reactivity of functional groups and protein-substrate interactions. The frontier molecular orbital analysis and density of states map showed the reactivity level and stability of the drug itself, among which 1i had the smallest energy gap difference (ΔEgap = 3.64 ev), showing good reactivity. The analysis of global reactivity descriptors such as electrophilic index (ω) and chemical potential (μ) also showed that our newly designed Mpro inhibitors had stronger interactions. Molecular dynamics simulation further revealed the stable binding of the Mpro inhibitors in a solvent environment. The binding free energy results calculated by Molecular Mechanics / Poisson Boltzmann Surface Area (MM/PBSA) all exceeded the Food and Drug Administration-approved standard reference drug (Nirmatrelvir), and the free energy landscape and principal component analysis also further described the energy sites formed during the binding process between the drug molecule and the ligand-protein and the changes in conformation. These new series of small molecule inhibitors studied in this work will provide the necessary theoretical basis for the synthesis and activity evaluation of novel SARS-CoV-2 Mpro inhibitors.
Collapse
Affiliation(s)
- Huiying Jiang
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, P. R. China
| | - Heping Xia
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, P. R. China
| | - Zhonghua Wang
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, P. R. China
| | - Fei Xiong
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, P. R. China
| |
Collapse
|
17
|
Yang H, Xie Y, Li S, Bao C, Wang J, Li C, Nie J, Quan Y. Immunogenicity of intranasal vaccine based on SARS-CoV-2 spike protein during primary and booster immunizations in mice. Hum Vaccin Immunother 2024; 20:2364519. [PMID: 38880868 PMCID: PMC11181929 DOI: 10.1080/21645515.2024.2364519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/18/2024] Open
Abstract
Mucosal immunity plays a crucial role in combating and controlling the spread of highly mutated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Recombinant subunit vaccines have shown safety and efficacy in clinical trials, but further investigation is necessary to evaluate their feasibility as mucosal vaccines. This study developed a SARS-CoV-2 mucosal vaccine using spike (S) proteins from a prototype strain and the omicron variant, along with a cationic chitosan adjuvant, and systematically evaluated its immunogenicity after both primary and booster immunization in mice. Primary immunization through intraperitoneal and intranasal administration of the S protein elicited cross-reactive antibodies against prototype strains, as well as delta and omicron variants, with particularly strong effects observed after mucosal vaccination. In the context of booster immunization following primary immunization with inactivated vaccines, the omicron-based S protein mucosal vaccine resulted in a broader and more robust neutralizing antibody response in both serum and respiratory mucosa compared to the prototype vaccine, enhancing protection against different variants. These findings indicate that mucosal vaccination with the S protein has the potential to trigger a broader and stronger antibody response during primary and booster immunization, making it a promising strategy against respiratory pathogens.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Administration, Intranasal
- Mice
- Immunization, Secondary/methods
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Mice, Inbred BALB C
- Female
- Immunity, Mucosal
- Immunogenicity, Vaccine
- Cross Reactions/immunology
- Chitosan/immunology
- Chitosan/administration & dosage
- Adjuvants, Vaccine/administration & dosage
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
Collapse
Affiliation(s)
- Huijie Yang
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
| | - Ying Xie
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, People’s Republic of China
| | - Shuyan Li
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
| | - Chunting Bao
- Changchun Institute of Biological Products, Changchun, China
| | - Jiahao Wang
- Sinovac Life Sciences Co., Ltd., Beijing, China
| | - Changgui Li
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
| | - Jiaojiao Nie
- Department of R&D, Beijing Yunling Biotechnology Co., Ltd., Beijing, China
| | - Yaru Quan
- Division of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
| |
Collapse
|
18
|
Padilla-Bórquez DL, Matuz-Flores MG, Hernández-Bello J, Rosas-Rodríguez JA, Turrubiates-Hernández FJ, García-Arellano S, González-Estevez G, Ceja-Galvez HR, Oregon-Romero E, López-Reyes A, Muñoz-Valle JF. Influence of previous COVID-19 exposure and vaccine type (CoronaVac, ChAdOx1 nCov-19 or BNT162b2) on antibody and cytokine (Th1 or Th2) responses. Hum Vaccin Immunother 2024; 20:2394265. [PMID: 39246041 PMCID: PMC11385164 DOI: 10.1080/21645515.2024.2394265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024] Open
Abstract
To achieve global herd immunity, widespread vaccination is the most effective strategy. Vaccines stimulate the immune system, generating cytokines and chemokines, isotype antibodies, and neutralizing antibodies; all these molecules collectively provide a more comprehensive characterization of the immune response post-vaccination. We conducted a longitudinal study in northwestern Mexico, involving 120 individuals before vaccination and after the first dose of the SARS-CoV-2 vaccine, and 46 individuals after their second dose. Our findings reveal that antibody levels stabilize over time; cytokine levels generally increase following the first dose but decrease after the second dose and higher than normal levels in IgG1 and IgG3 concentrations are present. Most of the innate cytokines determined in this study were higher after the first dose of the vaccine. Regardless of previous infection history, this finding suggests that the first dose of the vaccine is crucial and may stimulate immunity by enhancing the innate immune response. Conversely, increased levels of IL-4, indicative of a Th2 response, were found in individuals without prior exposure to the virus and in those vaccinated with CoronaVac. These results suggest that the immune response to COVID-19 vaccines is multi-faceted, with preexisting immunity potentiating a more robust innate response. Vaccine type plays a critical role, with genetic vaccines favoring a Th1 response and inactivated vaccines like CoronaVac skewing toward a Th2 profile.
Collapse
Affiliation(s)
- Diana Lourdes Padilla-Bórquez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Mónica Guadalupe Matuz-Flores
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Jorge Hernández-Bello
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Jesús Alfredo Rosas-Rodríguez
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Navojoa, México
| | - Francisco Javier Turrubiates-Hernández
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Samuel García-Arellano
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Guillermo González-Estevez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Hazael Ramiro Ceja-Galvez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Edith Oregon-Romero
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Ciudad de México, México
| | - Jose Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| |
Collapse
|
19
|
Gonzalez-Carmona MA, Schmitz AM, Berger M, Baier LI, Gorny JG, Sadeghlar F, Anhalt T, Zhou X, Zhou T, Mahn R, Möhring C, Linnemann T, Schmid M, Strassburg CP, Boesecke C, Rockstroh JK, Eis-Hübinger AM, Monin MB. Longitudinal Study of SARS-CoV-2 Vaccinations and Infections in Patients with Gastrointestinal Cancer: Stabilizing Immune Responses and Neutralizing Emerging Variants with Variant-Adapted Antigen Exposures. Int J Mol Sci 2024; 25:13613. [PMID: 39769379 PMCID: PMC11728159 DOI: 10.3390/ijms252413613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
This longitudinal study examined how active gastrointestinal (GI) cancer types affect immune responses to SARS-CoV-2, focusing on the ability to neutralize the Omicron variants. Patients with GI cancer (n = 168) were categorized into those with hepatocellular carcinoma, hepatic metastatic GI cancer, non-hepatic metastatic GI cancer, and two control groups of patients with and without underlying liver diseases. Humoral and cellular immune responses were evaluated before and after Omicron antigen exposures. In the pre-Omicron era, humoral SARS-CoV-2 immunity decreased after three antigen contacts without further antigen exposure. While Omicron neutralization was significantly lower than wildtype neutralization (p < 0.01), Omicron infections were yet mild to moderate. Additional Omicron exposures improved IgG levels (p < 0.01) and Omicron neutralization (p < 0.01). However, this effect was significantly less intense in patients with active GI cancer, particularly in patients with pancreaticobiliary neoplasms (PBN; p = 0.04), with underlying immunodeficiency (p = 0.05), and/or under conventional chemotherapy (p = 0.05). Pre-Omicron SARS-CoV-2 immunity prevented severe clinical courses of infections with Omicron variants in patients with GI cancer. However, in patients with PBN, with underlying immunodeficiency, and/or under conventional chemotherapy initial contacts with Omicron antigens triggered only reduced immune responses. Thus, subgroups could be identified for whom booster vaccinations are of special clinical significance.
Collapse
Affiliation(s)
- Maria A. Gonzalez-Carmona
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alina M. Schmitz
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Moritz Berger
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Leona I. Baier
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jens G. Gorny
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Farsaneh Sadeghlar
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Thomas Anhalt
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Xin Zhou
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Taotao Zhou
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Robert Mahn
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christian Möhring
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Thomas Linnemann
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Matthias Schmid
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christian P. Strassburg
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christoph Boesecke
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jürgen K. Rockstroh
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | | | - Malte B. Monin
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Partner-Site Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Infektionsmedizinisches Centrum Hamburg (ICH), Glockengießerwall 1, 20095 Hamburg, Germany
| |
Collapse
|
20
|
Kawamura S, Yamaguchi F, Kusakado R, Go Y, Nohmi S, Yoshizaki C, Yoshida Y, Izumizaki K, Saito Y, Kobayashi H, Hirata K, Miyo K, Kondo C, Kanzaki M, Ding Y, Yokoe T, Kobayashi S, Suzuki H. Changes in Clinical Features and Severity of COVID-19 with the Emergence of Omicron Variants: A Shift Towards a Common Disease. Infect Drug Resist 2024; 17:5595-5603. [PMID: 39711829 PMCID: PMC11663384 DOI: 10.2147/idr.s492816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024] Open
Abstract
Background The emergence of the Omicron variant of severe acute respiratory syndrome coronavirus-2 has significantly altered the clinical features and severity of coronavirus disease 2019 (COVID-19). Objective This study aims to evaluate whether the clinical factors that previously predicted COVID-19 remain valid following the emergence of the Omicron variant. Methods This cross-sectional study was conducted at Showa University Fujigaoka Hospital from April 2022 to March 2023. A total of 576 patients with suspected COVID-19 were included, of which 258 (44.8%) were diagnosed with COVID-19 based on real-time reverse-transcription polymerase chain reaction tests. Clinical data were collected retrospectively, and multivariate logistic regression was used to analyze factors associated with a COVID-19 diagnosis. Results Of the 258 patients diagnosed with COVID-19, 60% had mild disease, and the overall severity was lower than in previous reports prior to the emergence of the Omicron variant. In the multivariate analysis, only C-reactive protein (CRP) levels were significantly associated with COVID-19 (odds ratio, 0.3164; 95% confidence interval, 0.2077-0.4819), while factors such as age, sex, body mass index, lactate dehydrogenase, and comorbidities were not significantly associated. Non-COVID-19 cases were primarily bacterial infections, accounting for 57.2% of the non-COVID-19 diagnoses. Mortality rates did not differ significantly between the COVID-19 and non-COVID-19 groups. Conclusion The clinical characteristics of COVID-19 have become less distinct since the emergence of the Omicron variant, with CRP being the primary marker associated with a COVID-19 diagnosis. As COVID-19 continues to transition towards a more common infectious disease, distinguishing it will become increasingly challenging.
Collapse
Affiliation(s)
- Saori Kawamura
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Fumihiro Yamaguchi
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Rui Kusakado
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yoshihiro Go
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Shiho Nohmi
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Chinatsu Yoshizaki
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yuki Yoshida
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kensuke Izumizaki
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yuichiro Saito
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hitoshi Kobayashi
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kento Hirata
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kenta Miyo
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Chika Kondo
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Mamiko Kanzaki
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yize Ding
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Takuya Yokoe
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Sei Kobayashi
- Department of Otolaryngology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hiroshi Suzuki
- Department of Cardiovascular Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| |
Collapse
|
21
|
Brandi R, Paganelli A, D’Amelio R, Giuliani P, Lista F, Salemi S, Paganelli R. mRNA Vaccines Against COVID-19 as Trailblazers for Other Human Infectious Diseases. Vaccines (Basel) 2024; 12:1418. [PMID: 39772079 PMCID: PMC11680146 DOI: 10.3390/vaccines12121418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
mRNA vaccines represent a milestone in the history of vaccinology, because they are safe, very effective, quick and cost-effective to produce, easy to adapt should the antigen vary, and able to induce humoral and cellular immunity. METHODS To date, only two COVID-19 mRNA and one RSV vaccines have been approved. However, several mRNA vaccines are currently under development for the prevention of human viral (influenza, human immunodeficiency virus [HIV], Epstein-Barr virus, cytomegalovirus, Zika, respiratory syncytial virus, metapneumovirus/parainfluenza 3, Chikungunya, Nipah, rabies, varicella zoster virus, and herpes simplex virus 1 and 2), bacterial (tuberculosis), and parasitic (malaria) diseases. RESULTS RNA viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV)-2, HIV, and influenza, are characterized by high variability, thus creating the need to rapidly adapt the vaccines to the circulating viral strain, a task that mRNA vaccines can easily accomplish; however, the speed of variability may be higher than the time needed for a vaccine to be adapted. mRNA vaccines, using lipid nanoparticles as the delivery system, may act as adjuvants, thus powerfully stimulating innate as well as adaptive immunity, both humoral, which is rapidly waning, and cell-mediated, which is highly persistent. Safety profiles were satisfactory, considering that only a slight increase in prognostically favorable anaphylactic reactions in young females and myopericarditis in young males has been observed. CONCLUSIONS The COVID-19 pandemic determined a shift in the use of RNA: after having been used in medicine as micro-RNAs and tumor vaccines, the new era of anti-infectious mRNA vaccines has begun, which is currently in great development, to either improve already available, but unsatisfactory, vaccines or develop protective vaccines against infectious agents for which no preventative tools have been realized yet.
Collapse
Affiliation(s)
- Rossella Brandi
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | | | | | - Paolo Giuliani
- Poliambulatorio Montezemolo, Ente Sanitario Militare del Ministero Della Difesa Presso la Corte dei Conti, 00195 Rome, Italy;
| | - Florigio Lista
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | - Simonetta Salemi
- Division of Internal Medicine, Azienda Ospedaliero-Universitaria S. Andrea, 00189 Rome, Italy
| | - Roberto Paganelli
- Internal Medicine, Faculty of Medicine and Surgery, Unicamillus, International School of Medicine, 00131 Rome, Italy
| |
Collapse
|
22
|
Bayarri-Olmos R, Sutta A, Rosbjerg A, Mortensen MM, Helgstrand C, Nielsen PF, Pérez-Alós L, González-García B, Johnsen LB, Matthiesen F, Egebjerg T, Hansen CB, Sette A, Grifoni A, da Silva Antunes R, Garred P. Unraveling the impact of SARS-CoV-2 mutations on immunity: insights from innate immune recognition to antibody and T cell responses. Front Immunol 2024; 15:1412873. [PMID: 39720734 PMCID: PMC11666439 DOI: 10.3389/fimmu.2024.1412873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024] Open
Abstract
Throughout the COVID-19 pandemic, the emergence of new viral variants has challenged public health efforts, often evading antibody responses generated by infections and vaccinations. This immune escape has led to waves of breakthrough infections, raising questions about the efficacy and durability of immune protection. Here we focus on the impact of SARS-CoV-2 Delta and Omicron spike mutations on ACE-2 receptor binding, protein stability, and immune response evasion. Delta and Omicron variants had 3-5 times higher binding affinities to ACE-2 than the ancestral strain (KDwt = 23.4 nM, KDDelta = 8.08 nM, KDBA.1 = 4.77 nM, KDBA.2 = 4.47 nM). The pattern recognition molecule mannose-binding lectin (MBL) has been shown to recognize the spike protein. Here we found that MBL binding remained largely unchanged across the variants, even after introducing mutations at single glycan sites. Although MBL binding decreased post-vaccination, it increased by 2.6-fold upon IgG depletion, suggesting a compensatory or redundant role in immune recognition. Notably, we identified two glycan sites (N717 and N801) as potentially essential for the structural integrity of the spike protein. We also evaluated the antibody and T cell responses. Neutralization by serum immunoglobulins was predominantly mediated by IgG rather than IgA and was markedly impaired against the Delta (5.8-fold decrease) and Omicron variants BA.1 (17.4-fold) and BA.2 (14.2-fold). T cell responses, initially conserved, waned rapidly within 3 months post-Omicron infection. Our data suggests that immune imprinting may have hindered antibody and T cell responses toward the variants. Overall, despite decreased antibody neutralization, MBL recognition and T cell responses were generally unaffected by the variants. These findings extend our understanding of the complex interplay between viral adaptation and immune response, underscoring the importance of considering MBL interactions, immune imprinting, and viral evolution dynamics in developing new vaccine and treatment strategies.
Collapse
Affiliation(s)
- Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | - Adrian Sutta
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | - Anne Rosbjerg
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Beatriz González-García
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Huuskonen S, Liu X, Pöhner I, Redchuk T, Salokas K, Lundberg R, Maljanen S, Belik M, Reinholm A, Kolehmainen P, Tuhkala A, Tripathi G, Laine P, Belanov S, Auvinen P, Vartiainen M, Keskitalo S, Österlund P, Laine L, Poso A, Julkunen I, Kakkola L, Varjosalo M. The comprehensive SARS-CoV-2 'hijackome' knowledge base. Cell Discov 2024; 10:125. [PMID: 39653747 PMCID: PMC11628605 DOI: 10.1038/s41421-024-00748-y] [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: 04/30/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
The continuous evolution of SARS-CoV-2 has led to the emergence of several variants of concern (VOCs) that significantly affect global health. This study aims to investigate how these VOCs affect host cells at proteome level to better understand the mechanisms of disease. To achieve this, we first analyzed the (phospho)proteome changes of host cells infected with Alpha, Beta, Delta, and Omicron BA.1 and BA.5 variants over time frames extending from 1 to 36 h post infection. Our results revealed distinct temporal patterns of protein expression across the VOCs, with notable differences in the (phospho)proteome dynamics that suggest variant-specific adaptations. Specifically, we observed enhanced expression and activation of key components within crucial cellular pathways such as the RHO GTPase cycle, RNA splicing, and endoplasmic reticulum-associated degradation (ERAD)-related processes. We further utilized proximity biotinylation mass spectrometry (BioID-MS) to investigate how specific mutation of these VOCs influence viral-host protein interactions. Our comprehensive interactomics dataset uncovers distinct interaction profiles for each variant, illustrating how specific mutations can change viral protein functionality. Overall, our extensive analysis provides a detailed proteomic profile of host cells for each variant, offering valuable insights into how specific mutations may influence viral protein functionality and impact therapeutic target identification. These insights are crucial for the potential use and design of new antiviral substances, aiming to enhance the efficacy of treatments against evolving SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Sini Huuskonen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Xiaonan Liu
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ina Pöhner
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Taras Redchuk
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kari Salokas
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Sari Maljanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Milja Belik
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arttu Reinholm
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Antti Tuhkala
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Garima Tripathi
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Pia Laine
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sergei Belanov
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Petri Auvinen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Maria Vartiainen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Salla Keskitalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Pamela Österlund
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland
| | - Larissa Laine
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland
| | - Antti Poso
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
- InFlames Research Flagship Center, University of Turku, Turku, Finland
| | - Laura Kakkola
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
24
|
Qing E, Salgado J, Wilcox A, Gallagher T. SARS-CoV-2 Omicron variations reveal mechanisms controlling cell entry dynamics and antibody neutralization. PLoS Pathog 2024; 20:e1012757. [PMID: 39621785 PMCID: PMC11637440 DOI: 10.1371/journal.ppat.1012757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/12/2024] [Accepted: 11/17/2024] [Indexed: 12/14/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is adapting to continuous presence in humans. Transitions to endemic infection patterns are associated with changes in the spike (S) proteins that direct virus-cell entry. These changes generate antigenic drift and thereby allow virus maintenance in the face of prevalent human antiviral antibodies. These changes also fine tune virus-cell entry dynamics in ways that optimize transmission and infection into human cells. Focusing on the latter aspect, we evaluated the effects of several S protein substitutions on virus-cell membrane fusion, an essential final step in enveloped virus-cell entry. Membrane fusion is executed by integral-membrane "S2" domains, yet we found that substitutions in peripheral "S1" domains altered late-stage fusion dynamics, consistent with S1-S2 heterodimers cooperating throughout cell entry. A specific H655Y change in S1 stabilized a fusion-intermediate S protein conformation and thereby delayed membrane fusion. The H655Y change also sensitized viruses to neutralization by S2-targeting fusion-inhibitory peptides and stem-helix antibodies. The antibodies did not interfere with early fusion-activating steps; rather they targeted the latest stages of S2-directed membrane fusion in a novel neutralization mechanism. These findings demonstrate that single amino acid substitutions in the S proteins both reset viral entry-fusion kinetics and increase sensitivity to antibody neutralization. The results exemplify how selective forces driving SARS-CoV-2 fitness and antibody evasion operate together to shape SARS-CoV-2 evolution.
Collapse
Affiliation(s)
- Enya Qing
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Julisa Salgado
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Alexandria Wilcox
- McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Tom Gallagher
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, United States of America
| |
Collapse
|
25
|
Sun H, Xia L, Li J, Zhang Y, Zhang G, Huang P, Wang X, Cui Y, Fang T, Fan P, Zhou Q, Chi X, Yu C. A novel bispecific antibody targeting two overlapping epitopes in RBD improves neutralizing potency and breadth against SARS-CoV-2. Emerg Microbes Infect 2024; 13:2373307. [PMID: 38953857 PMCID: PMC11249148 DOI: 10.1080/22221751.2024.2373307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/06/2024] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
SARS-CoV-2 has been evolving into a large number of variants, including the highly pathogenic Delta variant, and the currently prevalent Omicron subvariants with extensive evasion capability, which raises an urgent need to develop new broad-spectrum neutralizing antibodies. Herein, we engineer two IgG-(scFv)2 form bispecific antibodies with overlapping epitopes (bsAb1) or non-overlapping epitopes (bsAb2). Both bsAbs are significantly superior to the parental monoclonal antibodies in terms of their antigen-binding and virus-neutralizing activities against all tested circulating SARS-CoV-2 variants including currently dominant JN.1. The bsAb1 can efficiently neutralize all variants insensitive to parental monoclonal antibodies or the cocktail with IC50 lower than 20 ng/mL, even slightly better than bsAb2. Furthermore, the cryo-EM structures of bsAb1 in complex with the Omicron spike protein revealed that bsAb1 with overlapping epitopes effectively locked the S protein, which accounts for its conserved neutralization against Omicron variants. The bispecific antibody strategy engineered from overlapping epitopes provides a novel solution for dealing with viral immune evasion.
Collapse
MESH Headings
- Antibodies, Bispecific/biosynthesis
- Antibodies, Bispecific/chemistry
- Antibodies, Bispecific/metabolism
- Antibodies, Bispecific/ultrastructure
- Protein Domains
- Epitopes/metabolism
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/metabolism
- Antibodies, Neutralizing/ultrastructure
- SARS-CoV-2/immunology
- Immune Evasion
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/chemistry
- Antibodies, Viral/metabolism
- Antibodies, Viral/ultrastructure
- Neutralization Tests
- Antigens, Viral/metabolism
- Cryoelectron Microscopy
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/metabolism
- Spike Glycoprotein, Coronavirus/ultrastructure
- Models, Molecular
- Protein Structure, Quaternary
- Mutation
- Humans
Collapse
Affiliation(s)
- Hancong Sun
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Lingyun Xia
- Center for Infectious Disease Research, Research Center for Industries of the Future, Zhejiang Key Laboratory of Structural Biology, School of Life Sciences, Westlake University, Institute of Biology, Westlake Institute for Advanced Study, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Jianhua Li
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Yuanyuan Zhang
- Center for Infectious Disease Research, Research Center for Industries of the Future, Zhejiang Key Laboratory of Structural Biology, School of Life Sciences, Westlake University, Institute of Biology, Westlake Institute for Advanced Study, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Guanying Zhang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Ping Huang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Xingxing Wang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Yue Cui
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Ting Fang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Pengfei Fan
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Qiang Zhou
- Center for Infectious Disease Research, Research Center for Industries of the Future, Zhejiang Key Laboratory of Structural Biology, School of Life Sciences, Westlake University, Institute of Biology, Westlake Institute for Advanced Study, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Xiangyang Chi
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Changming Yu
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
26
|
Erukainure OL, Muhammad A, Ravichandran R, Abarshi MM, Katsayal SB, Abubakar MB, Abiodun YU, Atolani O, Preissner R, Banerjee P. Targeting mutation sites in the omicron variant of SARS-CoV-2 as potential therapeutic strategy against COVID-19 by antiretroviral drugs. Toxicol Rep 2024; 13:101825. [PMID: 39654998 PMCID: PMC11626820 DOI: 10.1016/j.toxrep.2024.101825] [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: 08/20/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
The multiple mutation of the spike (S) protein of the Omicron SARS-CoV-2 variant is a major concern, as it has been implicated in the severity of COVID-19 and its complications. These mutations have been attributed to COVID-19-infected immune-compromised individuals, with HIV patients being suspected to top the list. The present study investigated the mutation of the S protein of the omicron variant in comparison to the Delta and Wuhan variants. It also investigated the molecular interactions of antiretroviral drugs (ARVd) vis-à-vis dolutegravir, lamivudine, tenofovir-disoproxilfumarate and lenacapavir with the initiation and termination codons of the mRNAs of the mutated proteins of the omicron variant using computational tools. The complete genome sequences of the respective S proteins for omicron (OM066778.1), Delta (OK091006.1) and Wuhan (NC 045512.2) SARS-CoV-2 variants were retrieved from the National Center for Biotechnology Information (NCBI) database. Evolutionary analysis revealed high trends of mutations in the S protein of the omicron SARS-CoV-2 variant compared to the delta and Wuhan variants coupled with 68 % homology. The sequences of the translation initiation sites (TISs), translation termination sites (TTSs), high mutation region-1 (HMR1) and high region mutation-2 (HMR2) mRNAs were retrieved from the full genome of the omicron variant S protein. Molecular docking analysis revealed strong molecular interactions of ARVd with TISs, TTSs, HMR1 and HMR2 of the S protein mRNA. These results indicate mutations in the S protein of the Omicron SARS-CoV-2 variant compared to the Delta and Wuhan variants. These mutation points may present new therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- Ochuko L. Erukainure
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein 2028, South Africa
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Rahul Ravichandran
- DiSTABiF, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, Caserta 81100, Italy
| | - Musa M. Abarshi
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Sanusi B. Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Murtala B. Abubakar
- Center for Advanced Medical Research and Training (CAMRET), Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Ya'qub U. Abiodun
- Center for Advanced Medical Research and Training (CAMRET), Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | | | - Robert Preissner
- Institute for Physiology, Charité and Science-IT– University Medicine Berlin, Berlin, Germany
| | - Priyanka Banerjee
- Institute for Physiology, Charité and Science-IT– University Medicine Berlin, Berlin, Germany
| |
Collapse
|
27
|
Huan X, Zhan J, Gao H. Research progress of spike protein mutation of SARS-CoV-2 mutant strain and antibody development. Front Immunol 2024; 15:1407149. [PMID: 39624100 PMCID: PMC11609190 DOI: 10.3389/fimmu.2024.1407149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/28/2024] [Indexed: 01/03/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a respiratory disease with a very high infectious rate caused by the Severe Acute Respiratory Syndrome Coronavirus-2(SARS-CoV-2). Because SARS-CoV-2 is easy to mutate, the continuous emergence of SARS-CoV-2 variant strains not only enhances the infectivity of the SARS-CoV-2 but also brings great obstacles to the treatment of COVID-19. Neutralizing antibodies have achieved good results in the clinical application of the novel coronavirus pneumonia, which can be used for pre-infection protection and treatment of novel coronavirus patients. This review makes a detailed introduction to the mutation characteristics of SARS-CoV-2, focusing on the molecular mechanism of mutation affecting the infectivity of SARS-CoV-2, and the impact of mutation on monoclonal antibody therapy, providing scientific reference for the prevention of SARS-CoV-2 variant strains and the research and development of antibody drugs.
Collapse
Affiliation(s)
| | | | - Hongwei Gao
- School of Life Science, Ludong University, Yantai, Shandong, China
| |
Collapse
|
28
|
Chang F, Wu Q, Hu Y, Pan Z, Liu YC, Li YZ, Bostina M, Liu W, Zhao P, Qu X, Li YP. Engineered bispecific antibodies with enhanced breadth and potency against SARS-CoV-2 variants and SARS-related coronaviruses. Med Microbiol Immunol 2024; 213:24. [PMID: 39520579 DOI: 10.1007/s00430-024-00809-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
The concern of COVID-19 persists due to the continuous emergence of variants and the potential spillover of animal coronaviruses. The broad-spectrum neutralizing antibodies play a pivotal role in the prevention and treatment of coronavirus (CoV) infections. Here, we constructed 18 bi-specific antibodies (bsAbs) using 9 antibodies isolated from COVID-19 convalescents and vaccinated individuals, designed as dual variable domain immunoglobulin (DVD-Ig). A bsAb 5-HI showed a high binding capability to the S1 subunit of spike and exhibited breadth and potency against pseudotyped SARS-CoV-2 variants of concerns (VOCs) and SARS-related-CoVs (SARSr-CoVs), with half maximal effective concentration (EC50) of 0.028-3.444 nM and 50% inhibitory concentration (IC50) of 0.008-0.800 nM. In addition, it retained neutralization potency against the peudotyped virus of recently prevalent JN.1 strain (IC50, 12.74 nM). We found that the parental antibodies showed weak or no binding to the receptor binding domain (RBD) of the SARS-CoV, EG.5.1, and JN.1. However, the 5-HI maintained the binding with RBD and prevented the binding between hACE2 and RBD (IC50 for the RBD of SARS-CoV, 1.067 nM; EG.5.1, 0.423 nM; JN.1, 0.223 nM). In neutralization assays with the authentic virus, we found that the 5-HI effectively neutralized Omicron variants XBB.1.5 (IC50, 0.308 nM), EG.5.1 (IC50, 0.129 nM), and JN.1 (IC50, 13.692 nM), while its parental antibodies showed weakened or no neutralization. Therefore, the 5-HI represents a promising candidate for further development in the treatment and prevention of ongoing evolved SARS-CoV-2 VOCs and other SARSr-CoVs that potentially emerge in the future.
Collapse
Affiliation(s)
- Fangfang Chang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qian Wu
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yabin Hu
- Translational Medicine Institute, Hengyang Medical School, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Zhendong Pan
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Yong-Chen Liu
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yue-Zhou Li
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Wenpei Liu
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, Hengyang, China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China.
| | - Xiaowang Qu
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, Hengyang, China.
| | - Yi-Ping Li
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
29
|
Dey S, Pahari P, Mukherjee S, Munro JB, Das DK. Conformational dynamics of SARS-CoV-2 Omicron spike trimers during fusion activation at single molecule resolution. Structure 2024; 32:1910-1925.e6. [PMID: 39366371 PMCID: PMC11560620 DOI: 10.1016/j.str.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/26/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron entry involves spike (S) glycoprotein-mediated fusion of viral and late endosomal membranes. Here, using single-molecule Förster resonance energy transfer (sm-FRET) imaging and biochemical measurements, we directly visualized conformational changes of individual spike trimers on the surface of SARS-CoV-2 Omicron pseudovirions during fusion activation. We observed that the S2 domain of the Omicron spike is a dynamic fusion machine. S2 reversibly interchanges between the pre-fusion conformation and two previously undescribed intermediate conformations. Acidic pH shifts the conformational equilibrium of S2 toward an intermediate conformation and promotes the membrane hemi-fusion reaction. Moreover, we captured conformational reversibility in the S2 domain, which suggests that spike can protect itself from pre-triggering. Furthermore, we determined that Ca2+ directly promotes the S2 conformational change from an intermediate conformation to post-fusion conformation. In the presence of a target membrane, low pH and Ca2+ stimulate the irreversible transition to S2 post-fusion state and promote membrane fusion.
Collapse
Affiliation(s)
- Shuvankar Dey
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Purba Pahari
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Srija Mukherjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - James B Munro
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dibyendu Kumar Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India; Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
30
|
An R, Yang H, Tang C, Li Q, Huang Q, Wang H, Wang J, Zhou Y, Yang Y, Chen H, Yu W, Li B, Wu D, Zhang Y, Luo F, Quan W, Xu J, Lin D, Liang X, Yan Y, Yuan L, Du X, Yuan Y, Li Y, Sun Q, Wang Y, Lu S. A protein vaccine of RBD integrated with immune evasion mutation shows broad protection against SARS-CoV-2. Signal Transduct Target Ther 2024; 9:301. [PMID: 39500906 PMCID: PMC11538548 DOI: 10.1038/s41392-024-02007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continue to emerge and evade immunity, resulting in breakthrough infections in vaccinated populations. There is an urgent need for the development of vaccines with broad protective effects. In this study, we selected hotspot mutations in the receptor-binding domain (RBD) that contribute to immune escape properties and integrated them into the original RBD protein to obtain a complex RBD protein (cRBD), and we found cRBDs have broad protective effects against SARS-CoV-2 variants. Three cRBDs were designed in our study. Compared with the BA.1 RBD protein, the cRBDs induced the production of higher levels of broader-spectrum neutralizing antibodies, suggesting stronger and broader protective efficacy. In viral challenge experiments, cRBDs were more effective than BA.1 RBD in attenuating lung pathologic injury. Among the three constructs, cRBD3 showed optimal broad-spectrum and protective effects and is a promising candidate for a broad-spectrum SARS-CoV-2 vaccine. In conclusion, immunization with cRBDs triggered immunity against a wide range of variants, including those that emerged after we had completed designing the cRBDs. This study preliminarily explores and validates the feasibility of incorporating hotspot mutations that contribute to immune evasion into the RBD to expand the activity spectrum of antigen-induced antibodies.
Collapse
Affiliation(s)
- Ran An
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Hao Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Cong Tang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Qianqian Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China
| | - Qing Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Haixuan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Junbin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yanan Zhou
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Hongyu Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Bai Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Daoju Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yong Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Fangyu Luo
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Wenqi Quan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Jingwen Xu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Dongdong Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Xiaoming Liang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yuhuan Yan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Longhai Yuan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Xuena Du
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yuxia Yuan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yanwen Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Yunnan Provincial Key Laboratory of Vector-borne Diseases Control and Research, Kunming, China.
| | - Youchun Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Yunnan Provincial Key Laboratory of Vector-borne Diseases Control and Research, Kunming, China.
| |
Collapse
|
31
|
Handrejk K, Schmitz KS, Veldhuis Kroeze EJB, van Dijk LLA, van Run P, Haagmans B, Moscona A, Porotto M, de Swart RL, de Vries RD, Rissmann M. Characterization of a SARS-CoV-2 Omicron BA.5 direct-contact transmission model in hamsters. NPJ VIRUSES 2024; 2:52. [PMID: 39512864 PMCID: PMC11537969 DOI: 10.1038/s44298-024-00061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024]
Abstract
As SARS-CoV-2 continues to evolve antigenically to escape vaccine- or infection-induced immunity, suitable animal models are needed to study novel interventions against viral variants. Syrian hamsters are often used because of their high susceptibility to SARS-CoV-2 and associated tissue damage in the respiratory tract. Here, we established a direct-contact transmission model for SARS-CoV-2 Omicron BA.5 in hamsters. First, we determined whether 103 or 104 TCID50 in a low-volume inoculum led to reproducible infection and viral shedding in male and female hamsters. Next, we determined the optimal co-housing timing and duration between donor and recipient hamsters required for consistent direct-contact transmission. Finally, we compared viral loads and histopathological lesions in the respiratory tissues of donor and recipient hamsters. Intranasal inoculation of hamsters with 103 TCID50 and 104 TCID50 Omicron BA.5 in 10 µl per nostril led to reproducible infection. Viral loads in the throat measured by RT-qPCR were comparable between male and female hamsters. Notably, the shedding of infectious virus was significantly higher in male hamsters. Compared to SARS-CoV-2 D614G, Omicron BA.5 infection reached lower viral loads, had a delayed peak of virus replication, and induced limited body weight loss. To ensure consistent direct-contact transmission from inoculated donor hamsters to naïve recipients, a co-housing duration of 24 h starting 20 h post-infection of the donors was optimal. We detected mild inflammation in the respiratory tract of donor and recipient hamsters, and viral loads were higher and peaked earlier in donor hamsters compared to recipient hamsters. Taken together, we developed a robust Omicron BA.5 direct-contact transmission model in hamsters, that provides a valuable tool to study novel interventions.
Collapse
Affiliation(s)
- Kim Handrejk
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | - Peter van Run
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Bart Haagmans
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Anne Moscona
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Center for Host–Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
| | - Matteo Porotto
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Center for Host–Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Rik L. de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Melanie Rissmann
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
32
|
Oliveira BR, Nehlmeier I, Kempf AM, Venugopalan V, Rehders M, Ceniza MEP, Cavalcanti PADTPV, Hoffmann M, Pöhlmann S, Brix K. Cytoskeletal β-tubulin and cysteine cathepsin L deregulation by SARS-CoV-2 spike protein interaction with the neuronal model cell line SH-SY5Y. Biochimie 2024; 226:49-61. [PMID: 38432290 DOI: 10.1016/j.biochi.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
SARS-CoV-2 mainly infects the respiratory tract but can also target other organs, including the central nervous system. While it was recently shown that cells of the blood-brain-barrier are permissive to SARS-CoV-2 infection in vitro, it remains debated whether neurons can be infected. In this study, we demonstrate that vesicular stomatitis virus particles pseudotyped with the spike protein of SARS-CoV-2 variants WT, Alpha, Delta and Omicron enter the neuronal model cell line SH-SY5Y. Cell biological analyses of the pseudo-virus treated cultures showed marked alterations in microtubules of SH-SY5Y cells. Because the changes in β-tubulin occurred in most cells, but only few were infected, we further asked whether interaction of the cells with spike protein might be sufficient to cause molecular and structural changes. For this, SH-SY5Y cells were incubated with trimeric spike proteins for time intervals of up to 24 h. CellProfiler™-based image analyses revealed changes in the intensities of microtubule staining in spike protein-incubated cells. Furthermore, expression of the spike protein-processing protease cathepsin L was found to be up-regulated by wild type, Alpha and Delta spike protein pseudotypes and cathepsin L was found to be secreted from spike protein-treated cells. We conclude that the mere interaction of the SARS-CoV-2 with neuronal cells can affect cellular architecture and proteolytic capacities. The molecular mechanisms underlying SARS-CoV-2 spike protein induced cytoskeletal changes in neuronal cells remain elusive and require future studies.
Collapse
Affiliation(s)
- Bernardo R Oliveira
- Constructor University, School of Science, Campus Ring 1, D-28759, Bremen, Germany
| | - Inga Nehlmeier
- Deutsches Primatenzentrum - Leibniz-Institut für Primatenforschung, Abteilung Infektionsbiologie, Kellnerweg 4, D-37077, Göttingen, Germany.
| | - Amy Madeleine Kempf
- Deutsches Primatenzentrum - Leibniz-Institut für Primatenforschung, Abteilung Infektionsbiologie, Kellnerweg 4, D-37077, Göttingen, Germany; Faculty of Biology and Psychology, Georg-August University Göttingen, Wilhelmsplatz 1, D-37073, Göttingen, Germany.
| | | | - Maren Rehders
- Constructor University, School of Science, Campus Ring 1, D-28759, Bremen, Germany.
| | - Marianne E P Ceniza
- Constructor University, School of Science, Campus Ring 1, D-28759, Bremen, Germany.
| | | | - Markus Hoffmann
- Deutsches Primatenzentrum - Leibniz-Institut für Primatenforschung, Abteilung Infektionsbiologie, Kellnerweg 4, D-37077, Göttingen, Germany; Faculty of Biology and Psychology, Georg-August University Göttingen, Wilhelmsplatz 1, D-37073, Göttingen, Germany.
| | - Stefan Pöhlmann
- Deutsches Primatenzentrum - Leibniz-Institut für Primatenforschung, Abteilung Infektionsbiologie, Kellnerweg 4, D-37077, Göttingen, Germany; Faculty of Biology and Psychology, Georg-August University Göttingen, Wilhelmsplatz 1, D-37073, Göttingen, Germany.
| | - Klaudia Brix
- Constructor University, School of Science, Campus Ring 1, D-28759, Bremen, Germany.
| |
Collapse
|
33
|
Yang YF, Lin YJ, You SH, Lu TH, Chen CY, Wang WM, Ling MP, Chen SC, Liao CM. A Regional-Scale Assessment-Based SARS-CoV-2 Variants Control Modeling with Implications for Infection Risk Characterization. Infect Drug Resist 2024; 17:4791-4805. [PMID: 39498414 PMCID: PMC11533883 DOI: 10.2147/idr.s480086] [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: 05/27/2024] [Accepted: 10/25/2024] [Indexed: 11/07/2024] Open
Abstract
Background The emergence and progression of highly divergent SARS-CoV-2 variants have posed increased risks to global public health, triggering the significant impacts on countermeasures since 2020. However, in addition to vaccination, the effectiveness of non-pharmaceutical interventions, such as social distancing, masking, or hand washing, on different variants of concern (VOC) remains largely unknown. Objective This study provides a mechanistic approach by implementing a control measure model and a risk assessment framework to quantify the impacts of control measure combinations on the transmissions of five VOC (Alpha, Beta, Delta, Gamma, and Omicron), along with a different perspective of risk assessment application. Materials and Methods We applied uncontrollable ratios as an indicator by adopting basic reproduction number (R 0) data collected from a regional-scale survey. A risk assessment strategy was established by constructing VOC-specific dose-response profiles to implicate practical uses in risk characterization when exposure data are available. Results We found that social distancing alone was ineffective without vaccination in almost all countries and VOC when the median R 0 was greater than two. Our results indicated that Omicron could not be contained, even when all control measure combinations were applied, due to its low threshold of infectivity (~3×10-4 plague-forming unit (PFU) mL-1). Conclusion To facilitate better decision-making in future interventions, we provide a comprehensive evaluation of how combined control measures impact on different countries and various VOC. Our findings indicate the potential application of threshold estimates of infectivity in the context of risk communication and policymaking for controlling future emerging SARS-CoV-2 variant infections.
Collapse
Affiliation(s)
- Ying-Fei Yang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 10617, Taiwan
| | - Yi-Jun Lin
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, 11230, Taiwan
| | - Shu-Han You
- Institute of Food Safety and Risk Management, National Taiwan Ocean University, Keelung City, 20224, Taiwan
| | - Tien-Hsuan Lu
- Department of Science Education and Application, National Taichung University of Education, Taichung, 403514, Taiwan
| | - Chi-Yun Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32608, USA
| | - Wei-Min Wang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 10617, Taiwan
| | - Min-Pei Ling
- Department of Food Science, National Taiwan Ocean University, Keelung City, 20224, Taiwan
| | - Szu-Chieh Chen
- Department of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
- Department of Family and Community Medicine, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Chung-Min Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
34
|
Chen N, Decker KE, Schulz SR, Kempf A, Nehlmeier I, Moldenhauer AS, Dopfer-Jablonka A, Behrens GMN, Stankov MV, Manthey L, Jäck HM, Hoffmann M, Pöhlmann S, Arora P. Comparative Analysis of Host Cell Entry Efficiency and Neutralization Sensitivity of Emerging SARS-CoV-2 Lineages KP.2, KP.2.3, KP.3, and LB.1. Vaccines (Basel) 2024; 12:1236. [PMID: 39591139 PMCID: PMC11598761 DOI: 10.3390/vaccines12111236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
New SARS-CoV-2 lineages continue to evolve and may exhibit new characteristics regarding host cell entry efficiency and potential for antibody evasion. Here, employing pseudotyped particles, we compared the host cell entry efficiency, ACE2 receptor usage, and sensitivity to antibody-mediated neutralization of four emerging SARS-CoV-2 lineages, KP.2, KP.2.3, KP.3, and LB.1. The XBB.1.5 and JN.1 lineages served as controls. Our findings reveal that KP.2, KP.2.3, KP.3, and LB.1 lineages enter host cells efficiently and in an ACE2-dependent manner, and that KP.3 is more adept at entering Calu-3 lung cells than JN.1. However, the variants differed in their capacity to employ ACE2 orthologues from animal species for entry, suggesting differences in ACE2 interactions. Moreover, we demonstrate that only two out of seven therapeutic monoclonal antibody (mAbs) in preclinical development retain robust neutralizing activity against the emerging JN.1 sublineages tested, while three mAbs displayed strongly reduced neutralizing activity and two mAbs lacked neutralizing activity against any of the lineages tested. Furthermore, our results show that KP.2, KP.2.3, KP.3, and LB.1 lineages evade neutralization by antibodies induced by infection or vaccination with greater efficiency than JN.1, particularly in individuals without hybrid immunity. This study indicates that KP.2, KP.2.3, KP.3, and LB.1 differ in ACE2 interactions and the efficiency of lung cell entry and suggest that evasion of neutralizing antibodies drove the emergence of these variants.
Collapse
Affiliation(s)
- Nianzhen Chen
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Katharina Emma Decker
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Sebastian R. Schulz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany; (S.R.S.); (H.-M.J.)
| | - Amy Kempf
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
| | - Anna-Sophie Moldenhauer
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
| | - Alexandra Dopfer-Jablonka
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany
| | - Georg M. N. Behrens
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany
- Center for Individualized Infection Medicine (CiiM), 30625 Hannover, Germany
| | - Metodi V. Stankov
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
| | - Luis Manthey
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany; (S.R.S.); (H.-M.J.)
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Prerna Arora
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
35
|
Bozkus CC, Brown M, Velazquez L, Thomas M, Wilson EA, O’Donnell T, Ruchnewitz D, Geertz D, Bykov Y, Kodysh J, Oguntuyo KY, Roudko V, Hoyos D, Srivastava KD, Kleiner G, Alshammary H, Karekar N, McClain C, Gopal R, Nie K, Del Valle D, Delbeau-Zagelbaum D, Rodriguez D, Setal J, The Mount Sinai COVID-19 Biobank Team, Carroll E, Wiesendanger M, Gulko PS, Charney A, Merad M, Kim-Schulze S, Lee B, Wajnberg A, Simon V, Greenbaum BD, Chowell D, Vabret N, Luksza M, Bhardwaj N. T cell epitope mapping reveals immunodominance of evolutionarily conserved regions within SARS-CoV-2 proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619918. [PMID: 39484455 PMCID: PMC11527131 DOI: 10.1101/2024.10.23.619918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
As SARS-CoV-2 variants continue to emerge capable of evading neutralizing antibodies, it has become increasingly important to fully understand the breadth and functional profile of T cell responses to determine their impact on the immune surveillance of variant strains. Here, sampling healthy individuals, we profiled the kinetics and polyfunctionality of T cell immunity elicited by mRNA vaccination. Modeling of anti-spike T cell responses against ancestral and variant strains of SARS-CoV-2 suggested that epitope immunodominance and cross-reactivity are major predictive determinants of T cell immunity. To identify immunodominant epitopes across the viral proteome, we generated a comprehensive map of CD4+ and CD8+ T cell epitopes within non-spike proteins that induced polyfunctional T cell responses in convalescent patients. We found that immunodominant epitopes mainly resided within regions that were minimally disrupted by mutations in emerging variants. Conservation analysis across historical human coronaviruses combined with in silico alanine scanning mutagenesis of non-spike proteins underscored the functional importance of mutationally-constrained immunodominant regions. Collectively, these findings identify immunodominant T cell epitopes across the mutationally-constrained SARS-CoV-2 proteome, potentially providing immune surveillance against emerging variants, and inform the design of next-generation vaccines targeting antigens throughout SARS-CoV-2 proteome for broader and more durable protection.
Collapse
Affiliation(s)
- Cansu Cimen Bozkus
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
| | - Matthew Brown
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leandra Velazquez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcus Thomas
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric A. Wilson
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy O’Donnell
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Denis Ruchnewitz
- Institute for Biological Physics, University of Cologne, 50937 Cologne, Germany
| | - Douglas Geertz
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yonina Bykov
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Kodysh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kasopefoluwa Y. Oguntuyo
- The Department of Medicine, The Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vladimir Roudko
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Hoyos
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Komal D. Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulio Kleiner
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hala Alshammary
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neha Karekar
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher McClain
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ramya Gopal
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Del Valle
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Denise Rodriguez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Setal
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Emily Carroll
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margrit Wiesendanger
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Percio S. Gulko
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ania Wajnberg
- The Department of Medicine, The Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), 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
- The Department of Medicine, The Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin D Greenbaum
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Diego Chowell
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicolas Vabret
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta Luksza
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
36
|
Declercq J, Gerlo S, Van Nevel S, De Ruyck N, Holtappels G, Delesie L, Tobback E, Lammens I, Gerebtsov N, Sedeyn K, Saelens X, Lambrecht BN, Gevaert P, Vandekerckhove L, Vanhee S. Repeated COVID-19 mRNA-based vaccination contributes to SARS-CoV-2 neutralizing antibody responses in the mucosa. Sci Transl Med 2024; 16:eadn2364. [PMID: 39441904 DOI: 10.1126/scitranslmed.adn2364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
To prevent infection by respiratory viruses and consequently limit virus circulation, vaccines need to promote mucosal immunity. The extent to which the currently used messenger RNA (mRNA)-based COVID-19 vaccines induce mucosal immunity remains poorly characterized. We evaluated mucosal neutralizing antibody responses in a cohort of 183 individuals. Participants were sampled at several time points after primary adenovirus vector-based or mRNA-based COVID-19 vaccination and after mRNA-based booster vaccinations. Our findings revealed that repeated vaccination with mRNA boosters promoted severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies in nasal secretions. Nasal and serum neutralizing antibody titers of both IgG and IgA isotypes correlated to one another. We investigated the source of these mucosal antibodies in a mouse model wherein mice received repeated mRNA vaccines for SARS-CoV-2. These experiments indicated that neutralizing antibody-producing cells reside in the spleen and bone marrow, whereas no proof of tissue homing to the respiratory mucosa was observed, despite the detection of mucosal antibodies. Serum transfer experiments confirmed that circulating antibodies were able to migrate to the respiratory mucosa. Collectively, these results demonstrate that, especially upon repeated vaccination, the currently used COVID-19 mRNA vaccines can elicit mucosal neutralizing antibodies and that vaccination might also stimulate mucosal immunity induced by previous SARS-CoV-2 infection. Moreover, migration of circulating antibodies to the respiratory mucosa might be a main mechanism. These findings advance our understanding of mRNA vaccine-induced immunity and have implications for the design of vaccine strategies to combat respiratory infections.
Collapse
Affiliation(s)
- Jozefien Declercq
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sarah Gerlo
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Sharon Van Nevel
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Natalie De Ruyck
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Gabriele Holtappels
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Liesbeth Delesie
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Els Tobback
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Inés Lammens
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Nikita Gerebtsov
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Koen Sedeyn
- VIB Center for Medical Biotechnology, 9000 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Xavier Saelens
- VIB Center for Medical Biotechnology, 9000 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus Medical Center, 3015 GD Rotterdam, Netherlands
| | - Philippe Gevaert
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Linos Vandekerckhove
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Stijn Vanhee
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
37
|
Liu Y, Zhao X, Shi J, Wang Y, Liu H, Hu YF, Hu B, Shuai H, Yuen TTT, Chai Y, Liu F, Gong HR, Li J, Wang X, Jiang S, Zhang X, Zhang Y, Li X, Wang L, Hartnoll M, Zhu T, Hou Y, Huang X, Yoon C, Wang Y, He Y, Zhou M, Du L, Zhang X, Chan WM, Chen LL, Cai JP, Yuan S, Zhou J, Huang JD, Yuen KY, To KKW, Chan JFW, Zhang BZ, Sun L, Wang P, Chu H. Lineage-specific pathogenicity, immune evasion, and virological features of SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3. Nat Commun 2024; 15:8728. [PMID: 39379369 PMCID: PMC11461813 DOI: 10.1038/s41467-024-53033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
SARS-CoV-2 JN.1 with an additional L455S mutation on spike when compared with its parental variant BA.2.86 has outcompeted all earlier variants to become the dominant circulating variant. Recent studies investigated the immune resistance of SARS-CoV-2 JN.1 but additional factors are speculated to contribute to its global dominance, which remain elusive until today. Here, we find that SARS-CoV-2 JN.1 has a higher infectivity than BA.2.86 in differentiated primary human nasal epithelial cells (hNECs). Mechanistically, we demonstrate that the gained infectivity of SARS-CoV-2 JN.1 over BA.2.86 associates with increased entry efficiency conferred by L455S and better spike cleavage in hNECs. Structurally, S455 altered the mode of binding of JN.1 spike protein to ACE2 when compared to BA.2.86 spike at ACE2H34, and modified the internal structure of JN.1 spike protein by increasing the number of hydrogen bonds with neighboring residues. These findings indicate that a single mutation (L455S) enhances virus entry in hNECs and increases immune evasiveness, which contribute to the robust transmissibility of SARS-CoV-2 JN.1. We further evaluate the in vitro and in vivo virological characteristics between SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3, and identify key lineage-specific features of the two Omicron sublineages that contribute to our understanding on Omicron antigenicity, transmissibility, and pathogenicity.
Collapse
Affiliation(s)
- Yuanchen Liu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Xiaoyu Zhao
- Shanghai Sci-Tech Inno Center for Infection & Immunity, National Medical Center for Infectious Diseases, Huashan Hospital, Institute of Infection and Health, Fudan University, Shanghai, China
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jialu Shi
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yajie Wang
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Huan Liu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Ye-Fan Hu
- BayVax Biotech Limited, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, China
| | - Bingjie Hu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Huiping Shuai
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Terrence Tsz-Tai Yuen
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Yue Chai
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Feifei Liu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Hua-Rui Gong
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiayan Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Xun Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Shujun Jiang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiang Zhang
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yanliang Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiangnan Li
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, China
| | - Lei Wang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Madeline Hartnoll
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Tianrenzheng Zhu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Yuxin Hou
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Xiner Huang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Chaemin Yoon
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yang Wang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yixin He
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Minmin Zhou
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Lianzhao Du
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Xiaojuan Zhang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wan-Mui Chan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Lin-Lei Chen
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jian-Piao Cai
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Jie Zhou
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Jian-Dong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Haikou, Hainan Province, China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
- Guangzhou Laboratory, Guangzhou, Guangdong Province, China
| | - Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
- Guangzhou Laboratory, Guangzhou, Guangdong Province, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China.
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China.
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Haikou, Hainan Province, China.
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China.
- Guangzhou Laboratory, Guangzhou, Guangdong Province, China.
| | - Bao-Zhong Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Lei Sun
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China.
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China.
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, China.
| |
Collapse
|
38
|
Tang Y, Tang K, Hu Y, Ye ZW, Luo W, Luo C, Cao H, Wang R, Yue X, Liu D, Liu C, Ge X, Liu T, Chen Y, Yuan S, Deng L. M protein ectodomain-specific immunity restrains SARS-CoV-2 variants replication. Front Immunol 2024; 15:1450114. [PMID: 39416782 PMCID: PMC11480003 DOI: 10.3389/fimmu.2024.1450114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction The frequent occurrence of mutations in the SARS-CoV-2 Spike (S) protein, with up to dozens of mutations, poses a severe threat to the current efficacy of authorized COVID-19 vaccines. Membrane (M) protein, which is the most abundant viral structural protein, exhibits a high level of amino acid sequence conservation. M protein ectodomain could be recognized by specific antibodies; however, the extent to which it is immunogenic and provides protection remains unclear. Methods We designed and synthesized multiple peptides derived from coronavirus M protein ectodomains, and determined the secondary structure of specific peptides using circular dichroism (CD) spectroscopy. Enzyme-linked immunosorbent assay (ELISA) was utilized to detect IgG responses against the synthesized peptides in clinical samples. To evaluate the immunogenicity of peptide vaccines, BALB/c mice were intraperitoneally immunized with peptide-keyhole limpet hemocyanin (KLH) conjugates adjuvanted with incomplete Freund's adjuvant (IFA). The humoral and T-cell immune responses induced by peptide-KLH conjugates were assessed using ELISA and ELISpot assays, respectively. The efficacy of the S2M2-30-KLH vaccine against SARS-CoV-2 variants was evaluated in vivo using the K18-hACE2 transgenic mouse model. The inhibitory effect of mouse immune serum on SARS-CoV-2 virus replication in vitro was evaluated using microneutralization assays. The subcellular localization of the M protein was evaluated using an immunofluorescent staining method, and the Fc-mediated antibody-dependent cellular cytotoxicity (ADCC) activity of the S2M2-30-specific monoclonal antibody (mAb) was measured using an ADCC reporter assay. Results Seroconversion rates for ectodomain-specific IgG were observed to be high in both SARS-CoV-2 convalescent patients and individuals immunized with inactivated vaccines. To assess the protective efficacy of the M protein ectodomain-based vaccine, we initially identified a highly immunogenic peptide derived from this ectodomain, named S2M2-30. The mouse serum specific to S2M2-30 showed inhibitory effects on the replication of SARS-CoV-2 variants in vitro. Immunizations of K18-hACE2-transgenic mice with the S2M2-30-keyhole limpet hemocyanin (KLH) vaccine significantly reduced the lung viral load caused by B.1.1.7/Alpha (UK) infection. Further mechanism investigations reveal that serum neutralizing activity, specific T-cell response and Fc-mediated antibody-dependent cellular cytotoxicity (ADCC) correlate with the specific immuno-protection conferred by S2M2-30. Discussion The findings of this study suggest that the antibody responses against M protein ectodomain in the population most likely exert a beneficial effect on preventing various SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Yibo Tang
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Kaiming Tang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yunqi Hu
- School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Zi-Wei Ye
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wanyu Luo
- School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Cuiting Luo
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hehe Cao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ran Wang
- Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xinyu Yue
- School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Dejian Liu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Cuicui Liu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Xingyi Ge
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Tianlong Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yaoqing Chen
- School of Public Health, Sun Yat-sen University, Shenzhen, China
- National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Sun Yat-sen University, Guangzhou, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Lei Deng
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
- Research and Development Department, Beijing Weimiao Biotechnology Co. Ltd., Beijing, China
| |
Collapse
|
39
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
40
|
Murayama G, Kusaoi M, Horiuchi Y, Tabe Y, Naito T, Ito S, Yamaji K, Tamura N. Effects of the induction of humoral and cellular immunity by third vaccination for SARS-CoV-2. J Infect Chemother 2024; 30:1021-1027. [PMID: 38570139 DOI: 10.1016/j.jiac.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/08/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION To control the spread of severe disease caused by mutant strains of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), it is necessary to determine whether continued vaccination enhances humoral and cellular immunity. AIM In this study, we examined the changes in humoral and cellular immunity to SARS-CoV-2 after administration of the third vaccination in Japanese adults who had received the second dose of messenger ribonucleic acid (mRNA)-1273 vaccine and the third vaccination (BNT162b2 or mRNA-1273). METHODS We measured anti-spike antibodies in immunoglobulin G (IgG) and anti-nucleocapsid IgG titers in the serum of the vaccinated subjects. To evaluate cellular immunity, the peripheral blood mononuclear cells of inoculated individuals were cultured with spiked proteins, including those of the SARS-CoV-2 conventional strain and Omicron strain, and then subjected to enzyme-linked immunospot (ELISPOT). RESULTS The results revealed that the anti-SARS-CoV-2 spike protein antibody titer increased after the third vaccination and was maintained; however, a decrease was observed at 6 months after vaccination. SARS-CoV-2 antigen-specific T helper (Th)1 and Th2 cell responses were also induced after the third vaccination and were maintained for 6 months after vaccination. Furthermore, induction of cellular immunity against Omicron strains by the omicron non-compliant vaccines, BNT162b2 or mRNA-1273, was observed. CONCLUSION These findings demonstrate the effectiveness of vaccination against unknown mutant strains that may occur in the future and provide important insights into vaccination strategies.
Collapse
Affiliation(s)
- Goh Murayama
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan.
| | - Makio Kusaoi
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Yuki Horiuchi
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, 113-8421, Japan
| | - Suminobu Ito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, 113-8421, Japan; Medical Technology Innovation Centre, Juntendo University, Tokyo, 113-8421, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| |
Collapse
|
41
|
Sarkar M, Madabhavi I. COVID-19 mutations: An overview. World J Methodol 2024; 14:89761. [PMID: 39310238 PMCID: PMC11230071 DOI: 10.5662/wjm.v14.i3.89761] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/07/2024] [Accepted: 04/17/2024] [Indexed: 06/25/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) belongs to the genus Beta coronavirus and the family of Coronaviridae. It is a positive-sense, non-segmented single-strand RNA virus. Four common types of human coronaviruses circulate globally, particularly in the fall and winter seasons. They are responsible for 10%-30% of all mild upper respiratory tract infections in adults. These are 229E, NL63 of the Alfacoronaviridae family, OC43, and HKU1 of the Betacoronaviridae family. However, there are three highly pathogenic human coronaviruses: SARS-CoV-2, Middle East respiratory syndrome coronavirus, and the latest pandemic caused by the SARS-CoV-2 infection. All viruses, including SARS-CoV-2, have the inherent tendency to evolve. SARS-CoV-2 is still evolving in humans. Additionally, due to the development of herd immunity, prior infection, use of medication, vaccination, and antibodies, the viruses are facing immune pressure. During the replication process and due to immune pressure, the virus may undergo mutations. Several SARS-CoV-2 variants, including the variants of concern (VOCs), such as B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617/B.1.617.2 (Delta), P.1 (Gamma), and B.1.1.529 (Omicron) have been reported from various parts of the world. These VOCs contain several important mutations; some of them are on the spike proteins. These mutations may lead to enhanced infectivity, transmissibility, and decreased neutralization efficacy by monoclonal antibodies, convalescent sera, or vaccines. Mutations may also lead to a failure of detection by molecular diagnostic tests, leading to a delayed diagnosis, increased community spread, and delayed treatment. We searched PubMed, EMBASE, Covariant, the Stanford variant Database, and the CINAHL from December 2019 to February 2023 using the following search terms: VOC, SARS-CoV-2, Omicron, mutations in SARS-CoV-2, etc. This review discusses the various mutations and their impact on infectivity, transmissibility, and neutralization efficacy.
Collapse
Affiliation(s)
- Malay Sarkar
- Department of Pulmonary Medicine, Indira Gandhi Medical College, Shimla 171001, Himachal Pradesh, India
| | - Irappa Madabhavi
- Department of Medical and Pediatric Oncology and Hematology, J N Medical College, and KAHER, Belagavi, Karnataka 590010, India
- Department of Medical and Pediatric Oncology and Hematology, Kerudi Cancer Hospital, Bagalkot, Karnataka 587103, India
| |
Collapse
|
42
|
Dhankher S, Yadav P, Sharma S, Gupta E, Yadav RG, Dash PK, Parida M. Structural and genomic evolutionary dynamics of Omicron variant of SARS-CoV-2 circulating in Madhya Pradesh, India. Front Med (Lausanne) 2024; 11:1416006. [PMID: 39323472 PMCID: PMC11422100 DOI: 10.3389/fmed.2024.1416006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/06/2024] [Indexed: 09/27/2024] Open
Abstract
The SARS-CoV-2 Omicron (B.1.1.529) variant emerged in early November 2021 and its rapid spread created fear worldwide. This was attributed to its increased infectivity and escaping immune mechanisms. The spike protein of Omicron has more mutations (>30) than any other previous variants and was declared as the variant of concern (VOC) by the WHO. The concern among the scientific community was huge about this variant, and a piece of updated information on circulating viral strains is important in order to better understand the epidemiology, virus pathogenicity, transmission, therapeutic interventions, and vaccine development. A total of 710 samples were processed for sequencing and identification up to a resolution of sub-lineage. The sequence analysis revealed Omicron variant with distribution as follows: B.1.1, B.1.1.529, BA.1, BA.2, BA.2.10, BA.2.10.1, BA.2.23, BA.2.37, BA.2.38, BA.2.43, BA.2.74, BA.2.75, BA.2.76, and BA.4 sub-lineages. There is a shift noted in circulating lineage from BA.1 to BA.2 to BA.4 over a period from January to September 2022. Multiple signature mutations were identified in S protein T376A, D405N, and R408S mutations, which were new and common to all BA.2 variants. Additionally, R346T was seen in emerging BA.2.74 and BA.2.76 variants. The emerging BA.4 retained the common T376A, D405N, and R408S mutations of BA.2 along with a new mutation F486V. The samples sequenced were from different districts of Madhya Pradesh and showed a predominance of BA.2 and its variants circulating in this region. The current study identified circulation of BA.1 and BA.1.1 variants during initial phase. The predominant Delta strain of the second wave has been replaced by the Omicron variant in this region over a period of time. This study successfully deciphers the dynamics of the emergence and replacement of various sub-lineages of SARS-CoV-2 in central India on real real-time basis.
Collapse
Affiliation(s)
| | | | | | | | | | - Paban Kumar Dash
- Virology Division, Defence Research and Development Establishment, Gwalior, India
| | | |
Collapse
|
43
|
Ye G, Bu F, Pan R, Mendoza A, Yang G, Spiller B, Wadzinski BE, Du L, Perlman S, Liu B, Li F. Structure-guided in vitro evolution of nanobodies targeting new viral variants. PLoS Pathog 2024; 20:e1012600. [PMID: 39325826 PMCID: PMC11460708 DOI: 10.1371/journal.ppat.1012600] [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: 05/08/2024] [Revised: 10/08/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
A major challenge in antiviral antibody therapy is keeping up with the rapid evolution of viruses. Our research shows that nanobodies - single-domain antibodies derived from camelids - can be rapidly re-engineered to combat new viral strains through structure-guided in vitro evolution. Specifically, for viral mutations occurring at nanobody-binding sites, we introduce randomized amino acid sequences into nanobody residues near these mutations. We then select nanobody variants that effectively bind to the mutated viral target from a phage display library. As a proof of concept, we used this approach to adapt Nanosota-3, a nanobody originally identified to target the receptor-binding domain (RBD) of early Omicron subvariants, making it highly effective against recent Omicron subvariants. Remarkably, this adaptation process can be completed in less than two weeks, allowing drug development to keep pace with viral evolution and provide timely protection to humans.
Collapse
Affiliation(s)
- Gang Ye
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Fan Bu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ruangang Pan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Alise Mendoza
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ge Yang
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Benjamin Spiller
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Brian E. Wadzinski
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Bin Liu
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
44
|
Koolaparambil Mukesh R, Yinda CK, Munster VJ, van Doremalen N. Beyond COVID-19: the promise of next-generation coronavirus vaccines. NPJ VIRUSES 2024; 2:39. [PMID: 40295763 PMCID: PMC11721646 DOI: 10.1038/s44298-024-00043-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/26/2024] [Indexed: 04/30/2025]
Abstract
Coronaviruses (CoVs) have caused three global outbreaks: severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) in 2003, Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012, and SARS-CoV-2 in 2019, with significant mortality and morbidity. The impact of coronavirus disease 2019 (COVID-19) raised serious concerns about the global preparedness for a pandemic. Furthermore, the changing antigenic landscape of SARS-CoV-2 led to new variants with increased transmissibility and immune evasion. Thus, the development of broad-spectrum vaccines against current and future emerging variants of CoVs will be an essential tool in pandemic preparedness. Distinct phylogenetic features within CoVs complicate and limit the process of generating a pan-CoV vaccine capable of targeting the entire Coronaviridae family. In this review, we aim to provide a detailed overview of the features of CoVs, their phylogeny, current vaccines against various CoVs, the efforts in developing broad-spectrum coronavirus vaccines, and the future.
Collapse
Affiliation(s)
| | - Claude K Yinda
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, MT, USA
| | - Vincent J Munster
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, MT, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
45
|
Han X, Pan H, Jin P, Wei M, Jia S, Wang W, Chu K, Gao S, Zhou L, Li J, Zhu F. A head-to-head comparison of humoral and cellular immune responses of five COVID-19 vaccines in adults in China. Front Immunol 2024; 15:1455730. [PMID: 39234239 PMCID: PMC11371563 DOI: 10.3389/fimmu.2024.1455730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/25/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Various COVID-19 vaccine trials have shown that vaccines can successfully prevent symptomatic cases of COVID-19 and death. Head-to-head comparisons help to better understand the immune response characteristics of different COVID-19 vaccines in humans. Methods We randomly selected 20 participants from each of five ongoing Phase II trials of COVID-19 vaccines. Here, SARS-CoV 2-specific immune responses to DNA vaccine (INO-4800), mRNA vaccine (BNT162b2), Adenovirus-vectored vaccine (CONVIDECIA), Protein subunit vaccine (Recombinant COVID- 19 Vaccine (Sf9 Cells)), Inactivated Vaccine (KCONVAC) were examined longitudinally in healthy adults between Jan 15, 2021 and July 5, 2021 for 6 months. RBD-IgG titres were detected by ELISA, neutralising antibody titer were detected by pseudoviral neutralization and immune cell response were detected by flow cytometry. Results At the first visit (V1), 100% of individuals who received the BNT162b2, CONVIDECIA, or KCONVAC vaccines experienced seroconversion of neutralizing and binding antibodies in the serum. Except for the Recombinant COVID-19 Vaccine (Sf9 Cells) vaccine having the highest neutralizing antibody GMT at the second visit (although there was no statistically significant difference in geometric mean titers between V1 and V2), the rest of the vaccines had the highest levels of binding antibodies and neutralizing antibodies at V1. The neutralizing antibodies GMT of all vaccines showed a significant decrease at V3 compared to V1. The neutralizing antibody GMT against the omicron variant of all vaccines at V1 showed a significant decrease compared to the wild strain. We observed statistically significant differences in Tcm cells and RBD-specific memory B cells among various vaccines. Discussion BNT162b2 (mRNA vaccine) exhibits the highest antibody levels among the five vaccines evaluated, regardless of whether the target is the wild-type virus or its variants. However, its cellular immune response may be weaker compared to CONVIDECIA (adenovirus type 5 vector vaccine).
Collapse
MESH Headings
- Humans
- COVID-19 Vaccines/immunology
- Adult
- COVID-19/immunology
- COVID-19/prevention & control
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Immunity, Humoral
- SARS-CoV-2/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Male
- Female
- Immunity, Cellular
- China
- Middle Aged
- Young Adult
- Vaccines, Subunit/immunology
- Vaccines, DNA/immunology
- BNT162 Vaccine/immunology
- Immunogenicity, Vaccine
- Vaccines, Inactivated/immunology
Collapse
Affiliation(s)
- Xu Han
- National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongxing Pan
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Pengfei Jin
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Mingwei Wei
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Siyue Jia
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Wenjuan Wang
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Kai Chu
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Shuyu Gao
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Li Zhou
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jingxin Li
- National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, China
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Fengcai Zhu
- National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, China
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| |
Collapse
|
46
|
Kotaki R, Moriyama S, Oishi S, Onodera T, Adachi Y, Sasaki E, Ishino K, Morikawa M, Takei H, Takahashi H, Takano T, Nishiyama A, Yumoto K, Terahara K, Isogawa M, Matsumura T, Shinkai M, Takahashi Y. Repeated Omicron exposures redirect SARS-CoV-2-specific memory B cell evolution toward the latest variants. Sci Transl Med 2024; 16:eadp9927. [PMID: 39167666 DOI: 10.1126/scitranslmed.adp9927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Immunological imprinting by ancestral SARS-CoV-2 strains is thought to impede the robust induction of Omicron-specific humoral responses by Omicron-based booster vaccines. Here, we analyzed the specificity and neutralization activity of memory B (Bmem) cells after repeated BA.5 exposure in individuals previously imprinted by ancestral strain-based mRNA vaccines. After a second BA.5 exposure, Bmem cells with BA.5 spike protein-skewed reactivity were promptly elicited, correlating with preexisting antibody titers. Clonal lineage analysis identified BA.5-skewed Bmem cells that had redirected their specificity from the ancestral strain to BA.5 through somatic hypermutations. Moreover, Bmem cells with redirected BA.5 specificity exhibited accelerated development compared with de novo Bmem cells derived from naïve repertoires. This redirected BA.5 specificity demonstrated greater resilience to viral point mutation and adaptation to recent Omicron variants HK.3 and JN.1, months after the second BA.5 exposure, suggesting that existing Bmem cells elicited by older vaccines can redirect their specificity toward newly evolving variants.
Collapse
Affiliation(s)
- Ryutaro Kotaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Shintaro Oishi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Eita Sasaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kota Ishino
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | | | | | - Tomohiro Takano
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Ayae Nishiyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kohei Yumoto
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kazutaka Terahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Masanori Isogawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido 001-0021, Japan
| |
Collapse
|
47
|
Goh YS, Fong SW, Hor PX, Loh CY, Wang B, Salleh SNM, Ngoh EZX, Lee RTC, Poh XY, Rao S, Chia PY, Ong SWX, Lee TH, Lim C, Teo J, Pada S, Sun LJ, Ong DLS, Somani J, Lee ES, Maurer-Stroh S, Wang CI, Leo YS, Lye DC, Young BE, Ng LFP, Renia L. Variant-Specific IgA Protects Against Omicron Infection. J Infect Dis 2024; 230:e287-e291. [PMID: 37996071 PMCID: PMC11326848 DOI: 10.1093/infdis/jiad525] [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: 09/01/2023] [Revised: 10/25/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The emergence of rapidly evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, coupled with waning vaccine-induced immunity, has contributed to the rise of vaccine breakthrough infections. It is crucial to understand how vaccine-induced protection is mediated. METHODS We examined 2 prospective cohorts of mRNA vaccinated and boosted individuals during the Omicron wave of infection in Singapore. RESULTS We found that individuals who remain uninfected over the follow-up period had a higher variant-specific IgA, but not IgG, antibody response at 1 month after booster vaccination, compared with individuals who became infected. CONCLUSIONS We conclude that IgA may have a potential contributory role in protection against Omicron infection. Clinical Trials Registration . NCT05142319.
Collapse
Affiliation(s)
- Yun Shan Goh
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Siew-Wai Fong
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Pei Xiang Hor
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chiew Yee Loh
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Bei Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Eve Zi Xian Ngoh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Raphael Tze Chuen Lee
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
- GISAID Global Data Science Initiative, Munich, Germany
| | - Xuan Ying Poh
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Suma Rao
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Po Ying Chia
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Sean W X Ong
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tau Hong Lee
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Clarissa Lim
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Jefanie Teo
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Surinder Pada
- Division of Infectious Diseases, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Louisa Jin Sun
- Infectious Diseases, Alexandra Hospital, Singapore, Singapore
| | | | - Jyoti Somani
- Division of Infectious Diseases, Department of Medicine, National University Hospital, National University Health System, Singapore, Singapore
| | - Eng Sing Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Healthcare Group Polyclinics, Singapore, Singapore
| | - Sebastian Maurer-Stroh
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
- GISAID Global Data Science Initiative, Munich, Germany
- National Public Health Laboratory, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yee-Sin Leo
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David C Lye
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Barnaby Edward Young
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
- Health Protection Research Unit in Emerging and Zoonotic Infections, National Institute of Health Research, University of Liverpool, Liverpool, United Kingdom
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Laurent Renia
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
48
|
Gagne M, Flynn BJ, Honeycutt CC, Flebbe DR, Andrew SF, Provost SJ, McCormick L, Van Ry A, McCarthy E, Todd JPM, Bao S, Teng IT, Marciano S, Rudich Y, Li C, Jain S, Wali B, Pessaint L, Dodson A, Cook A, Lewis MG, Andersen H, Zahradník J, Suthar MS, Nason MC, Foulds KE, Kwong PD, Roederer M, Schreiber G, Seder RA, Douek DC. Variant-proof high affinity ACE2 antagonist limits SARS-CoV-2 replication in upper and lower airways. Nat Commun 2024; 15:6894. [PMID: 39134521 PMCID: PMC11319446 DOI: 10.1038/s41467-024-51046-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: 11/21/2023] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
SARS-CoV-2 has the capacity to evolve mutations that escape vaccine- and infection-acquired immunity and antiviral drugs. A variant-agnostic therapeutic agent that protects against severe disease without putting selective pressure on the virus would thus be a valuable biomedical tool that would maintain its efficacy despite the ongoing emergence of new variants. Here, we challenge male rhesus macaques with SARS-CoV-2 Delta-the most pathogenic variant in a highly susceptible animal model. At the time of challenge, we also treat the macaques with aerosolized RBD-62, a protein developed through multiple rounds of in vitro evolution of SARS-CoV-2 RBD to acquire 1000-fold enhanced ACE2 binding affinity. RBD-62 treatment equivalently suppresses virus replication in both upper and lower airways, a phenomenon not previously observed with clinically approved vaccines. Importantly, RBD-62 does not block the development of virus-specific T- and B-cell responses and does not elicit anti-drug immunity. These data provide proof-of-concept that RBD-62 can prevent severe disease from a highly virulent variant.
Collapse
Affiliation(s)
- Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dillon R Flebbe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shayne F Andrew
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Samantha J Provost
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lauren McCormick
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Elizabeth McCarthy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Fred Hutch Cancer Center, Seattle, WA, USA
| | - John-Paul M Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Saran Bao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shir Marciano
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yinon Rudich
- Department of Earth and Planetary Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Chunlin Li
- Department of Earth and Planetary Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shilpi Jain
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Bushra Wali
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | | | | | | | | | | | - Jiří Zahradník
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Mehul S Suthar
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
49
|
Karim B, Barary M, Fereydouni Z, Sanjari E, Hosseinzadeh R, Salehi-Vaziri M, Maleki A. The nuts and bolts of recombination in the generation of SARS-CoV-2 variants; from XA to XBB. Lett Appl Microbiol 2024; 77:ovae074. [PMID: 39081071 DOI: 10.1093/lambio/ovae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/23/2024] [Accepted: 07/29/2024] [Indexed: 01/28/2025]
Abstract
Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), new variants with enhanced transmissibility and pathogenicity have surfaced. The World Health Organization has designated five such variants-Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529)-as variants of concern. Each variant exhibits distinct characteristics, with many displaying a combination of point mutations and insertions/deletions (indels). These genetic alterations, including mutations, recombinations, and rearrangements, contribute to the emergence of new strains that may exhibit modified phenotypes. However, identifying recombinant forms can be challenging due to their resemblance to other lineages. It is critical to monitor the evolution of new recombinant variants, particularly in light of the potential for vaccine-resistant strains and their accelerated propagation. Recombination has played a pivotal role in the development of certain SARS-CoV-2 variants, such as XA, XD, XF, XE, and XBB, among others. This report delves into the significance of recombination in the evolution of SARS-CoV-2 variants, especially Omicron sublineages, underscoring the necessity for continuous surveillance of the SARS-CoV-2 genome to identify newly emerged recombinant variants.
Collapse
Affiliation(s)
- Bardia Karim
- Student Research Committee, Babol University of Medical Sciences, Babol 4717647745, Iran
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Zahra Fereydouni
- COVID-19 National Reference Laboratory (CNRL), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran
| | - Elaheh Sanjari
- Student Research Committee, Faculty of Pharmacy, Islamic Azad University, Ayatollah Amoli Branch, Amol 678, Iran
| | - Rezvan Hosseinzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol 4717647745, Iran
| | - Mostafa Salehi-Vaziri
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Pasteur Ave., Tehran 01316943551, Iran
| | - Ali Maleki
- COVID-19 National Reference Laboratory (CNRL), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran
| |
Collapse
|
50
|
Rhodin MHJ, Reyes AC, Balakrishnan A, Bisht N, Kelly NM, Gibbons JS, Lloyd J, Vaine M, Cressey T, Crepeau M, Shen R, Manalo N, Castillo J, Levene RE, Leonard D, Zang T, Jiang L, Daniels K, Cox RM, Lieber CM, Wolf JD, Plemper RK, Leist SR, Scobey T, Baric RS, Wang G, Goodwin B, Or YS. The small molecule inhibitor of SARS-CoV-2 3CLpro EDP-235 prevents viral replication and transmission in vivo. Nat Commun 2024; 15:6503. [PMID: 39090095 PMCID: PMC11294338 DOI: 10.1038/s41467-024-50931-8] [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: 12/21/2023] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
The COVID-19 pandemic has led to the deaths of millions of people and severe global economic impacts. Small molecule therapeutics have played an important role in the fight against SARS-CoV-2, the virus responsible for COVID-19, but their efficacy has been limited in scope and availability, with many people unable to access their benefits, and better options are needed. EDP-235 is specifically designed to inhibit the SARS-CoV-2 3CLpro, with potent nanomolar activity against all SARS-CoV-2 variants to date, as well as clinically relevant human and zoonotic coronaviruses. EDP-235 maintains potency against variants bearing mutations associated with nirmatrelvir resistance. Additionally, EDP-235 demonstrates a ≥ 500-fold selectivity index against multiple host proteases. In a male Syrian hamster model of COVID-19, EDP-235 suppresses SARS-CoV-2 replication and viral-induced hamster lung pathology. In a female ferret model, EDP-235 inhibits production of SARS-CoV-2 infectious virus and RNA at multiple anatomical sites. Furthermore, SARS-CoV-2 contact transmission does not occur when naïve ferrets are co-housed with infected, EDP-235-treated ferrets. Collectively, these results demonstrate that EDP-235 is a broad-spectrum coronavirus inhibitor with efficacy in animal models of primary infection and transmission.
Collapse
Affiliation(s)
| | | | | | - Nalini Bisht
- Enanta Pharmaceuticals, Inc., Watertown, MA, USA
| | | | | | | | | | | | | | - Ruichao Shen
- Enanta Pharmaceuticals, Inc., Watertown, MA, USA
| | | | | | | | | | - Tianzhu Zang
- Enanta Pharmaceuticals, Inc., Watertown, MA, USA
| | - Lijuan Jiang
- Enanta Pharmaceuticals, Inc., Watertown, MA, USA
| | | | - Robert M Cox
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Carolin M Lieber
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Josef D Wolf
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Richard K Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sarah R Leist
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Trevor Scobey
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Yat Sun Or
- Enanta Pharmaceuticals, Inc., Watertown, MA, USA
| |
Collapse
|