151
|
Jeong YJ, Wi YM, Park H, Lee JE, Kim SH, Lee KS. Current and Emerging Knowledge in COVID-19. Radiology 2023; 306:e222462. [PMID: 36625747 PMCID: PMC9846833 DOI: 10.1148/radiol.222462] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/11/2023]
Abstract
COVID-19 has emerged as a pandemic leading to a global public health crisis of unprecedented morbidity. A comprehensive insight into the imaging of COVID-19 has enabled early diagnosis, stratification of disease severity, and identification of potential sequelae. The evolution of COVID-19 can be divided into early infectious, pulmonary, and hyperinflammatory phases. Clinical features, imaging features, and management are different among the three phases. In the early stage, peripheral ground-glass opacities are predominant CT findings, and therapy directly targeting SARS-CoV-2 is effective. In the later stage, organizing pneumonia or diffuse alveolar damage pattern are predominant CT findings and anti-inflammatory therapies are more beneficial. The risk of severe disease or hospitalization is lower in breakthrough or Omicron variant infection compared with nonimmunized or Delta variant infections. The protection rates of the fourth dose of mRNA vaccination were 34% and 67% against overall infection and hospitalizations for severe illness, respectively. After acute COVID-19 pneumonia, most residual CT abnormalities gradually decreased in extent, but they may remain as linear or multifocal reticular or cystic lesions. Advanced insights into the pathophysiologic and imaging features of COVID-19 along with vaccine benefits have improved patient care, but emerging knowledge of post-COVID-19 condition, or long COVID, also presents radiology with new challenges.
Collapse
Affiliation(s)
- Yeon Joo Jeong
- From the Department of Radiology, Research Institute for Convergence
of Biomedical Science and Technology, Pusan National University Yangsan
Hospital, Pusan National University School of Medicine, Yangsan, Korea (Y.J.J.);
Division of Infectious Diseases, Department of Internal Medicine (Y.M.W.,
S.H.K.) and Department of Radiology (K.S.L.), Samsung Changwon Hospital,
Sungkyunkwan University School of Medicine (SKKU-SOM), Changwon 51353, Korea;
Department of Electrical and Computer Engineering, Sungkyunkwan University,
Suwon, Korea (H.P.); Center for Neuroscience Imaging Research, Institute for
Basic Science, Suwon, Korea (H.P.); and Department of Radiology, Chonnam
National University Hospital, Gwangju, Korea (J.E.L.)
| | - Yu Mi Wi
- From the Department of Radiology, Research Institute for Convergence
of Biomedical Science and Technology, Pusan National University Yangsan
Hospital, Pusan National University School of Medicine, Yangsan, Korea (Y.J.J.);
Division of Infectious Diseases, Department of Internal Medicine (Y.M.W.,
S.H.K.) and Department of Radiology (K.S.L.), Samsung Changwon Hospital,
Sungkyunkwan University School of Medicine (SKKU-SOM), Changwon 51353, Korea;
Department of Electrical and Computer Engineering, Sungkyunkwan University,
Suwon, Korea (H.P.); Center for Neuroscience Imaging Research, Institute for
Basic Science, Suwon, Korea (H.P.); and Department of Radiology, Chonnam
National University Hospital, Gwangju, Korea (J.E.L.)
| | - Hyunjin Park
- From the Department of Radiology, Research Institute for Convergence
of Biomedical Science and Technology, Pusan National University Yangsan
Hospital, Pusan National University School of Medicine, Yangsan, Korea (Y.J.J.);
Division of Infectious Diseases, Department of Internal Medicine (Y.M.W.,
S.H.K.) and Department of Radiology (K.S.L.), Samsung Changwon Hospital,
Sungkyunkwan University School of Medicine (SKKU-SOM), Changwon 51353, Korea;
Department of Electrical and Computer Engineering, Sungkyunkwan University,
Suwon, Korea (H.P.); Center for Neuroscience Imaging Research, Institute for
Basic Science, Suwon, Korea (H.P.); and Department of Radiology, Chonnam
National University Hospital, Gwangju, Korea (J.E.L.)
| | - Jong Eun Lee
- From the Department of Radiology, Research Institute for Convergence
of Biomedical Science and Technology, Pusan National University Yangsan
Hospital, Pusan National University School of Medicine, Yangsan, Korea (Y.J.J.);
Division of Infectious Diseases, Department of Internal Medicine (Y.M.W.,
S.H.K.) and Department of Radiology (K.S.L.), Samsung Changwon Hospital,
Sungkyunkwan University School of Medicine (SKKU-SOM), Changwon 51353, Korea;
Department of Electrical and Computer Engineering, Sungkyunkwan University,
Suwon, Korea (H.P.); Center for Neuroscience Imaging Research, Institute for
Basic Science, Suwon, Korea (H.P.); and Department of Radiology, Chonnam
National University Hospital, Gwangju, Korea (J.E.L.)
| | - Si-Ho Kim
- From the Department of Radiology, Research Institute for Convergence
of Biomedical Science and Technology, Pusan National University Yangsan
Hospital, Pusan National University School of Medicine, Yangsan, Korea (Y.J.J.);
Division of Infectious Diseases, Department of Internal Medicine (Y.M.W.,
S.H.K.) and Department of Radiology (K.S.L.), Samsung Changwon Hospital,
Sungkyunkwan University School of Medicine (SKKU-SOM), Changwon 51353, Korea;
Department of Electrical and Computer Engineering, Sungkyunkwan University,
Suwon, Korea (H.P.); Center for Neuroscience Imaging Research, Institute for
Basic Science, Suwon, Korea (H.P.); and Department of Radiology, Chonnam
National University Hospital, Gwangju, Korea (J.E.L.)
| | - Kyung Soo Lee
- From the Department of Radiology, Research Institute for Convergence
of Biomedical Science and Technology, Pusan National University Yangsan
Hospital, Pusan National University School of Medicine, Yangsan, Korea (Y.J.J.);
Division of Infectious Diseases, Department of Internal Medicine (Y.M.W.,
S.H.K.) and Department of Radiology (K.S.L.), Samsung Changwon Hospital,
Sungkyunkwan University School of Medicine (SKKU-SOM), Changwon 51353, Korea;
Department of Electrical and Computer Engineering, Sungkyunkwan University,
Suwon, Korea (H.P.); Center for Neuroscience Imaging Research, Institute for
Basic Science, Suwon, Korea (H.P.); and Department of Radiology, Chonnam
National University Hospital, Gwangju, Korea (J.E.L.)
| |
Collapse
|
152
|
Kullappan M, Mary U, Ambrose JM, Veeraraghavan VP, Surapaneni KM. Elucidating the role of N440K mutation in SARS-CoV-2 spike - ACE-2 binding affinity and COVID-19 severity by virtual screening, molecular docking and dynamics approach. J Biomol Struct Dyn 2023; 41:912-929. [PMID: 34904526 DOI: 10.1080/07391102.2021.2014973] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
COVID-19 has become a public health concern around the world. The frequency of N440K variant was higher during the second wave in South India. The mutation was observed in the Receptor Binding Domain region (RBD) of the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) spike (S) protein. The binding affinity of SARS-CoV-2-Angiotensin-Converting Enzyme-2 (ACE-2) plays a major role in the transmission and severity of the disease. To understand the binding affinity of the wild and mutant SARS-CoV-2 S with ACE2, molecular modeling studies were carried out. We discovered that the wild SARS-CoV-2 S RBD-ACE-2 complex has a high binding affinity and stability than that of the mutant. The N440K strain escapes from antibody neutralization, which might increase reinfection and decrease vaccine efficiency. To find a potential inhibitor against mutant N440K SARS-CoV-2, a virtual screening process was carried out and found ZINC169293961, ZINC409421825 and ZINC22060839 as the best binding energy compounds. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Malathi Kullappan
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India
| | - Usha Mary
- Department of Chemistry, Panimalar Engineering College, Varadharajapuram, Poonamallee, Chennai, India
| | - Jenifer M Ambrose
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Velappanchavadi, Chennai, Tamil Nadu, India
| | - Krishna Mohan Surapaneni
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India.,Department of Biochemistry, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India.,Department of Molecular Virology, Clinical Skills & Simulation, Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India.,Department of Clinical Skills & Simulation, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India
| |
Collapse
|
153
|
Mao S, Li S, Zhang Y, Long L, Peng J, Cao Y, Mao JZ, Qi X, Xin Q, San G, Ding J, Jiang J, Bai X, Wang Q, Xu P, Xia H, Lu L, Xie L, Kong D, Zhu S, Xu W. A highly efficient needle-free-injection delivery system for mRNA-LNP vaccination against SARS-CoV-2. NANO TODAY 2023; 48:101730. [PMID: 36570700 PMCID: PMC9767897 DOI: 10.1016/j.nantod.2022.101730] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 05/14/2023]
Abstract
Despite the various vaccines that have been developed to combat the coronavirus disease 2019 (COVID-19) pandemic, the persistent and unpredictable mutations of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) require innovative and unremitting solutions to cope with the resultant immune evasion and establish a sustainable immune barrier. Here we introduce a vaccine-delivery system with a combination of a needle-free injection (NFI) device and a SARS-CoV-2-Spike-specific mRNA-Lipid Nanoparticle (LNP) vaccine. The benefits are duller pain and a significant increase of immunogenicity compared to the canonical needle injection (NI). From physicochemical and bioactivity analyses, the structure of the mRNA-LNP maintains stability upon NFI, contradictory to the belief that LNPs are inclined towards destruction under the high-pressure conditions of NFI. Moreover, mRNA-LNP vaccine delivered by NFI induces significantly more binding and neutralizing antibodies against SARS-CoV-2 variants than the same vaccine delivered by NI. Heterogeneous vaccination of BA.5-LNP vaccine with NFI enhanced the generation of neutralizing antibodies against Omicron BA.5 variants in rabbits previously vaccinated with non-BA.5-specific mRNA-LNP or other COVID-19 vaccines. NFI parameters can be adjusted to deliver mRNA-LNP subcutaneously or intramuscularly. Taken together, our results suggest that NFI-based mRNA-LNP vaccination is an effective substitute for the traditional NI-based mRNA-LNP vaccination.
Collapse
Affiliation(s)
- Shanhong Mao
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
- School of Light Industry, Beijing Technology and Business University, Beijing 100048, China
| | - Shiyou Li
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Yuxin Zhang
- Beijing QS Medical Technology Co.,Ltd., Beijing 100176, China
| | - Luoxin Long
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
| | - Junfeng Peng
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Yuanyan Cao
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
| | - Jessica Z Mao
- School of Veterinary Medicine & Biomedical Sciences, Texas A&M, College Station, TX 77843, USA
| | - Xin Qi
- Beijing QS Medical Technology Co.,Ltd., Beijing 100176, China
| | - Qi Xin
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Guoliang San
- Beijing QS Medical Technology Co.,Ltd., Beijing 100176, China
| | - Jing Ding
- Beijing QS Medical Technology Co.,Ltd., Beijing 100176, China
| | - Jun Jiang
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Xuejiao Bai
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Qianting Wang
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Pengfei Xu
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Huan Xia
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Lijun Lu
- Tricision Biotherapeutic Inc, Beijing 100176, Zhuhai 519040, China
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China
| | - Desheng Kong
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China
| | - Shuangli Zhu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Wenbo Xu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
154
|
Gomari MM, Tarighi P, Choupani E, Abkhiz S, Mohamadzadeh M, Rostami N, Sadroddiny E, Baammi S, Uversky VN, Dokholyan NV. Structural evolution of Delta lineage of SARS-CoV-2. Int J Biol Macromol 2023; 226:1116-1140. [PMID: 36435470 PMCID: PMC9683856 DOI: 10.1016/j.ijbiomac.2022.11.227] [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: 08/17/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
One of the main obstacles in prevention and treatment of COVID-19 is the rapid evolution of the SARS-CoV-2 Spike protein. Given that Spike is the main target of common treatments of COVID-19, mutations occurring at this virulent factor can affect the effectiveness of treatments. The B.1.617.2 lineage of SARS-CoV-2, being characterized by many Spike mutations inside and outside of its receptor-binding domain (RBD), shows high infectivity and relative resistance to existing cures. Here, utilizing a wide range of computational biology approaches, such as immunoinformatics, molecular dynamics (MD), analysis of intrinsically disordered regions (IDRs), protein-protein interaction analyses, residue scanning, and free energy calculations, we examine the structural and biological attributes of the B.1.617.2 Spike protein. Furthermore, the antibody design protocol of Rosetta was implemented for evaluation the stability and affinity improvement of the Bamlanivimab (LY-CoV55) antibody, which is not capable of interactions with the B.1.617.2 Spike. We observed that the detected mutations in the Spike of the B1.617.2 variant of concern can cause extensive structural changes compatible with the described variation in immunogenicity, secondary and tertiary structure, oligomerization potency, Furin cleavability, and drug targetability. Compared to the Spike of Wuhan lineage, the B.1.617.2 Spike is more stable and binds to the Angiotensin-converting enzyme 2 (ACE2) with higher affinity.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- Student Research Committee, Iran University of Medical Sciences, Tehran 1449614535, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Edris Choupani
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Shadi Abkhiz
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Masoud Mohamadzadeh
- Department of Chemistry, Faculty of Sciences, University of Hormozgan, Bandar Abbas 7916193145, Iran
| | - Neda Rostami
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak 3848177584, Iran
| | - Esmaeil Sadroddiny
- Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Soukayna Baammi
- African Genome Centre (AGC), Mohammed VI Polytechnic University, Benguerir 43150, Morocco
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA; Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia.
| | - Nikolay V Dokholyan
- Department of Pharmacology, Department of Biochemistry & Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 16802, USA.
| |
Collapse
|
155
|
Huo J, Dijokaite-Guraliuc A, Liu C, Zhou D, Ginn HM, Das R, Supasa P, Selvaraj M, Nutalai R, Tuekprakhon A, Duyvesteyn HME, Mentzer AJ, Skelly D, Ritter TG, Amini A, Bibi S, Adele S, Johnson SA, Paterson NG, Williams MA, Hall DR, Plowright M, Newman TAH, Hornsby H, de Silva TI, Temperton N, Klenerman P, Barnes E, Dunachie SJ, Pollard AJ, Lambe T, Goulder P, Fry EE, Mongkolsapaya J, Ren J, Stuart DI, Screaton GR. A delicate balance between antibody evasion and ACE2 affinity for Omicron BA.2.75. Cell Rep 2023; 42:111903. [PMID: 36586406 PMCID: PMC9747698 DOI: 10.1016/j.celrep.2022.111903] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have caused successive global waves of infection. These variants, with multiple mutations in the spike protein, are thought to facilitate escape from natural and vaccine-induced immunity and often increase in affinity for ACE2. The latest variant to cause concern is BA.2.75, identified in India where it is now the dominant strain, with evidence of wider dissemination. BA.2.75 is derived from BA.2 and contains four additional mutations in the receptor-binding domain (RBD). Here, we perform an antigenic and biophysical characterization of BA.2.75, revealing an interesting balance between humoral evasion and ACE2 receptor affinity. ACE2 affinity for BA.2.75 is increased 9-fold compared with BA.2; there is also evidence of escape of BA.2.75 from immune serum, particularly that induced by Delta infection, which may explain the rapid spread in India, where where there is a high background of Delta infection. ACE2 affinity appears to be prioritized over greater escape.
Collapse
Affiliation(s)
- Jiandong Huo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, the Wellcome Centre for Human Genetics, Oxford, UK; Guangzhou Laboratory, Bio-island, Guangzhou 510320, China.
| | - Aiste Dijokaite-Guraliuc
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chang Liu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Daming Zhou
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, the Wellcome Centre for Human Genetics, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Helen M Ginn
- Diamond Light Source, Ltd., Harwell Science and Innovation Campus, Didcot, UK
| | - Raksha Das
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Muneeswaran Selvaraj
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rungtiwa Nutalai
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aekkachai Tuekprakhon
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Helen M E Duyvesteyn
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, the Wellcome Centre for Human Genetics, Oxford, UK
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Donal Skelly
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Thomas G Ritter
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Ali Amini
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Sandra Adele
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Neil G Paterson
- Diamond Light Source, Ltd., Harwell Science and Innovation Campus, Didcot, UK
| | - Mark A Williams
- Diamond Light Source, Ltd., Harwell Science and Innovation Campus, Didcot, UK
| | - David R Hall
- Diamond Light Source, Ltd., Harwell Science and Innovation Campus, Didcot, UK
| | - Megan Plowright
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK; Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Thomas A H Newman
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK; Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK; Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich Chatham Maritime, Kent ME4 4TB, UK
| | - Paul Klenerman
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Eleanor Barnes
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Susanna J Dunachie
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand; Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Teresa Lambe
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK
| | - Elizabeth E Fry
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, the Wellcome Centre for Human Genetics, Oxford, UK.
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK.
| | - Jingshan Ren
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, the Wellcome Centre for Human Genetics, Oxford, UK.
| | - David I Stuart
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, the Wellcome Centre for Human Genetics, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK; Diamond Light Source, Ltd., Harwell Science and Innovation Campus, Didcot, UK.
| | - Gavin R Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK.
| |
Collapse
|
156
|
Lee MX, Peng S, Lee ARYB, Wong SY, Tay RYK, Li J, Tariq A, Goh CXY, Tan YK, Tan BKJ, Teo CB, Chan E, Ooi M, Chng WJ, Chee CE, Ho CLF, Walsh RJ, Wong M, Su Y, Alexander L, Sethi SK, Tan SSY, Chan YH, Tan KB, Lee SC, Chai LYA, Sundar R. Clinical efficacy and long-term immunogenicity of an early triple dose regimen of SARS-CoV-2 mRNA vaccination in cancer patients. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2023. [DOI: 10.47102/annals-acadmedsg.2022302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Introduction: Three doses of SARS-CoV-2 mRNA vaccines have been recommended for cancer patients to reduce the risk of severe disease. Anti-neoplastic treatment, such as chemotherapy, may affect long-term vaccine immunogenicity.
Method: Patients with solid or haematological cancer were recruited from 2 hospitals between July 2021 and March 2022. Humoral response was evaluated using GenScript cPASS surrogate virus neutralisation assays. Clinical outcomes were obtained from medical records and national mandatory-reporting databases.
Results: A total of 273 patients were recruited, with 40 having haematological malignancies and the rest solid tumours. Among the participants, 204 (74.7%) were receiving active cancer therapy, including 98 (35.9%) undergoing systemic chemotherapy and the rest targeted therapy or immunotherapy. All patients were seronegative at baseline. Seroconversion rates after receiving 1, 2 and 3 doses of SARS-CoV-2 mRNA vaccination were 35.2%, 79.4% and 92.4%, respectively. After 3 doses, patients on active treatment for haematological malignancies had lower antibodies (57.3%±46.2) when compared to patients on immunotherapy (94.1%±9.56, P<0.05) and chemotherapy (92.8%±18.1, P<0.05). SARS-CoV-2 infection was reported in 77 (28.2%) patients, of which 18 were severe. No patient receiving a third dose within 90 days of the second dose experienced severe infection.
Conclusion: This study demonstrates the benefit of early administration of the third dose among cancer patients.
Keywords: Cancer, oncology, SARS-CoV-2, third dose, vaccination
Collapse
Affiliation(s)
| | - Siyu Peng
- National University Hospital, Singapore
| | | | - Shi Yin Wong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ryan Yong Kiat Tay
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jiaqi Li
- School of Clinical Medicine, University of Cambridge, United Kingdom
| | - Areeba Tariq
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Claire Xin Yi Goh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ying Kiat Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Chong Boon Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Esther Chan
- National University Cancer Institute, Singapore, Singapore
| | - Melissa Ooi
- National University Cancer Institute, Singapore, Singapore
| | - Wee Joo Chng
- National University Cancer Institute, Singapore, Singapore
| | - Cheng Ean Chee
- National University Cancer Institute, Singapore, Singapore
| | - Carol LF Ho
- National University Cancer Institute, Singapore, Singapore
| | | | - Maggie Wong
- National University Cancer Institute, Singapore, Singapore
| | - Yan Su
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Lezhava Alexander
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | | | | | - Yiong Huak Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Soo-Chin Lee
- National University Cancer Institute, Singapore, Singapore
| | | | - Raghav Sundar
- National University Cancer Institute, Singapore, Singapore
| |
Collapse
|
157
|
Chrysostomou AC, Aristokleous A, Rodosthenous JH, Christodoulou C, Stathi G, Kostrikis LG. Detection of Circulating SARS-CoV-2 Variants of Concern (VOCs) Using a Multiallelic Spectral Genotyping Assay. Life (Basel) 2023; 13:life13020304. [PMID: 36836661 PMCID: PMC9960118 DOI: 10.3390/life13020304] [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: 12/19/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Throughout the coronavirus disease 2019 (COVID-19) pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has continuously evolved, resulting in new variants, some of which possess increased infectivity, immune evasion, and virulence. Such variants have been denoted by the World Health Organization as variants of concern (VOC) because they have resulted in an increased number of cases, posing a strong risk to public health. Thus far, five VOCs have been designated, Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529), including their sublineages. Next-generation sequencing (NGS) can produce a significant amount of information facilitating the study of variants; however, NGS is time-consuming and costly and not efficient during outbreaks, when rapid identification of VOCs is urgently needed. In such periods, there is a need for fast and accurate methods, such as real-time reverse transcription PCR in combination with probes, which can be used for monitoring and screening of the population for these variants. Thus, we developed a molecular beacon-based real-time RT-PCR assay according to the principles of spectral genotyping. This assay employs five molecular beacons that target ORF1a:ΔS3675/G3676/F3677, S:ΔH69/V70, S:ΔE156/F157, S:ΔΝ211, S:ins214EPE, and S:ΔL242/A243/L244, deletions and an insertion found in SARS-CoV-2 VOCs. This assay targets deletions/insertions because they inherently provide higher discrimination capacity. Here, the design process of the molecular beacon-based real-time RT-PCR assay for detection and discrimination of SARS-CoV-2 is presented, and experimental testing using SARS-CoV-2 VOC samples from reference strains (cultured virus) and clinical patient samples (nasopharyngeal samples), which have been previously classified using NGS, were evaluated. Based on the results, it was shown that all molecular beacons can be used under the same real-time RT-PCR conditions, consequently improving the time and cost efficiency of the assay. Furthermore, this assay was able to confirm the genotype of each of the tested samples from various VOCs, thereby constituting an accurate and reliable method for VOC detection and discrimination. Overall, this assay is a valuable tool that can be used for screening and monitoring the population for VOCs or other emerging variants, contributing to limiting their spread and protecting public health.
Collapse
Affiliation(s)
| | - Antonia Aristokleous
- Department of Biological Sciences, University of Cyprus, Aglantzia, 2109 Nicosia, Cyprus
| | | | | | - Georgia Stathi
- Department of Biological Sciences, University of Cyprus, Aglantzia, 2109 Nicosia, Cyprus
| | - Leondios G. Kostrikis
- Department of Biological Sciences, University of Cyprus, Aglantzia, 2109 Nicosia, Cyprus
- Cyprus Academy of Sciences, Letters, and Arts, 60-68 Phaneromenis Street, 1011 Nicosia, Cyprus
- Correspondence: ; Tel.: +35-72-289-2885
| |
Collapse
|
158
|
Marzi R, Bassi J, Silacci-Fregni C, Bartha I, Muoio F, Culap K, Sprugasci N, Lombardo G, Saliba C, Cameroni E, Cassotta A, Low JS, Walls AC, McCallum M, Tortorici MA, Bowen JE, Dellota EA, Dillen JR, Czudnochowski N, Pertusini L, Terrot T, Lepori V, Tarkowski M, Riva A, Biggiogero M, Franzetti-Pellanda A, Garzoni C, Ferrari P, Ceschi A, Giannini O, Havenar-Daughton C, Telenti A, Arvin A, Virgin HW, Sallusto F, Veesler D, Lanzavecchia A, Corti D, Piccoli L. Maturation of SARS-CoV-2 Spike-specific memory B cells drives resilience to viral escape. iScience 2023; 26:105726. [PMID: 36507220 PMCID: PMC9721160 DOI: 10.1016/j.isci.2022.105726] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Memory B cells (MBCs) generate rapid antibody responses upon secondary encounter with a pathogen. Here, we investigated the kinetics, avidity, and cross-reactivity of serum antibodies and MBCs in 155 SARS-CoV-2 infected and vaccinated individuals over a 16-month time frame. SARS-CoV-2-specific MBCs and serum antibodies reached steady-state titers with comparable kinetics in infected and vaccinated individuals. Whereas MBCs of infected individuals targeted both prefusion and postfusion Spike (S), most vaccine-elicited MBCs were specific for prefusion S, consistent with the use of prefusion-stabilized S in mRNA vaccines. Furthermore, a large fraction of MBCs recognizing postfusion S cross-reacted with human betacoronaviruses. The avidity of MBC-derived and serum antibodies increased over time resulting in enhanced resilience to viral escape by SARS-CoV-2 variants, including Omicron BA.1 and BA.2 sublineages, albeit only partially for BA.4 and BA.5 sublineages. Overall, the maturation of high-affinity and broadly reactive MBCs provides the basis for effective recall responses to future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Roberta Marzi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Jessica Bassi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Istvan Bartha
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Francesco Muoio
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Katja Culap
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nicole Sprugasci
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Gloria Lombardo
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Christian Saliba
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | | | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | | | | | - Laura Pertusini
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Tatiana Terrot
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Maciej Tarkowski
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Maira Biggiogero
- Clinical Research Unit, Clinica Luganese Moncucco, Lugano, Switzerland
| | | | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, Lugano, Switzerland
| | - Paolo Ferrari
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Alessandro Ceschi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | | | | | - Ann Arvin
- Vir Biotechnology, San Francisco, CA, USA
| | - Herbert W. Virgin
- Vir Biotechnology, San Francisco, CA, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - Davide Corti
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| |
Collapse
|
159
|
S Trimer Derived from SARS-CoV-2 B.1.351 and B.1.618 Induced Effective Immune Response against Multiple SARS-CoV-2 Variants. Vaccines (Basel) 2023; 11:vaccines11010193. [PMID: 36680037 PMCID: PMC9863711 DOI: 10.3390/vaccines11010193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/30/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
The spread of SARS-CoV-2 and its variants leads to a heavy burden on healthcare and the global economy, highlighting the need for developing vaccines that induce broad immunity against coronavirus. Here, we explored the immunogenicity of monovalent or bivalent spike (S) trimer subunit vaccines derived from SARS-CoV-2 B.1.351 (S1-2P) or/and B.1. 618 (S2-2P) in Balb/c mice. Both S1-2P and S2-2P elicited anti-spike antibody responses, and alum adjuvant induced higher levels of antibodies than Addavax adjuvant. The dose responses of the vaccines on immunogenicity were evaluated in vivo. A low dose of 5 μg monovalent recombinant protein or 2.5 μg bivalent vaccine triggered high-titer antibodies that showed cross-activity to Beta, Delta, and Gamma RBD in mice. The third immunization dose could boost (1.1 to 40.6 times) high levels of cross-binding antibodies and elicit high titers of neutralizing antibodies (64 to 1024) prototype, Beta, Delta, and Omicron variants. Furthermore, the vaccines were able to provoke a Th1-biased cellular immune response. Significantly, at the same antigen dose, S1-2P immune sera induced stronger broadly neutralizing antibodies against prototype, Beta, Delta, and Omicron variants compared to that induced by S2-2P. At the same time, the low dose of bivalent vaccine containing S2-2P and S1-2P (2.5 μg for each antigen) significantly improved the cross-neutralizing antibody responses. In conclusion, our results showed that monovalent S1-2P subunit vaccine or bivalent vaccine (S1-2P and S2-2P) induced potent humoral and cellular responses against multiple SARS-CoV-2 variants and provided valuable information for the development of recombinant protein-based SARS-CoV-2 vaccines that protect against emerging SARS-CoV-2 variants.
Collapse
|
160
|
Xu H, Wang T, Sun P, Hou X, Gong X, Zhang B, Wu J, Liu B. A bivalent subunit vaccine efficiently produced in Pichia pastoris against SARS-CoV-2 and emerging variants. Front Microbiol 2023; 13:1093080. [PMID: 36704561 PMCID: PMC9871450 DOI: 10.3389/fmicb.2022.1093080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus type II (SARS-CoV-2) variants have led to a decline in the protection of existing vaccines and antibodies, and there is an urgent need for a broad-spectrum vaccination strategy to reduce the pressure on the prevention and control of the pandemic. In this study, the receptor binding domain (RBD) of the SARS-CoV-2 Beta variant was successfully expressed through a glycoengineered yeast platform. To pursue a more broad-spectrum vaccination strategy, RBD-Beta and RBD-wild type were mixed at the ratio of 1:1 with Al(OH)3 and CpG double adjuvants for the immunization of BALB/c mice. This bivalent vaccine stimulated robust conjugated antibody titers and a broader spectrum of neutralizing antibody titers. These results suggested that a bivalent vaccine of RBD-Beta and RBD-wild type could be a possible broad-spectrum vaccination strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun Wu
- *Correspondence: Jun Wu, ✉
| | | |
Collapse
|
161
|
Venkatakrishnan AJ, Anand P, Lenehan PJ, Ghosh P, Suratekar R, Silvert E, Pawlowski C, Siroha A, Chowdhury DR, O'Horo JC, Yao JD, Pritt BS, Norgan AP, Hurt RT, Badley AD, Halamka J, Soundararajan V. Expanding repertoire of SARS-CoV-2 deletion mutations contributes to evolution of highly transmissible variants. Sci Rep 2023; 13:257. [PMID: 36604461 PMCID: PMC9815892 DOI: 10.1038/s41598-022-26646-5] [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: 08/13/2021] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
The emergence of highly transmissible SARS-CoV-2 variants and vaccine breakthrough infections globally mandated the characterization of the immuno-evasive features of SARS-CoV-2. Here, we systematically analyzed 2.13 million SARS-CoV-2 genomes from 188 countries/territories (up to June 2021) and performed whole-genome viral sequencing from 102 COVID-19 patients, including 43 vaccine breakthrough infections. We identified 92 Spike protein mutations that increased in prevalence during at least one surge in SARS-CoV-2 test positivity in any country over a 3-month window. Deletions in the Spike protein N-terminal domain were highly enriched for these 'surge-associated mutations' (Odds Ratio = 14.19, 95% CI 6.15-32.75, p value = 3.41 × 10-10). Based on a longitudinal analysis of mutational prevalence globally, we found an expanding repertoire of Spike protein deletions proximal to an antigenic supersite in the N-terminal domain that may be one of the key contributors to the evolution of highly transmissible variants. Finally, we generated clinically annotated SARS-CoV-2 whole genome sequences from 102 patients and identified 107 unique mutations, including 78 substitutions and 29 deletions. In five patients, we identified distinct deletions between residues 85-90, which reside within a linear B cell epitope. Deletions in this region arose contemporaneously on a diverse background of variants across the globe since December 2020. Overall, our findings based on genomic-epidemiology and clinical surveillance suggest that the genomic deletion of dispensable antigenic regions in SARS-CoV-2 may contribute to the evasion of immune responses and the evolution of highly transmissible variants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Venky Soundararajan
- nference, Cambridge, MA, 02139, USA.
- nference Labs, Bengaluru, Karnataka, India.
- Anumana, Cambridge, MA, 02139, USA.
| |
Collapse
|
162
|
Wang Q, Ye S, Zhou Z, Song A, Zhu X, Peng J, Liang R, Yang C, Yu X, Huang X, Yu J, Qiu Y, Ge X. Key mutations in the spike protein of SARS-CoV-2 affecting neutralization resistance and viral internalization. J Med Virol 2023; 95:e28407. [PMID: 36519597 PMCID: PMC9877909 DOI: 10.1002/jmv.28407] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/17/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
To control the ongoing COVID-19 pandemic, a variety of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines have been developed. However, the rapid mutations of SARS-CoV-2 spike (S) protein may reduce the protective efficacy of the existing vaccines which is mainly determined by the level of neutralizing antibodies targeting S. In this study, we screened prevalent S mutations and constructed 124 pseudotyped lentiviral particles carrying these mutants. We challenged these pseudoviruses with sera vaccinated by Sinovac CoronaVac and ZF2001 vaccines, two popular vaccines designed for the initial strain of SARS-CoV-2, and then systematically assessed the susceptivity of these SARS-CoV-2 variants to the immune sera of vaccines. As a result, 14 S mutants (H146Y, V320I + S477N, V382L, K444R, L455F + S477N, L452M + F486L, F486L, Y508H, P521R, A626S, S477N + S698L, A701V, S477N + T778I, E1144Q) were found to be significantly resistant to neutralization, indicating reduced protective efficacy of the vaccines against these SARS-CoV-2 variants. In addition, F486L and Y508H significantly enhanced the utilization of human angiotensin-converting enzyme 2, suggesting a potentially elevated infectivity of these two mutants. In conclusion, our results show that some prevalent S mutations of SARS-CoV-2 reduced the protective efficacy of current vaccines and enhance the infectivity of the virus, indicating the necessity of vaccine renewal and providing direction for the development of new vaccines.
Collapse
Affiliation(s)
- Qiong Wang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Sheng‐Bao Ye
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Zhi‐Jian Zhou
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - A‐Ling Song
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Xi Zhu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Jia‐Mei Peng
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Rui‐Min Liang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Chen‐Hui Yang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Xiao‐Wei Yu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina,Hunan Prevention and Treatment Institute for Occupational DiseasesChangshaHunanChina
| | - Xun Huang
- Department of Hospital Infection Control CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Jie Yu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina
| | - Xing‐Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of BiologyHunan UniversityChangshaHunanChina,Department of Hospital Infection Control CenterXiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
163
|
Alamri SS, Alsaieedi A, Khouqeer Y, Afeef M, Alharbi S, Algaissi A, Alghanmi M, Altorki T, Zawawi A, Alfaleh MA, Hashem AM, Alhabbab R. The importance of combining serological testing with RT-PCR assays for efficient detection of COVID-19 and higher diagnostic accuracy. PeerJ 2023; 11:e15024. [PMID: 37065688 PMCID: PMC10103696 DOI: 10.7717/peerj.15024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/17/2023] [Indexed: 04/18/2023] Open
Abstract
Misdiagnosing suspected COVID-19 individuals could largely contribute to the viruses transmission, therefore, making an accurate diagnosis of infected subjects vital in minimizing and containing the disease. Although RT-PCR is the standard method in detecting COVID-19, it is associated with some limitations, including possible false negative results. Therefore, serological testing has been suggested as a complement assay to RT-PCR to support the diagnosis of acute infections. In this study, 15 out of 639 unvaccinated healthcare workers (HCWs) were tested negative for COVID-19 by RT-PCR and were found seropositive for SARS-CoV-2 nucleocapsid protein-specific IgM and IgG antibodies. These participants underwent additional confirmatory RT-PCR and SARS-CoV-2 spike-specific ELISA tests. Of the 15 individuals, nine participants were found negative by second RT-PCR but seropositive for anti-spike IgM and IgG antibodies and neutralizing antibodies confirming their acute infection. At the time of collection, these nine individuals were in close contact with COVID-19-confirmed patients, with 77.7% reporting COVID-19-related symptoms. These results indicate that including serological tests in the current testing profile can provide better outcomes and help contain the spread of the virus by increasing diagnostic accuracy to prevent future outbreaks rapidly.
Collapse
Affiliation(s)
- Sawsan S. Alamri
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahdab Alsaieedi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Yousef Khouqeer
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Marwah Afeef
- Study & Research Department, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Samiyah Alharbi
- Intensive Care Unit, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Abdullah Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Medical Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Maimonah Alghanmi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tarfa Altorki
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat Zawawi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed A. Alfaleh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rowa Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
164
|
Emrani J, Hefner EN. Socio-demographic Heterogeneity in Prevalence of SARS-COV-2 Infection and Death Rate: Relevance to Black College Student Knowledge of COVID-19 and SARS-COV-2. J Racial Ethn Health Disparities 2023; 10:14-31. [PMID: 35119679 PMCID: PMC8815385 DOI: 10.1007/s40615-021-01193-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 02/03/2023]
Abstract
Black and Brown communities are affected disproportionately by COVID-19. In an attempt to learn if young Black college students unknowingly contribute to the spread of the COVID-19 in their communities, using surveys, this pilot study gauges the general safety knowledge and basic scientific knowledge of Black college students about SARS-COV-2 virus and COVID-19 at an HBCU. We also investigated whether students enrolled in chemistry courses designed for STEM (Science, Technology, and Engineering Majors) majors displayed increased knowledge of SARS-COV-2 and COVID-19 in comparison to their non-STEM major peers. Two sets of surveys with multiple choice questions, one with 25 and the other with 34 questions, were designed to assess general safety knowledge and basic scientific knowledge of the students about COVID-19 and the SARS-COV-2 virus. Survey questions were administered through Blackboard learning management system to one hundred eighty-seven (187) students in the summer of 2020 to two freshman non-science majors and in the fall of 2020 to one freshman non-science-major class, two freshmen STEM-major classes, and one senior STEM-major class. All students self-registered in the 6 chemistry classes at North Carolina A&T State University at random with no predetermined criteria. Results of the study show that regardless of their year of study, majority (> 90%) of the students possess basic scientific knowledge and are aware of the safety precautions concerning SARS-COV-2 virus and COVID-19. Majority of non-science major freshmen answered the basic safety questions correctly but were not able to choose the correct answers for the more specific scientific questions concerning SARS-COV-2 and COVID-19. Surprisingly, there was no significant difference in basic scientific knowledge regarding SARS-COV-2 and COVID-19 between STEM and non-STEM student populations, and first year STEM students were just as knowledgeable as senior STEM students. Based on these data, we speculate that students surveyed here have an acceptable basic understanding of how SARS-CoV-2 is transmitted, and therefore, they may not be a source of COVID-19 transmission to Black and Brown communities as this study confirms they are receiving accurate information about SARS-COV-2 and COVID-19. Possession of crucial timely and accurate knowledge about the health and safety is important in fighting racism and to gain equity within the society at large. By sharing the acquired knowledge, students can serve as positive role models for others in the community thus encouraging them to pursue science. Education brings equity, sharing the acquired knowledge encourages others to continue their education and succeed in obtaining higher degrees and better jobs as remedies for social inequality. Spread of accurate knowledge on various aspects of COVID-19 will also help remove fears of vaccination and hesitation towards visits to health clinics to resolve health issues. Relying on the results of this pilot study, we plan to explore these important factors further in our next study.
Collapse
Affiliation(s)
- Jahangir Emrani
- Department of Chemistry, North Carolina A&T State University, Greensboro, NC 27410 USA
| | - Elia Nichelle Hefner
- Department of Chemistry, North Carolina A&T State University, Greensboro, NC 27410 USA
| |
Collapse
|
165
|
Vogrig M, Berger AE, Bourlet T, Waeckel L, Haccourt A, Chanavat A, Hupin D, Roche F, Botelho-Nevers E, Pozzetto B, Paul S. Monitoring of Both Humoral and Cellular Immunities Could Early Predict COVID-19 Vaccine Efficacy Against the Different SARS-CoV2 Variants. J Clin Immunol 2023; 43:31-45. [PMID: 36006568 PMCID: PMC9403229 DOI: 10.1007/s10875-022-01354-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/17/2022] [Indexed: 01/21/2023]
Abstract
Reliable immunoassays are essential to early predict and monitor vaccine efficacy against SARS-CoV-2. The performance of an Interferon Gamma Release Assay (IGRA, QuantiFERON® SARS-CoV-2), and a current anti-spike serological test, compared to a plaque reduction neutralization test (PRNT) taken as gold standard were compared. Eighty vaccinated individuals, whose 16% had a previous history of COVID-19, were included in a longitudinal prospective study and sampled before and two to four weeks after each dose of vaccine. In non-infected patients, 2 doses were required for obtaining both positive IGRA and PRNT assays, while serology was positive after one dose. Each dose of vaccine significantly increased the humoral and cellular response. By contrast, convalescent subjects needed a single dose of vaccine to be positive on all 3 tests. Both IGRA and current serology assay were found predictive of a positive titer of neutralizing antibodies that is correlated with vaccine protection. Patients over 65 or 80 years old had a significantly reduced response. The response tended to be better with the heterologous scheme (vs. homologous) and with the mRNA-1273 vaccine (vs. BNT162b2) in the homologous group, in patients under 55 and under 65 years old, respectively. Finally, decrease intensity or absence of IGRA response and to a less extent of anti-spike serology were also correlated to reinfection which has occurred during the follow up. In conclusion, both IGRA and current anti-spike serology assays could be used at defined thresholds to monitor the vaccine response against SARS-CoV-2 and to simply identify non-responding individuals after a complete vaccination scheme. Two available specific tests (IGRA and anti-spike antibodies) could early assess the vaccine-induced immunity against SARS-CoV-2 at the individual scale, to potentially adapt the vaccination scheme in non-responder patients.
Collapse
Affiliation(s)
- Manon Vogrig
- Immunology Department, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Priest-en-Jarez, France
| | - Anne-Emmanuelle Berger
- Immunology Department, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
| | - Thomas Bourlet
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Priest-en-Jarez, France
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
| | - Louis Waeckel
- Immunology Department, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
| | - Alice Haccourt
- Immunology Department, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
| | - Alice Chanavat
- Immunology Department, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
| | - David Hupin
- Clinical and Exercise Physiology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Inserm, U1059, SAINBIOSE, Université de Lyon, Université Jean-Monnet, Saint-Etienne, France
| | - Frederic Roche
- Clinical and Exercise Physiology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Inserm, U1059, SAINBIOSE, Université de Lyon, Université Jean-Monnet, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
- Infectious Diseases Department, University Hospital of Saint-Etienne, Saint-Priest-en-Jarez, France
| | - Bruno Pozzetto
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Priest-en-Jarez, France
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France
| | - Stéphane Paul
- Immunology Department, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France.
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, F42023, Saint-Etienne, France.
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, 42055, Saint-Etienne, France.
- Faculté de Médecine, Campus Santé Innovations, 10 Rue de la Marandière, BP 80019, 42270, Saint-Priest-en-Jarez, France.
| |
Collapse
|
166
|
Lamkiewicz K, Esquivel Gomez LR, Kühnert D, Marz M. Genome Structure, Life Cycle, and Taxonomy of Coronaviruses and the Evolution of SARS-CoV-2. Curr Top Microbiol Immunol 2023; 439:305-339. [PMID: 36592250 DOI: 10.1007/978-3-031-15640-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Coronaviruses have a broad host range and exhibit high zoonotic potential. In this chapter, we describe their genomic organization in terms of encoded proteins and provide an introduction to the peculiar discontinuous transcription mechanism. Further, we present evolutionary conserved genomic RNA secondary structure features, which are involved in the complex replication mechanism. With a focus on computational methods, we review the emergence of SARS-CoV-2 starting with the 2019 strains. In that context, we also discuss the debated hypothesis of whether SARS-CoV-2 was created in a laboratory. We focus on the molecular evolution and the epidemiological dynamics of this recently emerged pathogen and we explain how variants of concern are detected and characterised. COVID-19, the disease caused by SARS-CoV-2, can spread through different transmission routes and also depends on a number of risk factors. We describe how current computational models of viral epidemiology, or more specifically, phylodynamics, have facilitated and will continue to enable a better understanding of the epidemic dynamics of SARS-CoV-2.
Collapse
Affiliation(s)
- Kevin Lamkiewicz
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Leutragraben 1, 07743, Jena, Germany
- European Virus Bioinformatics Center, Leutragraben 1, 07743, Jena, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Puschstr. 4, 04103, Leipzig, Germany
| | - Luis Roger Esquivel Gomez
- Transmission, Infection, Diversification and Evolution Group, Max Planck Institute for the Science of Human History, Kahlaische Straße 10, 07745, Jena, Germany
| | - Denise Kühnert
- Transmission, Infection, Diversification and Evolution Group, Max Planck Institute for the Science of Human History, Kahlaische Straße 10, 07745, Jena, Germany
- European Virus Bioinformatics Center, Leutragraben 1, 07743, Jena, Germany
| | - Manja Marz
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Leutragraben 1, 07743, Jena, Germany.
- European Virus Bioinformatics Center, Leutragraben 1, 07743, Jena, Germany.
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Puschstr. 4, 04103, Leipzig, Germany.
- FLI Leibniz Institute for Age Research, Beutenbergstraße 11, 07745, Jena, Germany.
| |
Collapse
|
167
|
Fraser DD, Patel MA, Van Nynatten LR, Martin C, Seney SL, Miller MR, Daley M, Slessarev M, Cepinskas G, Juneja GK, Sabourin V, Fox-Robichaud A, Yeh CH, Kim PY, Badrnya S, Oehler S, Miholits M, Webb B. Cross-immunity against SARS-COV-2 variants of concern in naturally infected critically ill COVID-19 patients. Heliyon 2023; 9:e12704. [PMID: 36594041 PMCID: PMC9797417 DOI: 10.1016/j.heliyon.2022.e12704] [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: 07/12/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Critically ill patients infected with SARS-CoV-2 display adaptive immunity, but it is unknown if they develop cross-reactivity to variants of concern (VOCs). We profiled cross-immunity against SARS-CoV-2 VOCs in naturally infected, non-vaccinated, critically ill COVID-19 patients. Wave-1 patients (wild-type infection) were similar in demographics to Wave-3 patients (wild-type/alpha infection), but Wave-3 patients had higher illness severity. Wave-1 patients developed increasing neutralizing antibodies to all variants, as did patients during Wave-3. Wave-3 patients, when compared to Wave-1, developed more robust antibody responses, particularly for wild-type, alpha, beta and delta variants. Within Wave-3, neutralizing antibodies were significantly less to beta and gamma VOCs, as compared to wild-type, alpha and delta. Patients previously diagnosed with cancer or chronic obstructive pulmonary disease had significantly fewer neutralizing antibodies. Naturally infected ICU patients developed adaptive responses to all VOCs, with greater responses in those patients more likely to be infected with the alpha variant, versus wild-type.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme
- AUC, area-under-the-curve
- Adaptive immunity
- Antibodies
- COPD, chronic obstructive pulmonary disease
- COVID-19
- COVID-19, coronavirus disease 2019
- ICU, intensive care unit
- IQR, interquartile range
- Intensive care units
- MFI, median fluorescence intensity
- MODS, multi-organ dysfunction score
- Neutralizing
- P/F, arterial partial pressure to inspired oxygen
- RBD, receptor binding domain
- REB, research ethics board
- ROC, receiver operating characteristic
- SARS-CoV-2
- SARS-CoV-2 alpha variant
- SARS-CoV-2 beta variant
- SARS-CoV-2 delta variant
- SARS-CoV-2 gamma variant
- SOFA, sequential organ failure assessment
- VOC, variants of concern
- VTE, venous thromboembolism
- WT, wild-type
Collapse
Affiliation(s)
- Douglas D. Fraser
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada
- Pediatrics, Western University, London, ON, N6A 3K7, Canada
- Clinical Neurological Sciences, Western University, London, ON, N6A 3K7, Canada
- Physiology & Pharmacology, Western University, London, ON, N6A 3K7, Canada
| | | | | | - Claudio Martin
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada
- Medicine, Western University, London, ON, N6A 3K7, Canada
| | | | - Michael R. Miller
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada
- Pediatrics, Western University, London, ON, N6A 3K7, Canada
| | - Mark Daley
- Epidemiology, Western University, London, ON, N6A 3K7, Canada
| | - Marat Slessarev
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada
- Medicine, Western University, London, ON, N6A 3K7, Canada
| | - Gediminas Cepinskas
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada
- Medical Biophysics, Western University, London, ON, N6A 3K7, Canada
| | - Ganeem K. Juneja
- Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, L8L 2X2, Canada
| | - Vanessa Sabourin
- Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, L8L 2X2, Canada
| | - Alison Fox-Robichaud
- Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, L8L 2X2, Canada
| | - Calvin H. Yeh
- Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, L8L 2X2, Canada
| | - Paul Y. Kim
- Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, L8L 2X2, Canada
| | | | | | | | - Brian Webb
- Thermo Fisher Scientific, Rockford, IL, USA
| |
Collapse
|
168
|
Li E, Han Q, Bi J, Wei S, Wang S, Zhang Y, Liu J, Feng N, Wang T, Wu J, Yang S, Zhao Y, Liu B, Yan F, Xia X. Therapeutic equine hyperimmune antibodies with high and broad-spectrum neutralizing activity protect rodents against SARS-CoV-2 infection. Front Immunol 2023; 14:1066730. [PMID: 36875106 PMCID: PMC9981790 DOI: 10.3389/fimmu.2023.1066730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
The emergence of SARS-CoV-2 variants stresses the continued need for broad-spectrum therapeutic antibodies. Several therapeutic monoclonal antibodies or cocktails have been introduced for clinical use. However, unremitting emerging SARS-CoV-2 variants showed reduced neutralizing efficacy by vaccine induced polyclonal antibodies or therapeutic monoclonal antibodies. In our study, polyclonal antibodies and F(ab')2 fragments with strong affinity produced after equine immunization with RBD proteins produced strong affinity. Notably, specific equine IgG and F(ab')2 have broad and high neutralizing activity against parental virus, all SARS-CoV-2 variants of concern (VOCs), including B.1.1,7, B.1.351, B.1.617.2, P.1, B.1.1.529 and BA.2, and all variants of interest (VOIs) including B.1.429, P.2, B.1.525, P.3, B.1.526, B.1.617.1, C.37 and B.1.621. Although some variants weaken the neutralizing ability of equine IgG and F(ab')2 fragments, they still exhibited superior neutralization ability against mutants compared to some reported monoclonal antibodies. Furthermore, we tested the pre-exposure and post-exposure protective efficacy of the equine immunoglobulin IgG and F(ab')2 fragments in lethal mouse and susceptible golden hamster models. Equine immunoglobulin IgG and F(ab')2 fragments effectively neutralized SARS-CoV-2 in vitro, fully protected BALB/c mice from the lethal challenge, and reduced golden hamster's lung pathological change. Therefore, equine pAbs are an adequate, broad coverage, affordable and scalable potential clinical immunotherapy for COVID-19, particularly for SARS-CoV-2 VOCs or VOIs.
Collapse
Affiliation(s)
- Entao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Qiuxue Han
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, Jilin Agriculture University, Changchun, China
| | - Jinhao Bi
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,Institute of Laboratory Animal Science, Chinese Academy of Medical Science and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Shimeng Wei
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,Institute of Laboratory Animal Science, Chinese Academy of Medical Science and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Shen Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ying Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Jun Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jun Wu
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liu
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
169
|
Abulsoud AI, El-Husseiny HM, El-Husseiny AA, El-Mahdy HA, Ismail A, Elkhawaga SY, Khidr EG, Fathi D, Mady EA, Najda A, Algahtani M, Theyab A, Alsharif KF, Albrakati A, Bayram R, Abdel-Daim MM, Doghish AS. Mutations in SARS-CoV-2: Insights on structure, variants, vaccines, and biomedical interventions. Biomed Pharmacother 2023; 157:113977. [PMID: 36370519 PMCID: PMC9637516 DOI: 10.1016/j.biopha.2022.113977] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
COVID-19 is a worldwide pandemic caused by SARS-coronavirus-2 (SARS-CoV-2). Less than a year after the emergence of the Covid-19 pandemic, many vaccines have arrived on the market with innovative technologies in the field of vaccinology. Based on the use of messenger RNA (mRNA) encoding the Spike SARS-Cov-2 protein or on the use of recombinant adenovirus vectors enabling the gene encoding the Spike protein to be introduced into our cells, these strategies make it possible to envisage the vaccination in a new light with tools that are more scalable than the vaccine strategies used so far. Faced with the appearance of new variants, which will gradually take precedence over the strain at the origin of the pandemic, these new strategies will allow a much faster update of vaccines to fight against these new variants, some of which may escape neutralization by vaccine antibodies. However, only a vaccination policy based on rapid and massive vaccination of the population but requiring a supply of sufficient doses could make it possible to combat the emergence of these variants. Indeed, the greater the number of infected individuals, the faster the virus multiplies, with an increased risk of the emergence of variants in these RNA viruses. This review will discuss SARS-CoV-2 pathophysiology and evolution approaches in altered transmission platforms and emphasize the different mutations and how they influence the virus characteristics. Also, this article summarizes the common vaccines and the implication of the mutations and genetic variety of SARS-CoV-2 on the COVID-19 biomedical arbitrations.
Collapse
Affiliation(s)
- Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Hussein M El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt.
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829, Cairo, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Doaa Fathi
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Eman A Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Agnieszka Najda
- Department of Vegetable Crops and Medicinal Plants University of Life Sciences, Lublin 50A Doświadczalna Street, 20-280, Lublin, Poland.
| | - Mohammad Algahtani
- Department of Laboratory & Blood Bank, Security Forces Hospital, P.O. Box 14799, Mecca 21955, Saudi Arabia
| | - Abdulrahman Theyab
- Department of Laboratory & Blood Bank, Security Forces Hospital, P.O. Box 14799, Mecca 21955, Saudi Arabia; College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Khalaf F Alsharif
- Department of Clinical Laboratory sciences, College of Applied medical sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Roula Bayram
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| |
Collapse
|
170
|
Storti P, Marchica V, Vescovini R, Franceschi V, Russo L, Notarfranchi L, Raimondi V, Toscani D, Burroughs Garcia J, Costa F, Dalla Palma B, Iannozzi NT, Sammarelli G, Donofrio G, Giuliani N. Immune response to SARS-CoV-2 mRNA vaccination and booster dose in patients with multiple myeloma and monoclonal gammopathies: impact of Omicron variant on the humoral response. Oncoimmunology 2022; 11:2120275. [PMID: 36105747 PMCID: PMC9467550 DOI: 10.1080/2162402x.2022.2120275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Rosanna Vescovini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Luca Russo
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | | | - Vincenzo Raimondi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Federica Costa
- School of Medicine, “Università del Piemonte Orientale”, Novara, Italy
| | | | | | | | - Gaetano Donofrio
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Hematology, “Azienda Ospedaliero-Universitaria di Parma”, Parma, Italy
| |
Collapse
|
171
|
Campos C, Colomer-Castell S, Garcia-Cehic D, Gregori J, Andrés C, Piñana M, González-Sánchez A, Borràs B, Parés-Badell O, Adombi CM, Ibañez-Lligoña M, Esperalba J, Codina MG, Rando-Segura A, Saubí N, Esteban JI, Rodriguez-Frías F, Pumarola T, Antón A, Quer J. The frequency of defective genomes in Omicron differs from that of the Alpha, Beta and Delta variants. Sci Rep 2022; 12:22571. [PMID: 36581627 PMCID: PMC9799681 DOI: 10.1038/s41598-022-24918-8] [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: 07/21/2022] [Accepted: 11/22/2022] [Indexed: 12/31/2022] Open
Abstract
The SARS-CoV-2 Omicron variant emerged showing higher transmissibility and possibly higher resistance to current COVID-19 vaccines than other variants dominating the global pandemic. In March 2020 we performed a study in clinical samples, where we found that a portion of genomes in the SARS-CoV-2 viral population accumulated deletions immediately before the S1/S2 cleavage site (furin-like cleavage site, PRRAR/S) of the spike gene, generating a frameshift and appearance of a premature stop codon. The main aim of this study was to determine the frequency of defective deletions in prevalent variants from the first to sixth pandemic waves in our setting and discuss whether the differences observed might support epidemiological proposals. The complete SARS-CoV-2 spike gene was deeply studied by next-generation sequencing using the MiSeq platform. More than 90 million reads were obtained from respiratory swab specimens of 78 COVID-19 patients with mild infection caused by the predominant variants circulating in the Barcelona city area during the six pandemic waves: B.1.5, B.1.1, B.1.177, Alpha, Beta, Delta, and Omicron. The frequency of defective genomes found in variants dominating the first and second waves was similar to that seen in Omicron, but differed from the frequencies seen in the Alpha, Beta and Delta variants. The changing pattern of mutations seen in the various SARS-CoV-2 variants driving the pandemic waves over time can affect viral transmission and immune escape. Here we discuss the putative biological effects of defective deletions naturally occurring before the S1/S2 cleavage site during adaption of the virus to human infection.
Collapse
Affiliation(s)
- Carolina Campos
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Campus de La UAB, Plaça Cívica, 08193, Bellaterra, Spain
| | - Sergi Colomer-Castell
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Campus de La UAB, Plaça Cívica, 08193, Bellaterra, Spain
| | - Damir Garcia-Cehic
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | - Josep Gregori
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | - Cristina Andrés
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Maria Piñana
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Alejandra González-Sánchez
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Blanca Borràs
- Preventive Medicine, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Oleguer Parés-Badell
- Preventive Medicine, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Caroline Melanie Adombi
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Marta Ibañez-Lligoña
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Juliana Esperalba
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Maria Gema Codina
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Ariadna Rando-Segura
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Narcis Saubí
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Juan Ignacio Esteban
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona (UAB), Campus de La UAB, Plaça Cívica, 08193, Bellaterra, Spain
| | - Francisco Rodriguez-Frías
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Campus de La UAB, Plaça Cívica, 08193, Bellaterra, Spain
- Biochemistry Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Tomàs Pumarola
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Microbiology Department, Universitat Autònoma de Barcelona (UAB), Campus de La UAB, Plaça Cívica, 08193, Bellaterra, Spain
| | - Andrés Antón
- Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Microbiology Department, Universitat Autònoma de Barcelona (UAB), Campus de La UAB, Plaça Cívica, 08193, Bellaterra, Spain.
| | - Josep Quer
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Campus de La UAB, Plaça Cívica, 08193, Bellaterra, Spain.
| |
Collapse
|
172
|
Takeshita M, Fukuyama H, Kamada K, Matsumoto T, Makino-Okamura C, Uchikubo-Kamo T, Tomabechi Y, Hanada K, Moriyama S, Takahashi Y, Ishigaki H, Nakayama M, Nguyen CT, Kitagawa Y, Itoh Y, Imai M, Maemura T, Furusawa Y, Ueki H, Iwatsuki-Horimoto K, Ito M, Yamayoshi S, Kawaoka Y, Shirouzu M, Ishii M, Saya H, Kondo Y, Kaneko Y, Suzuki K, Fukunaga K, Takeuchi T. Potent SARS-CoV-2 neutralizing antibodies with therapeutic effects in two animal models. iScience 2022; 25:105596. [PMID: 36406861 PMCID: PMC9664764 DOI: 10.1016/j.isci.2022.105596] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/07/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
The use of therapeutic neutralizing antibodies against SARS-CoV-2 infection has been highly effective. However, there remain few practical antibodies against viruses that are acquiring mutations. In this study, we created 494 monoclonal antibodies from patients with COVID-19-convalescent, and identified antibodies that exhibited the comparable neutralizing ability to clinically used antibodies in the neutralization assay using pseudovirus and authentic virus including variants of concerns. These antibodies have different profiles against various mutations, which were confirmed by cell-based assay and cryo-electron microscopy. To prevent antibody-dependent enhancement, N297A modification was introduced. Our antibodies showed a reduction of lung viral RNAs by therapeutic administration in a hamster model. In addition, an antibody cocktail consisting of three antibodies was also administered therapeutically to a macaque model, which resulted in reduced viral titers of swabs and lungs and reduced lung tissue damage scores. These results showed that our antibodies have sufficient antiviral activity as therapeutic candidates.
Collapse
Affiliation(s)
- Masaru Takeshita
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hidehiro Fukuyama
- RIKEN Center for Integrative Medical Sciences, Infectious Diseases Research unit, Kanagawa 230-0045, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Lymphocyte Differentiation, Kanagawa 230-0045, Japan
- Cell Integrative Science Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa 230-0045, Japan
- INSERM EST, 67037 Strasbourg Cedex 2, France
- Near-InfraRed Photo-Immunotherapy Research Institute, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Katsuhiko Kamada
- RIKEN Center for Biosystems Dynamics Research, Kanagawa 230-0045, Japan
- Laboratory for Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama 351-0198, Japan
| | | | - Chieko Makino-Okamura
- RIKEN Center for Integrative Medical Sciences, Laboratory for Lymphocyte Differentiation, Kanagawa 230-0045, Japan
| | | | - Yuri Tomabechi
- RIKEN Center for Biosystems Dynamics Research, Kanagawa 230-0045, Japan
| | - Kazuharu Hanada
- RIKEN Center for Biosystems Dynamics Research, Kanagawa 230-0045, Japan
| | - Saya Moriyama
- 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
| | - Hirohito Ishigaki
- Department of Pathology, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Misako Nakayama
- Department of Pathology, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Cong Thanh Nguyen
- Department of Pathology, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Yoshinori Kitagawa
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Masaki Imai
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- Center for Global Viral Diseases, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Tadashi Maemura
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yuri Furusawa
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- Center for Global Viral Diseases, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Hiroshi Ueki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- Center for Global Viral Diseases, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | | | - Mutsumi Ito
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Seiya Yamayoshi
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- Center for Global Viral Diseases, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- Center for Global Viral Diseases, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Mikako Shirouzu
- RIKEN Center for Biosystems Dynamics Research, Kanagawa 230-0045, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 162-8640, Japan
| | - Yasushi Kondo
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
173
|
Zhao F, Zai X, Zhang Z, Xu J, Chen W. Challenges and developments in universal vaccine design against SARS-CoV-2 variants. NPJ Vaccines 2022; 7:167. [PMID: 36535982 PMCID: PMC9761649 DOI: 10.1038/s41541-022-00597-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had become a global concern because of its unexpectedly high pathogenicity and transmissibility. SARS-CoV-2 variants that reduce the immune protection elicited from previous vaccination or natural infection raise challenges in controlling the spread of the pandemic. The development of universal vaccines against these variants seems to be a practical solution to alleviate the physical and economic effects caused by this disease, but it is hard to achieve. In this review, we describe the high mutation rate of RNA viruses and dynamic molecular structures of SARS-CoV-2 variants in several major neutralizing epitopes, trying to answer the question of why universal vaccines are difficult to design. Understanding the biological basis of immune evasion is crucial for combating these obstacles. We then summarize several advancements worthy of further study, including heterologous prime-boost regimens, construction of chimeric immunogens, design of protein nanoparticle antigens, and utilization of conserved neutralizing epitopes. The fact that some immunogens can induce cross-reactive immune responses against heterologous coronaviruses provides hints for universal vaccine development. We hope this review can provide inspiration to current universal vaccine studies.
Collapse
Affiliation(s)
- Fangxin Zhao
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiaodong Zai
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China
| | - Zhiling Zhang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China.
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 10071, China.
| |
Collapse
|
174
|
Atmeh PA, Gay L, Levasseur A, La Scola B, Olive D, Mezouar S, Gorvel JP, Mege JL. Macrophages and γδ T cells interplay during SARS-CoV-2 variants infection. Front Immunol 2022; 13:1078741. [PMID: 36601113 PMCID: PMC9806226 DOI: 10.3389/fimmu.2022.1078741] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction The emergence of several SARS-CoV-2 variants during the COVID pandemic has revealed the impact of variant diversity on viral infectivity and host immune responses. While antibodies and CD8 T cells are essential to clear viral infection, the protective role of innate immunity including macrophages has been recognized. The aims of our study were to compare the infectivity of different SARS-CoV-2 variants in monocyte-derived macrophages (MDM) and to assess their activation profiles and the role of ACE2 (Angiotensin-converting enzyme 2), the main SARS-CoV-2 receptor. We also studied the ability of macrophages infected to affect other immune cells such as γδ2 T cells, another partner of innate immune response to viral infections. Results We showed that the SARS-CoV-2 variants α-B.1.1.7 (United Kingdom), β-B.1.351 (South Africa), γ-P.1 (Brazil), δ-B.1.617 (India) and B.1.1.529 (Omicron), infected MDM without replication, the γ-Brazil variant exhibiting increased infectivity for MDM. No clear polarization profile of SARS-CoV-2 variants-infected MDM was observed. The β-B.1.351 (South Africa) variant induced macrophage activation while B.1.1.529 (Omicron) was rather inhibitory. We observed that SARS-CoV-2 variants modulated ACE2 expression in MDM. In particular, the β-B.1.351 (South Africa) variant induced a higher expression of ACE2, related to MDM activation. Finally, all variants were able to activate γδ2 cells among which γ-P.1 (Brazil) and β-B.1.351 (South Africa) variants were the most efficient. Conclusion Our data show that SARS-CoV-2 variants can infect MDM and modulate their activation, which was correlated with the ACE2 expression. They also affect γδ2 T cell activation. The macrophage response to SARS-CoV-2 variants was stereotypical.
Collapse
Affiliation(s)
- Perla Abou Atmeh
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Laetitia Gay
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Anthony Levasseur
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Bernard La Scola
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Daniel Olive
- Institut Paoli-Calmettes; Aix-Marseille Univ, UM105, Centre National de la Recherche Scientifique (CNRS) UMR 7258, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Jean-Pierre Gorvel
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille Luminy (CIML), Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
- Aix-Marseille Univ, Assitance Publique Hopitaux de Marseille (APHM), Hôpital de la Conception, Laboratoire d’Immunologie, Marseille, France
| |
Collapse
|
175
|
Fryer HA, Hartley GE, Edwards ES, O'Hehir RE, van Zelm MC. Humoral immunity and B-cell memory in response to SARS-CoV-2 infection and vaccination. Biochem Soc Trans 2022; 50:1643-1658. [PMID: 36421662 PMCID: PMC9788580 DOI: 10.1042/bst20220415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 01/15/2024]
Abstract
Natural infection with SARS-CoV-2 induces a robust circulating memory B cell (Bmem) population, which remains stable in number at least 8 months post-infection despite the contraction of antibody levels after 1 month. Multiple vaccines have been developed to combat the virus. These include two new formulations, mRNA and adenoviral vector vaccines, which have varying efficacy rates, potentially related to their distinct capacities to induce humoral immune responses. The mRNA vaccines BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna) elicit significantly higher serum IgG and neutralizing antibody levels than the adenoviral vector ChAdOx1 (AstraZeneca) and Ad26.COV2.S (Janssen) vaccines. However, all vaccines induce Spike- and RBD-specific Bmem, which are vital in providing long-lasting protection in the form of rapid recall responses to subsequent infections. Past and current SARS-CoV-2 variants of concern (VoC) have shown the capacity to escape antibody neutralization to varying degrees. A booster dose with an mRNA vaccine following primary vaccination restores antibody levels and improves the capacity of these antibodies and Bmem to bind viral variants, including the current VoC Omicron. Future experimental research will be essential to evaluate the durability of protection against VoC provided by each vaccine and to identify immune markers of protection to enable prognostication of people who are at risk of severe complications from COVID-19.
Collapse
Affiliation(s)
- Holly A. Fryer
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Gemma E. Hartley
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Emily S.J. Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Robyn E. O'Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia
| | - Menno C. van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
176
|
Torchia JA, Tavares AH, Carstensen LS, Chen DY, Huang J, Xiao T, Mukherjee S, Reeves PM, Tu H, Sluder AE, Chen B, Kotton DN, Bowen RA, Saeed M, Poznansky MC, Freeman GJ. Optimized ACE2 decoys neutralize antibody-resistant SARS-CoV-2 variants through functional receptor mimicry and treat infection in vivo. SCIENCE ADVANCES 2022; 8:eabq6527. [PMID: 36475798 PMCID: PMC9728973 DOI: 10.1126/sciadv.abq6527] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/01/2022] [Indexed: 05/14/2023]
Abstract
As severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) evolves to escape natural antibodies, it also loses sensitivity to therapeutic antibody drugs. By contrast, evolution selects for binding to ACE2, the cell-surface receptor required for SARS-CoV-2 infection. Consistent with this, we find that an ACE2 decoy neutralizes antibody-resistant variants, including Omicron, with no loss in potency. To identify design features necessary for in vivo activity, we compare several enzymatically inactive, Fc effector-silenced ACE2-Fc decoys. Inclusion of the ACE2 collectrin-like domain not only improves affinity for the S protein but also unexpectedly extends serum half-life and is necessary to reduce disease severity and viral titer in Syrian hamsters. Fc effector function is not required. The activity of ACE2 decoy receptors is due, in part, to their ability to trigger an irreversible structural change in the viral S protein. Our studies provide a new understanding of how ACE2 decoys function and support their development as therapeutics to treat ACE2-dependent coronaviruses.
Collapse
Affiliation(s)
- James A. Torchia
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Alexander H. Tavares
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Laura S. Carstensen
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Da-Yuan Chen
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jessie Huang
- Center for Regenerative Medicine of Boston University, Boston Medical Center, and The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Tianshu Xiao
- Division of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Sonia Mukherjee
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Patrick M. Reeves
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Hua Tu
- LakePharma Inc., San Carlos, CA 94070, USA
| | - Ann E. Sluder
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Bing Chen
- Division of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine of Boston University, Boston Medical Center, and The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Richard A. Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
177
|
Zhang GF, Meng W, Chen L, Ding L, Feng J, Perez J, Ali A, Sun S, Liu Z, Huang Y, Guo H, Gao S. Neutralizing antibodies to SARS-CoV-2 variants of concern including Delta and Omicron in subjects receiving mRNA-1273, BNT162b2, and Ad26.COV2.S vaccines. J Med Virol 2022; 94:5678-5690. [PMID: 35902378 PMCID: PMC9353285 DOI: 10.1002/jmv.28032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 01/06/2023]
Abstract
SARS-CoV-2 vaccines have contributed to the control of COVID-19 in some parts of the world. However, the constant emergence of variants of concern (VOCs) challenges the effectiveness of SARS-CoV-2 vaccines over time. In particular, Omicron contains a high number of mutations in the spike (S) protein gene, on which most vaccines were developed. In this study, we quantitated neutralizing antibodies in vaccine recipients at various times postvaccination using S protein-based pseudoviruses derived from wild type (WT) SARS-CoV-2 and five VOCs including Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529). We found that two-dose mRNA-1273 and BNT162b2 vaccines elicited robust neutralizing antibodies against WT, Alpha, Beta, Gamma, and Delta, but wanned after 6 months with a faster decline observed for BNT162b2. Both mRNA-1273 and BNT162b2 elicited weak neutralizing antibodies against Omicron. One dose of Ad26.COV2.S vaccine induced weaker neutralizing antibodies against WT and most VOCs than mRNA-1273 and BNT162b2 did but moderate neutralizing antibodies against Delta and Omicron, which lasted for 6 months. These results support current recommendations of the Centers for Disease Control and Prevention for a booster 5 months after full immunization with an mRNA-based vaccine and the use of an mRNA-based vaccine 2 months after Ad26.COV2.S vaccination.
Collapse
Affiliation(s)
- George Fei Zhang
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Wen Meng
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Luping Chen
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ling Ding
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Jian Feng
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Joseph Perez
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Abid Ali
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Shenyu Sun
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Zhentao Liu
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Electrical and Computer EngineeringSwanson School and EngineeringPittsburghPennsylvaniaUSA
| | - Yufei Huang
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Electrical and Computer EngineeringSwanson School and EngineeringPittsburghPennsylvaniaUSA
| | - Haitao Guo
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Shou‐Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
178
|
Rothenberger S, Hurdiss DL, Walser M, Malvezzi F, Mayor J, Ryter S, Moreno H, Liechti N, Bosshart A, Iss C, Calabro V, Cornelius A, Hospodarsch T, Neculcea A, Looser T, Schlegel A, Fontaine S, Villemagne D, Paladino M, Schiegg D, Mangold S, Reichen C, Radom F, Kaufmann Y, Schaible D, Schlegel I, Zitt C, Sigrist G, Straumann M, Wolter J, Comby M, Sacarcelik F, Drulyte I, Lyoo H, Wang C, Li W, Du W, Binz HK, Herrup R, Lusvarghi S, Neerukonda SN, Vassell R, Wang W, Adler JM, Eschke K, Nascimento M, Abdelgawad A, Gruber AD, Bushe J, Kershaw O, Knutson CG, Balavenkatraman KK, Ramanathan K, Wyler E, Teixeira Alves LG, Lewis S, Watson R, Haeuptle MA, Zürcher A, Dawson KM, Steiner D, Weiss CD, Amstutz P, van Kuppeveld FJM, Stumpp MT, Bosch BJ, Engler O, Trimpert J. The trispecific DARPin ensovibep inhibits diverse SARS-CoV-2 variants. Nat Biotechnol 2022; 40:1845-1854. [PMID: 35864170 PMCID: PMC9750863 DOI: 10.1038/s41587-022-01382-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/02/2022] [Indexed: 01/14/2023]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with potential resistance to existing drugs emphasizes the need for new therapeutic modalities with broad variant activity. Here we show that ensovibep, a trispecific DARPin (designed ankyrin repeat protein) clinical candidate, can engage the three units of the spike protein trimer of SARS-CoV-2 and inhibit ACE2 binding with high potency, as revealed by cryo-electron microscopy analysis. The cooperative binding together with the complementarity of the three DARPin modules enable ensovibep to inhibit frequent SARS-CoV-2 variants, including Omicron sublineages BA.1 and BA.2. In Roborovski dwarf hamsters infected with SARS-CoV-2, ensovibep reduced fatality similarly to a standard-of-care monoclonal antibody (mAb) cocktail. When used as a single agent in viral passaging experiments in vitro, ensovibep reduced the emergence of escape mutations in a similar fashion to the same mAb cocktail. These results support further clinical evaluation of ensovibep as a broad variant alternative to existing targeted therapies for Coronavirus Disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Sylvia Rothenberger
- Spiez Laboratory, Austrasse, Spiez, Switzerland
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Daniel L Hurdiss
- Department Biomolecular Health Sciences, Division Infectious Diseases & Immunology - Virology section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Cryo-Electron Microscopy, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Marcel Walser
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | - Jennifer Mayor
- Spiez Laboratory, Austrasse, Spiez, Switzerland
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Sarah Ryter
- Spiez Laboratory, Austrasse, Spiez, Switzerland
| | - Hector Moreno
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | | | | | - Chloé Iss
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | | | | | | | - Thamar Looser
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | - Anja Schlegel
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | | | | | | | | | | | - Filip Radom
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | | | - Iris Schlegel
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | - Christof Zitt
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | | | - Julia Wolter
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | - Marco Comby
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | - Ieva Drulyte
- Materials and Structural Analysis, Thermo Fisher Scientific, Eindhoven, Netherlands
| | - Heyrhyoung Lyoo
- Department Biomolecular Health Sciences, Division Infectious Diseases & Immunology - Virology section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Chunyan Wang
- Department Biomolecular Health Sciences, Division Infectious Diseases & Immunology - Virology section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Wentao Li
- Department Biomolecular Health Sciences, Division Infectious Diseases & Immunology - Virology section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Wenjuan Du
- Department Biomolecular Health Sciences, Division Infectious Diseases & Immunology - Virology section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | | | - Rachel Herrup
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Sabrina Lusvarghi
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Sabari Nath Neerukonda
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Russell Vassell
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Wei Wang
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Julia M Adler
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Kathrin Eschke
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | | | - Azza Abdelgawad
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Achim D Gruber
- Freie Universität Berlin, Institut für Tierpathologie, Berlin, Germany
| | - Judith Bushe
- Freie Universität Berlin, Institut für Tierpathologie, Berlin, Germany
| | - Olivia Kershaw
- Freie Universität Berlin, Institut für Tierpathologie, Berlin, Germany
| | - Charles G Knutson
- Novartis Institutes for BioMedical Research, PK Sciences, Cambridge, MA, USA
| | | | | | - Emanuel Wyler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | | | - Seth Lewis
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | | | | | | | | | - Carol D Weiss
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Frank J M van Kuppeveld
- Department Biomolecular Health Sciences, Division Infectious Diseases & Immunology - Virology section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | | | - Berend-Jan Bosch
- Department Biomolecular Health Sciences, Division Infectious Diseases & Immunology - Virology section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | | | - Jakob Trimpert
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| |
Collapse
|
179
|
Ustianowski A. Tixagevimab/cilgavimab for prevention and treatment of COVID-19: a review. Expert Rev Anti Infect Ther 2022; 20:1517-1527. [PMID: 36217836 DOI: 10.1080/14787210.2022.2134118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION There is a need to protect vulnerable individuals who do not respond to vaccination against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), particularly following the emergence of new variants. Tixagevimab/cilgavimab, the only monoclonal antibody combination authorized for pre-exposure prophylaxis of coronavirus disease 2019 (COVID-19), demonstrated efficacy in unvaccinated individuals in the PROVENT study. AREAS COVERED This review focuses predominantly on real-world evidence examining the effectiveness and safety of tixagevimab/cilgavimab in populations who are immunocompromised and otherwise vulnerable. The ability of tixagevimab/cilgavimab to neutralize Omicron subvariants, the appropriate dosage in vulnerable populations, and the impact of prior vaccination on tixagevimab/cilgavimab effectiveness are also discussed. EXPERT OPINION The tixagevimab/cilgavimab combination is important in providing protection in people who either cannot have a full vaccination or respond poorly to COVID-19 vaccines. Abundant clinical data have emerged to inform clinical use in adults in need, although some additional data-formal pediatric and adolescent studies, plus information on optimal doses required to protect against emerging variants, and the ideal interval between tixagevimab/cilgavimab dosing and vaccination-would be welcomed. Importantly, despite the current effectiveness of tixagevimab/cilgavimab, we must recognize the possibility that resistant SARS-CoV-2 variants could emerge in the future.
Collapse
Affiliation(s)
- Andrew Ustianowski
- Regional Infectious Diseases Unit, North Manchester General Hospital and Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK
| |
Collapse
|
180
|
Dobaño C, Ramírez-Morros A, Alonso S, Ruiz-Olalla G, Rubio R, Vidal M, Prados de la Torre E, Jairoce C, Mitchell RA, Barrios D, Jiménez A, Rodrigo Melero N, Carolis C, Izquierdo L, Zanoncello J, Aguilar R, Vidal-Alaball J, Moncunill G, Ruiz-Comellas A. Eleven-month longitudinal study of antibodies in SARS-CoV-2 exposed and naïve primary health care workers upon COVID-19 vaccination. Immunology 2022; 167:528-543. [PMID: 36065677 DOI: 10.1111/imm.13551] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022] Open
Abstract
We evaluated the kinetics of antibody responses to Two years into the COVID-19 pandemic and 1 year after the start of vaccination rollout, the world faced a peak of cases associated with the highly contagious Omicron variant of concern (VoC) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) and nucleocapsid (N) antigens over five cross-sectional visits (January-November 2021), and the determinants of pre-booster immunoglobulin levels, in a prospective cohort of vaccinated primary health care workers in Catalonia, Spain. Antibodies against S antigens after a full primary vaccination course, mostly with BNT162b2, decreased steadily over time and were higher in pre-exposed (n = 247) than naïve (n = 200) individuals, but seropositivity was maintained at 100% (100% IgG, 95.5% IgA, 30.6% IgM) up to 319 days after the first dose. Antibody binding to variants of concern was highly maintained for IgG compared to wild type but significantly reduced for IgA and IgM, particularly for Beta and Gamma. Factors significantly associated with longer-term antibodies included age, sex, occupation, smoking, adverse reaction to vaccination, levels of pre-vaccination SARS-CoV-2 antibodies, interval between disease onset and vaccination, hospitalization, oxygen supply, post COVID and symptomatology. Earlier morning vaccination hours were associated with higher IgG responses in pre-exposed participants. Symptomatic breakthroughs occurred in 9/447 (2.01%) individuals, all among naïve (9/200, 4.5%) and generally boosted antibody responses. Additionally, an increase in IgA and/or IgM seropositivity to variants, and N seroconversion at later time points (6.54%), indicated asymptomatic breakthrough infections, even among pre-exposed. Seropositivity remained highly stable over almost a year after vaccination. However, gradually waning of anti-S IgGs that correlate with neutralizing activity, coupled to evidence of an increase in breakthrough infections during the Delta and Omicron predominance, provides a rationale for booster immunization.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Anna Ramírez-Morros
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
| | - Selena Alonso
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Rocío Rubio
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | | | - Chenjerai Jairoce
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Robert A Mitchell
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Diana Barrios
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBERESP, Barcelona, Spain
| | - Natalia Rodrigo Melero
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Jasmina Zanoncello
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Josep Vidal-Alaball
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
- Health Promotion in Rural Areas Research Group, Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Anna Ruiz-Comellas
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
- Health Promotion in Rural Areas Research Group, Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain
- Centre d'Atenció Primària (CAP) Sant Joan de Vilatorrada, Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain
| |
Collapse
|
181
|
Addai E, Zhang L, Asamoah JKK, Preko AK, Arthur YD. Fractal-fractional age-structure study of omicron SARS-CoV-2 variant transmission dynamics. PARTIAL DIFFERENTIAL EQUATIONS IN APPLIED MATHEMATICS : A SPIN-OFF OF APPLIED MATHEMATICS LETTERS 2022; 6:100455. [PMID: 36277845 PMCID: PMC9576209 DOI: 10.1016/j.padiff.2022.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/08/2022]
Abstract
This paper proposes a new fractal-fractional age-structure model for the omicron SARS-CoV-2 variant under the Caputo-Fabrizio fractional order derivative. Caputo-Fabrizio fractal-fractional order is particularly successful in modelling real-world phenomena due to its repeated memory effect and ability to capture the exponentially decreasing impact of disease transmission dynamics. We consider two age groups, the first of which has a population under 50 and the second of a population beyond 50. Our results show that at a population dynamics level, there is a high infection and recovery of omicron SARS-CoV-2 variant infection among the population under 50 (Group-1), while a high infection rate and low recovery of omicron SARS-CoV-2 variant infection among the population beyond 50 (Group-2) when the fractal-fractional order is varied.
Collapse
Affiliation(s)
- Emmanuel Addai
- College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Taiyuan 030024, China
- Department of Mathematics, Taiyuan University of Technology, Shanxi Taiyuan 030024, China
| | - Lingling Zhang
- Department of Mathematics, Taiyuan University of Technology, Shanxi Taiyuan 030024, China
| | - Joshua Kiddy K Asamoah
- Department of Mathematics, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Ama Kyerewaa Preko
- College of Teacher Education, Zhejiang Normal University, Zhejiang Jinhua, 321004, China
| | - Yarhands Dissou Arthur
- Department of Mathematics Education, Akenten Appiah-Menka University of Skills Training and Entrepreneurial Development, Kumasi, Ghana
| |
Collapse
|
182
|
Yuan H, Chen P, Wan C, Li Y, Liu BF. Merging microfluidics with luminescence immunoassays for urgent point-of-care diagnostics of COVID-19. Trends Analyt Chem 2022; 157:116814. [PMID: 36373139 PMCID: PMC9637550 DOI: 10.1016/j.trac.2022.116814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/29/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
The Coronavirus disease 2019 (COVID-19) outbreak has urged the establishment of a global-wide rapid diagnostic system. Current widely-used tests for COVID-19 include nucleic acid assays, immunoassays, and radiological imaging. Immunoassays play an irreplaceable role in rapidly diagnosing COVID-19 and monitoring the patients for the assessment of their severity, risks of the immune storm, and prediction of treatment outcomes. Despite of the enormous needs for immunoassays, the widespread use of traditional immunoassay platforms is still limited by high cost and low automation, which are currently not suitable for point-of-care tests (POCTs). Microfluidic chips with the features of low consumption, high throughput, and integration, provide the potential to enable immunoassays for POCTs, especially in remote areas. Meanwhile, luminescence detection can be merged with immunoassays on microfluidic platforms for their good performance in quantification, sensitivity, and specificity. This review introduces both homogenous and heterogenous luminescence immunoassays with various microfluidic platforms. We also summarize the strengths and weaknesses of the categorized methods, highlighting their recent typical progress. Additionally, different microfluidic platforms are described for comparison. The latest advances in combining luminescence immunoassays with microfluidic platforms for POCTs of COVID-19 are further explained with antigens, antibodies, and related cytokines. Finally, challenges and future perspectives were discussed.
Collapse
Affiliation(s)
- Huijuan Yuan
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chao Wan
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
183
|
Luo WR, Wu XM, Wang W, Yu JL, Chen QQ, Zhou X, Huang X, Pan HF, Liu ZR, Gao Y, He J. Novel coronavirus mutations: Vaccine development and challenges. Microb Pathog 2022; 173:105828. [PMID: 36243381 PMCID: PMC9561474 DOI: 10.1016/j.micpath.2022.105828] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The ongoing global pandemic of novel coronavirus pneumonia (COVID-19) caused by the SARS-CoV-2 has a significant impact on global health and economy system. In this context, there have been some landmark advances in vaccine development. Over 100 new coronavirus vaccine candidates have been approved for clinical trials, with ten WHO-approved vaccines including four inactivated virus vaccines, two mRNA vaccines, three recombinant viral vectored vaccines and one protein subunit vaccine on the "Emergency Use Listing". Although the SARS-CoV-2 has an internal proofreading mechanism, there have been a number of mutations emerged in the pandemic affecting its transmissibility, pathogenicity and immunogenicity. Of these, mutations in the spike (S) protein and the resultant mutant variants have posed new challenges for vaccine development and application. In this review article, we present an overview of vaccine development, the prevalence of new coronavirus variants and their impact on protective efficacy of existing vaccines and possible immunization strategies coping with the viral mutation and diversity.
Collapse
Affiliation(s)
- Wan-Rong Luo
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Xiao-Min Wu
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Wei Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), 17 Lujiang Road, Hefei, Anhui, China
| | - Jun-Ling Yu
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Qing-Qing Chen
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Xue Zhou
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Xin'er Huang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Zhi-Rong Liu
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China; Department of Microbiology Laboratory, Public Health Research Institute of Anhui Province, 12560, Fanhua Avenue, Hefei, Anhui, China.
| | - Yong Gao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), 17 Lujiang Road, Hefei, Anhui, China.
| | - Jun He
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China; Department of Microbiology Laboratory, Public Health Research Institute of Anhui Province, 12560, Fanhua Avenue, Hefei, Anhui, China; Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
184
|
Parn S, Savsani K, Dakshanamurthy S. SARS-CoV-2 Omicron (BA.1 and BA.2) specific novel CD8+ and CD4+ T cell epitopes targeting spike protein. IMMUNOINFORMATICS (AMSTERDAM, NETHERLANDS) 2022; 8:100020. [PMID: 36337685 PMCID: PMC9624113 DOI: 10.1016/j.immuno.2022.100020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The Omicron (BA.1/B.1.1.529) variant of SARS-CoV-2 harbors an alarming 37 mutations on its spike protein, reducing the efficacy of current COVID-19 vaccines. In this study, we identified CD8+ and CD4+ T cell epitopes from SARS-CoV-2 S protein mutants. To identify the highest quality CD8 and CD4 epitopes from the Omicron variant, we selected epitopes with a high binding affinity towards both MHC I and MHC II molecules. We applied other clinical checkpoint predictors, including immunogenicity, antigenicity, allergenicity, instability and toxicity. Subsequently, we found eight Omicron (BA.1/B.1.1.529) specific CD8+ and eleven CD4+ T cell epitopes with a world population coverage of 76.16% and 97.46%, respectively. Additionally, we identified common epitopes across Omicron BA.1 and BA.2 lineages that target mutations critical to SARS-CoV-2 virulence. Further, we identified common epitopes across B.1.1.529 and other circulating SARS-CoV-2 variants, such as B.1.617.2 (Delta). We predicted CD8 epitopes' binding affinity to murine MHC alleles to test the vaccine candidates in preclinical models. The CD8 epitopes were further validated using our previously developed software tool PCOptim. We then modeled the three-dimensional structures of our top CD8 epitopes to investigate the binding interaction between peptide-MHC and peptide-MHC-TCR complexes. Notably, our identified epitopes are targeting the mutations on the RNA-binding domain and the fusion sites of S protein. This could potentially eliminate viral infections and form long-term immune responses compared to relatively short-lived mRNA vaccines and maximize the efficacy of vaccine candidates against the current pandemic and potential future variants.
Collapse
Affiliation(s)
- Simone Parn
- University of the District of Columbia, Washington, D.C, USA
| | - Kush Savsani
- Virginia Commonwealth University, Richmond, VA, USA
| | - Sivanesan Dakshanamurthy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057 USA
| |
Collapse
|
185
|
Dhawan M, Sharma A, Priyanka, Thakur N, Rajkhowa TK, Choudhary OP. Delta variant (B.1.617.2) of SARS-CoV-2: Mutations, impact, challenges and possible solutions. Hum Vaccin Immunother 2022; 18:2068883. [PMID: 35507895 PMCID: PMC9359381 DOI: 10.1080/21645515.2022.2068883] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/04/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022] Open
Abstract
Since commencement of COVID-19 pandemic, several SARS-CoV-2 variants have emerged amid containment efforts via vaccination. The Delta variant (B.1.617.2), discovered in October 2020, was designated as a VOC by the WHO on May 11, 2021. The enhanced transmissibility of Delta variant has been associated with critical mutations such as D614G, L452R, P681R, and T478K in the S-protein. The increased affinity of the S-protein and ACE2 has been postulated as a key reason for decreased vaccine efficacy. As per evidence, the Delta variant possesses increased transmissibility and decreased vaccine efficacy compared to other VOCs like Alpha and Beta. This has led to concerns regarding the acquisition of novel mutations in the Delta variant and outbreaks in vulnerable communities, including vaccinated people. In this mini-review of Delta variant, we have explained its evolution and characteristics, the impact of spike mutations on infectivity and immune evasion, and measures to combat future outbreaks.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- Trafford College, Altrincham, Manchester, UK
| | - Abhilasha Sharma
- Department of Life Science, University School of Sciences, Gujarat University, Ahmedabad, India
| | - Priyanka
- Independent Researcher, 07, Type IV Quarter, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, Mizoram, India
| | - Nanamika Thakur
- Department of Medical Lab Technology, Faculty of Allied and Healthcare Sciences, GNA University, Phagwara, Punjab, India
| | - Tridib Kumar Rajkhowa
- Department of Veterinary Pathology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, Mizoram, India
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy and Histology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, Mizoram, India
| |
Collapse
|
186
|
Mohseni Afshar Z, Barary M, Hosseinzadeh R, Alijanpour A, Hosseinzadeh D, Ebrahimpour S, Nazary K, Sio TT, Sullman MJM, Carson-Chahhoud K, Babazadeh A. Breakthrough SARS-CoV-2 infections after vaccination: a critical review. Hum Vaccin Immunother 2022; 18:2051412. [PMID: 35302905 PMCID: PMC9115792 DOI: 10.1080/21645515.2022.2051412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/06/2022] [Indexed: 12/28/2022] Open
Abstract
At the beginning of the current pandemic, it was believed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection would induce lifelong immunity and that reinfections would be unlikely. However, after several cases of reinfection were documented in previously infected patients, this was understood to be a false assumption, and this waning humoral immunity has raised significant concerns. Accordingly, long-term and durable vaccine-induced antibody protection against infection have also become a challenge, as several breakthroughs of COVID-19 infection have been identified in individuals who were fully vaccinated. This review discusses the current evidence on breakthrough COVID-19 infections occurring after vaccination.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Rezvan Hosseinzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | | | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Kosar Nazary
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Terence T. Sio
- Department of Radiation Oncology, Mayo Clinic, Phoenix, Arizona, USA
| | - Mark J. M. Sullman
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | | | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
187
|
Bian L, Liu J, Gao F, Gao Q, He Q, Mao Q, Wu X, Xu M, Liang Z. Research progress on vaccine efficacy against SARS-CoV-2 variants of concern. Hum Vaccin Immunother 2022; 18:2057161. [PMID: 35438600 PMCID: PMC9115786 DOI: 10.1080/21645515.2022.2057161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 01/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to circulate worldwide and a variety of variants have emerged. Variants of concern (VOC) designated by the World Health Organization (WHO) have triggered epidemic waves due to their strong infectivity or pathogenicity and potential immune escape, among other reasons. Although large-scale vaccination campaigns undertaken globally have contributed to the improved control of SARS-CoV-2, the efficacies of current vaccines against VOCs have declined to various degrees. In particular, the highly infectious Delta and Omicron variants have caused recent epidemics and prompted concerns about control measures. This review summarizes current VOCs, the protective efficacy of vaccines against VOCs, and the shortcomings in methods for evaluating vaccine efficacy. In addition, strategies for responding to variants are proposed for future epidemic prevention and control as well as for vaccine research and development.
Collapse
Affiliation(s)
- Lianlian Bian
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Jianyang Liu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Fan Gao
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Qiushuang Gao
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Qian He
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Qunying Mao
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Xing Wu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Miao Xu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Zhenglun Liang
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
188
|
Casetti IC, Borsani O, Rumi E. COVID-19 in Patients with Hematologic Diseases. Biomedicines 2022; 10:3069. [PMID: 36551825 PMCID: PMC9775038 DOI: 10.3390/biomedicines10123069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
The COVID-19 outbreak had a strong impact on people's lives all over the world. Patients with hematologic diseases have been heavily affected by the pandemic, because their immune system may be compromised due to anti-cancer or immunosuppressive therapies and because diagnosis and treatment of their baseline conditions were delayed during lockdowns. Hematologic malignancies emerged very soon as risk factors for severe COVID-19 infection, increasing the mortality rate. SARS-CoV2 can also induce or exacerbate immune-mediated cytopenias, such as autoimmune hemolytic anemias, complement-mediated anemias, and immune thrombocytopenia. Active immunization with vaccines has been shown to be the best prophylaxis of severe COVID-19 in hematologic patients. However, the immune response to vaccines may be significantly impaired, especially in those receiving anti-CD20 monoclonal antibodies or immunosuppressive agents. Recently, antiviral drugs and monoclonal antibodies have become available for pre-exposure and post-exposure prevention of severe COVID-19. As adverse events after vaccines are extremely rare, the cost-benefit ratio is largely in favor of vaccination, even in patients who might be non-responders; in the hematological setting, all patients should be considered at high risk of developing complications due to SARS-CoV2 infection and should be offered all the therapies aimed to prevent them.
Collapse
Affiliation(s)
| | - Oscar Borsani
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Elisa Rumi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
189
|
Lu Y, Zhu Q, Fox DM, Gao C, Stanley SA, Luo K. SARS-CoV-2 down-regulates ACE2 through lysosomal degradation. Mol Biol Cell 2022; 33:ar147. [PMID: 36287912 PMCID: PMC9727799 DOI: 10.1091/mbc.e22-02-0045] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilizes its Spike (S) glycoprotein to bind to the angiotensin-converting enzyme 2 (ACE2) receptor for cellular entry. ACE2 is a critical negative regulator of the renin-angiotensin system and plays a protective role in preventing tissue injury. Expression of ACE2 has been shown to decrease upon infection by SARS-CoV. However, whether SARS-CoV-2 down-regulates ACE2 and the underlying mechanism and biological impact of this down-regulation have not been well defined. Here we show that the SARS-CoV-2 infection down-regulates ACE2 in vivo in an animal model, and in cultured cells in vitro, by inducing clathrin- and AP2-dependent endocytosis, leading to its degradation in the lysosome. SARS-CoV-2 S-treated cells and ACE2 knockdown cells exhibit similar alterations in downstream gene expression, with a pattern indicative of activated cytokine signaling that is associated with respiratory distress and inflammatory diseases often observed in COVID-19 patients. Finally, we have identified a soluble ACE2 fragment with a stronger binding to SARS-CoV-2 S that can efficiently block ACE2 down-regulation and viral infection. Thus, our study suggests that ACE2 down-regulation represents an important mechanism underlying SARS-CoV-2-associated pathology, and blocking this process could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yi Lu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Douglas M. Fox
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720,Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720
| | - Carol Gao
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Sarah A. Stanley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720,Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720,*Address correspondence to: Kunxin Luo ()
| |
Collapse
|
190
|
Dingemans J, van der Veer BMJW, Gorgels KMF, Hackert V, den Heijer CDJ, Hoebe CJPA, Savelkoul PHM, van Alphen LB. Investigating SARS-CoV-2 breakthrough infections per variant and vaccine type. Front Microbiol 2022; 13:1027271. [PMID: 36504818 PMCID: PMC9729533 DOI: 10.3389/fmicb.2022.1027271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
Breakthrough SARS-CoV-2 infections have been reported in fully vaccinated individuals, in spite of the high efficacy of the currently available vaccines, proven in trials and real-world studies. Several variants of concern (VOC) have been proffered to be associated with breakthrough infections following immunization. In this study, we investigated 378 breakthrough infections recorded between January and July 2021 and compared the distribution of SARS-CoV-2 genotypes identified in 225 fully vaccinated individuals to the frequency of circulating community lineages in the region of South Limburg (The Netherlands) in a week-by-week comparison. Although the proportion of breakthrough infections was relatively low and stable when the Alpha variant was predominant, the rapid emergence of the Delta variant lead to a strong increase in breakthrough infections, with a higher relative proportion of individuals vaccinated with Vaxzevria or Jcovden being infected compared to those immunized with mRNA-based vaccines. A significant difference in median age was observed when comparing fully vaccinated individuals with severe symptoms (83 years) to asymptomatic cases (46.5 years) or individuals with mild-to-moderate symptoms (42 years). There was no association between SARS-CoV-2 genotype or vaccine type and disease symptoms. Furthermore, the majority of adaptive mutations were concentrated in the N-terminal domain of the Spike protein, highlighting its role in immune evasion. Interestingly, symptomatic individuals harbored significantly higher SARS-CoV-2 loads than asymptomatic vaccinated individuals and breakthrough infections caused by the Delta variant were associated with increased viral loads compared to those caused by the Alpha variant. In addition, we investigated the role of the Omicron variant in causing breakthrough infections by analyzing 135 samples that were randomly selected for genomic surveillance during the transition period from Delta to Omicron. We found that the proportion of Omicron vs. Delta infections was significantly higher in individuals who received a booster vaccine compared to both unvaccinated and fully vaccinated individuals. Altogether, these results indicate that the emergence of the Delta variant and in particular Omicron has lowered the efficiency of particular vaccine types to prevent SARS-CoV-2 infections and that, although rare, the elderly are particularly at risk of becoming severely infected as the consequence of a breakthrough infection.
Collapse
Affiliation(s)
- Jozef Dingemans
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands,*Correspondence: Jozef Dingemans, ; Brian M. J. W. van der Veer,
| | - Brian M. J. W. van der Veer
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands,*Correspondence: Jozef Dingemans, ; Brian M. J. W. van der Veer,
| | - Koen M. F. Gorgels
- Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands
| | - Volker Hackert
- Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands,Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Casper D. J. den Heijer
- Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands,Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Christian J. P. A Hoebe
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands,Department of Sexual Health, Infectious Diseases and Environmental Health, South Limburg Public Health Service, Heerlen, Netherlands,Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Paul H. M. Savelkoul
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands
| | - Lieke B. van Alphen
- Department of Medical Microbiology, Infectious diseases and Infection prevention, Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands
| |
Collapse
|
191
|
Cox RM, Lieber CM, Wolf JD, Karimi A, Lieberman NAP, Sticher ZM, Roychoudhury P, Andrews MK, Krueger RE, Natchus MG, Painter GR, Kolykhalov AA, Greninger AL, Plemper RK. Paxlovid-like nirmatrelvir/ritonavir fails to block SARS-CoV-2 transmission in ferrets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.11.20.517271. [PMID: 36451893 PMCID: PMC9709798 DOI: 10.1101/2022.11.20.517271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Despite the continued spread of SARS-CoV-2 and emergence of variants of concern (VOC) that are capable of escaping preexisting immunity, therapeutic options are underutilized. In addition to preventing severe disease in high-risk patients, antivirals may contribute to interrupting transmission chains. The FDA has granted emergency use authorizations for two oral drugs, molnupiravir and paxlovid. Initial clinical trials suggested an efficacy advantage of paxlovid, giving it a standard-of-care-like status in the United States. However, recent retrospective clinical studies suggested a more comparable efficacy of both drugs in preventing complicated disease and case-fatalities in older adults. For a direct efficacy comparison under controlled conditions, we assessed potency of both drugs against SARS-CoV-2 in two relevant animal models; the Roborovski dwarf hamster model for severe COVID-19 in high-risk patients and the ferret model of upper respiratory tract disease and transmission. After infection of dwarf hamsters with VOC omicron, paxlovid and molnupiravir were efficacious in mitigating severe disease and preventing death. However, a pharmacokinetics-confirmed human equivalent dose of paxlovid did not significantly reduce shed SARS-CoV-2 titers in ferrets and failed to block virus transmission to untreated direct-contact ferrets, whereas transmission was fully suppressed in a group of animals treated with a human-equivalent dose of molnupiravir. Prophylactic administration of molnupiravir to uninfected ferrets in direct contact with infected animals blocked productive SARS-CoV-2 transmission, whereas all contacts treated with prophylactic paxlovid became infected. These data confirm retrospective reports of similar therapeutic benefit of both drugs for older adults, and reveal that treatment with molnupiravir, but not paxlovid, may be suitable to reduce the risk of SARS-CoV-2 transmission.
Collapse
|
192
|
Kodchakorn K, Kongtawelert P. Molecular dynamics study on the strengthening behavior of Delta and Omicron SARS-CoV-2 spike RBD improved receptor-binding affinity. PLoS One 2022; 17:e0277745. [PMID: 36395151 PMCID: PMC9671323 DOI: 10.1371/journal.pone.0277745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
The COVID-19 pandemic caused by a virus that can be transmitted from human to human via air droplets has changed the quality of life and economic systems all over the world. The viral DNA has mutated naturally over time leading to the diversity of coronavirus victims which has posed a serious threat to human security on a massive scale. The current variants have developed in a dominant way and are considered "Variants of Concern" by the World Health Organization (WHO). In this work, Kappa (B.1.617.1), Delta (B.1.617.2), and Omicron (B.1.1.529) variants were obtained to evaluate whether naturally occurring mutations have strengthened viral infectivity. We apply reliable in silico structural dynamics and energetic frameworks of the mutated S-RBD protein for ACE2-binding to analyze and compare the structural information related to the wild-type. In particular, the hotspot residues at Q493, Q498, and N501 on the S-RBD protein were determined as contributing factors to the employment stability of the relevant binding interface. The L452R mutation induces an increment of the hydrogen bonds formed by changing the Q493 environment for ACE2 binding. Moreover, the Q493K exchange in Omicron enables the formation of two additional salt bridges, leading to a strong binding affinity by increased electrostatic interaction energy. These results could be used in proposing concrete informative data for a structure-based design engaged in finding better therapeutics against novel variants.
Collapse
Affiliation(s)
- Kanchanok Kodchakorn
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- * E-mail:
| |
Collapse
|
193
|
Gupta AM, Chakrabarti J. Effect on the conformations of the spike protein of SARS-CoV-2 due to mutation. Biotechnol Appl Biochem 2022:10.1002/bab.2413. [PMID: 36314068 PMCID: PMC9874481 DOI: 10.1002/bab.2413] [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: 05/23/2022] [Accepted: 10/23/2022] [Indexed: 01/27/2023]
Abstract
The spike protein of SARS-CoV-2 mediates receptor binding and cell entry and is the key immunogenic target for virus neutralization and the present attention of many vaccine layouts. It exhibits significant conformational flexibility. We study the structural fluctuations of spike protein among the most common mutations that appeared in the variant of concerns (VOC). We report the thermodynamics of conformational changes in mutant spike protein with respect to the wild-type from the distributions of the dihedral angles obtained from the equilibrium configurations generated via all-atom molecular dynamics simulations. We find that the mutation causes the increase in distance between the N-terminal domain and receptor binding domain, leading to an obtuse angle cosine θ distribution in the trimeric structure in spike protein. Thus, an increase in open state is conferred to the more infectious variants of SARS-CoV-2. The thermodynamically destabilized and disordered residues of receptor binding motif among the mutant variants of spike protein are proposed to serve as better binding sites for the host factor. We identify a short stretch of region connecting the N-terminal domain and receptor binding domain forming a linker loop where many residues undergo stabilization in the open state compared to the closed one.
Collapse
Affiliation(s)
- Aayatti Mallick Gupta
- Department of Chemical, Biological & Macro‐Molecular SciencesS.N. Bose National Centre for Basic SciencesKolkataWest BengalIndia
| | - Jaydeb Chakrabarti
- Department of Chemical, Biological & Macro‐Molecular SciencesS.N. Bose National Centre for Basic SciencesKolkataWest BengalIndia
| |
Collapse
|
194
|
Toledo-Romani ME, García-Carmenate M, Verdecia-Sánchez L, Pérez-Rodríguez S, Rodriguez-González M, Valenzuela-Silva C, Paredes-Moreno B, Sanchez-Ramirez B, González-Mugica R, Hernández-Garcia T, Orosa-Vázquez I, Díaz-Hernández M, Pérez-Guevara MT, Enriquez-Puertas J, Noa-Romero E, Palenzuela-Diaz A, Baro-Roman G, Mendoza-Hernández I, Muñoz Y, Gómez-Maceo Y, Santos-Vega BL, Fernandez-Castillo S, Climent-Ruiz Y, Rodríguez-Noda L, Santana-Mederos D, García-Vega Y, Chen GW, Doroud D, Biglari A, Boggiano-Ayo T, Valdés-Balbín Y, Rivera DG, García-Rivera D, Vérez-Bencomo V. Safety and immunogenicity of anti-SARS-CoV-2 heterologous scheme with SOBERANA 02 and SOBERANA Plus vaccines: Phase IIb clinical trial in adults. MED (NEW YORK, N.Y.) 2022; 3:760-773.e5. [PMID: 35998623 PMCID: PMC9359498 DOI: 10.1016/j.medj.2022.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/17/2022] [Accepted: 08/02/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND SOBERANA 02 has been evaluated in phase I and IIa studies comparing homologous versus heterologous schedule (this one, including SOBERANA Plus). Here, we report results of immunogenicity, safety, and reactogenicity of SOBERANA 02 in a two- or three-dose heterologous scheme in adults. METHOD Phase IIb was a parallel, multicenter, adaptive, double-blind, randomized, and placebo-controlled trial. Subjects (n = 810) aged 19-80 years were randomized to receive two doses of SARS-CoV-2 RBD conjugated to tetanus toxoid (SOBERANA 02) and a third dose of dimeric RBD (SOBERANA Plus) 28 days apart; two production batches of active ingredients of SOBERANA 02 were evaluated. Primary outcome was the percentage of seroconverted subjects with ≥4-fold the anti-RBD immunoglobulin G (IgG) concentration. Secondary outcomes were safety, reactogenicity, and neutralizing antibodies. FINDINGS Seroconversion rate in vaccinees was 76.3% after two doses and 96.8% after the third dose of SOBERANA Plus (7.3% in the placebo group). Neutralizing IgG antibodies were detected against D614G and variants of concern (VOCs) Alpha, Beta, Delta, and Omicron. Specific, functional antibodies were detected 7-8 months after the third dose. The frequency of serious adverse events (AEs) associated with vaccination was very low (0.1%). Local pain was the most frequent AE. CONCLUSIONS Two doses of SOBERANA 02 were safe and immunogenic in adults. The heterologous combination with SOBERANA Plus increased neutralizing antibodies, detectable 7-8 months after the third dose. TRIAL REGISTRY https://rpcec.sld.cu/trials/RPCEC00000347 FUNDING: This work was supported by Finlay Vaccine Institute, BioCubaFarma, and the Fondo Nacional de Ciencia y Técnica (FONCI-CITMA-Cuba, contract 2020-20).
Collapse
Affiliation(s)
| | - Mayra García-Carmenate
- "19 de Abril" Polyclinic, Tulipan St. between Panorama y Oeste, Nuevo Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | | | - Suzel Pérez-Rodríguez
- "19 de Abril" Polyclinic, Tulipan St. between Panorama y Oeste, Nuevo Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | | | - Carmen Valenzuela-Silva
- Cybernetics, Mathematics and Physics Institute, 15th St. #55, Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | - Beatriz Paredes-Moreno
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | | | - Raúl González-Mugica
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Tays Hernández-Garcia
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | - Ivette Orosa-Vázquez
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | | | | | | | - Enrique Noa-Romero
- National Civil Defense Research Laboratory, San José de las Lajas, Mayabeque, Cuba
| | | | - Gerardo Baro-Roman
- Centre for Immunoassays, 134 St. and 25, Cubanacán, Playa, Havana 11600 Cuba
| | - Ivis Mendoza-Hernández
- National Clinical Trials Coordinating Center, 5th Avenue and 62, Miramar, Playa, Havana, Cuba
| | - Yaima Muñoz
- National Clinical Trials Coordinating Center, 5th Avenue and 62, Miramar, Playa, Havana, Cuba
| | | | - Bertha Leysi Santos-Vega
- "19 de Abril" Polyclinic, Tulipan St. between Panorama y Oeste, Nuevo Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | - Sonsire Fernandez-Castillo
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba,Corresponding author
| | - Yanet Climent-Ruiz
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Laura Rodríguez-Noda
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Darielys Santana-Mederos
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Yanelda García-Vega
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | - Guang-Wu Chen
- Chengdu Olisynn Biotech. Co. Ltd., Chengdu 610041, People’s Republic of China,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Delaram Doroud
- Pasteur Institute of Iran, No. 69, Pasteur Avenue, Tehran 1316943551, Islamic Republic of Iran
| | - Alireza Biglari
- Pasteur Institute of Iran, No. 69, Pasteur Avenue, Tehran 1316943551, Islamic Republic of Iran
| | - Tammy Boggiano-Ayo
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | - Yury Valdés-Balbín
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Daniel G. Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Havana 10400, Cuba
| | - Dagmar García-Rivera
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Vicente Vérez-Bencomo
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba,Corresponding author
| | | |
Collapse
|
195
|
Yu C, Fengzhao Z, Hongmei W, Zeyuan L, Yu L, Yuhang G, Rufei S, Qingzhu J, Xiaorong S, Xia W, Caiping S, Zhi X, Chunmei L. The impact of vaccination on patients with COVID-19 during the wave of Omicron in Shanghai. Front Public Health 2022; 10:1054313. [PMID: 36438303 PMCID: PMC9682288 DOI: 10.3389/fpubh.2022.1054313] [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: 09/26/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
Background The global health has been affected by the COVID-19 pandemic persistently, of which Omicron is currently the predominant variant. However, the impact of vaccination on Omicron remained uncertain. Objective This study sought to explore the effect of vaccination on patients infected with Omicron. Methods A retrospective observational cohort was conducted in the largest Fangcang shelter hospital in Shanghai from April 1 to May 30, 2022. The demographics, length of hospital stay, clinical symptoms, the comorbidities and vaccination status were recorded. Clinical outcomes of the vaccinated and non-vaccinated groups were compared and analyzed. Results Of the 3,119 patients who fulfilled the eligibility criteria and were enrolled in the study, 2,226 (71.4%) patients had received nCoV-19 vaccine while 893 (28.6%) patients had not received it before admission. Patients in the vaccinated group had significantly shorter length of hospital stay than those in the unvaccinated group (15.48 ± 2.708 vs. 15.85 ± 3.102, p < 0.001). More asymptomatic patients were observed in the vaccinated group than the non-vaccinated (70.4 vs. 64.5%, p < 0.001). Further subgroup analysis demonstrated that the older the age, the more significant the difference was (p < 0.005). Conclusions Vaccination was associated with a significant reduction in the severity of Omicron infection compared with no vaccination. Vaccination appears to make Omicron-infected people with milder symptoms than unvaccinated people. This suggests the potential effectiveness of current vaccines against Omicron.
Collapse
Affiliation(s)
- Chen Yu
- Department of Orthopaedics, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Zhu Fengzhao
- Department of Orthopaedics, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Wu Hongmei
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Lei Zeyuan
- Department of Plastic Surgery, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Liu Yu
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Guo Yuhang
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Shen Rufei
- Department of Endocrinology, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Jia Qingzhu
- Department of Oncology, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Sun Xiaorong
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Wang Xia
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Song Caiping
- Department of Office of the Hospital, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Xu Zhi
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Luo Chunmei
- Department of Orthopaedics, Xinqiao Hospital, The Army Medical University, Chongqing, China
| |
Collapse
|
196
|
Vanoni S, Matulevicius A, Avdiu B, Scantamburlo G, Ulekleiv C, Brzoska PM, Furtado MR, Feenstra JDM, Rico A, Gandhi M, Giacobazzi E, Masi E, Paulmichl M, Nofziger C. SARS-CoV-2 variants of concern surveillance including Omicron using RT-PCR–based genotyping offers comparable performance to whole genome sequencing. Front Cell Infect Microbiol 2022; 12:960065. [PMID: 36405967 PMCID: PMC9670535 DOI: 10.3389/fcimb.2022.960065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
Known SARS-CoV-2 variants of concern (VOCs) can be detected and differentiated using an RT-PCR–based genotyping approach, which offers quicker time to result, lower cost, higher flexibility, and use of the same laboratory instrumentation for detection of SARS-CoV-2 when compared with whole genome sequencing (WGS). In the current study, we demonstrate how we applied a genotyping approach for identification of all VOCs and that such technique can offer comparable performance to WGS for identification of known SARS-CoV-2 VOCs, including more recent strains, Omicron BA.1 and BA.2.
Collapse
Affiliation(s)
- Simone Vanoni
- PharmGenetix GmbH, Niederalm/Anif, Austria
- *Correspondence: Simone Vanoni,
| | | | | | | | - Camilla Ulekleiv
- Thermo Fisher Scientific, South San Francisco, CA, United States
| | - Pius M. Brzoska
- Thermo Fisher Scientific, South San Francisco, CA, United States
| | | | | | - Alain Rico
- Thermo Fisher Scientific, South San Francisco, CA, United States
| | - Manoj Gandhi
- Thermo Fisher Scientific, South San Francisco, CA, United States
| | - Elisabetta Giacobazzi
- Azienda sanitaria dell’Alto Adige, Laboratorio di microbiologia e virologia, Bolzano, Italy
| | - Elisa Masi
- Azienda sanitaria dell’Alto Adige, Laboratorio di microbiologia e virologia, Bolzano, Italy
| | - Markus Paulmichl
- Department of Personalised Medicine, Privatklinik Maria Hilf GmbH, Klagenfurt, Austria
| | | |
Collapse
|
197
|
Yang F, Jiang XL, Tariq A, Sadia S, Ahmed Z, Sardans J, Aleem M, Ullah R, Bussmann RW. Potential medicinal plants involved in inhibiting 3CL pro activity: A practical alternate approach to combating COVID-19. JOURNAL OF INTEGRATIVE MEDICINE 2022; 20:488-496. [PMID: 35985974 PMCID: PMC9359926 DOI: 10.1016/j.joim.2022.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/29/2022] [Indexed: 01/25/2023]
Abstract
At present, a variety of vaccines have been approved, and existing antiviral drugs are being tested to find an effective treatment for coronavirus disease 2019 (COVID-19). However, no standardized treatment has yet been approved by the World Health Organization. The virally encoded chymotrypsin-like protease (3CLpro) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which facilitates the replication of SARS-CoV in the host cells, is one potential pharmacological target for the development of anti-SARS drugs. Online search engines, such as Web of Science, Google Scholar, Scopus and PubMed, were used to retrieve data on the traditional uses of medicinal plants and their inhibitory effects against the SARS-CoV 3CLpro. Various pure compounds, including polyphenols, terpenoids, chalcones, alkaloids, biflavonoids, flavanones, anthraquinones and glycosides, have shown potent inhibition of SARS-CoV-2 3CLpro activity with 50% inhibitory concentration (IC50) values ranging from 2-44 µg/mL. Interestingly, most of these active compounds, including xanthoangelol E (isolated from Angelica keiskei), dieckol 1 (isolated from Ecklonia cava), amentoflavone (isolated from Torreya nucifera), celastrol, pristimerin, tingenone and iguesterin (isolated from Tripterygium regelii), tannic acid (isolated from Camellia sinensis), and theaflavin-3,3'-digallate, 3-isotheaflav1in-3 gallate and dihydrotanshinone I (isolated from Salvia miltiorrhiza), had IC50 values of less than 15 µg/mL. Kinetic mechanistic studies of several active compounds revealed that their mode of inhibition was dose-dependent and competitive, with Ki values ranging from 2.4-43.8 μmol/L. Given the significance of plant-based compounds and the many promising results obtained, there is still need to explore the phytochemical and mechanistic potentials of plants and their products. These medicinal plants could serve as an effective inexpensive nutraceutical for the general public to help manage COVID-19.
Collapse
Affiliation(s)
- Fan Yang
- The Medical Center of General Practice, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu 610041, Sichuan Province, China
| | - Xiao-Lan Jiang
- The Medical Center of General Practice, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu 610041, Sichuan Province, China.
| | - Akash Tariq
- Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830041, Xinjiang Uygur Autonomous Region, China.
| | - Sehrish Sadia
- Department of Biological Sciences, University of Veterinary and Animal Sciences, Ravi Campus, Pattoki 55300, Pakistan
| | - Zeeshan Ahmed
- Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830041, Xinjiang Uygur Autonomous Region, China
| | - Jordi Sardans
- Consejo Superior de Investigaciones Científicas, Global Ecology Unit, Centre for Ecological Research and Forestry Applications-Consejo Superior de Investigaciones Científicas-Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Catalonia, Spain; Centre for Ecological Research and Forestry Applications, Cerdanyola del Vallès 08193, Catalonia, Spain
| | - Muhammad Aleem
- Department of Biological Sciences, University of Veterinary and Animal Sciences, Ravi Campus, Pattoki 55300, Pakistan
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rainer W Bussmann
- Department of Ethnobotany, Institute of Botany, llia State University, Tbilisi 0105, Georgia
| |
Collapse
|
198
|
Pacheco-Olvera DL, Saint Remy-Hernández S, García-Valeriano MG, Rivera-Hernández T, López-Macías C. Bioinformatic Analysis of B- and T-cell Epitopes from SARS-CoV-2 Structural Proteins and their Potential Cross-reactivity with Emerging Variants and other Human Coronaviruses. Arch Med Res 2022; 53:694-710. [PMID: 36336501 PMCID: PMC9633039 DOI: 10.1016/j.arcmed.2022.10.007] [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: 02/25/2022] [Revised: 08/23/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The mutations in SARS-CoV-2 variants of concern (VOC) facilitate the virus' escape from the neutralizing antibodies induced by vaccines. However, the protection from hospitalization and death is not significantly diminished. Both vaccine boosters and infection improve immune responses and provide protection, suggesting that conserved and/or cross-reactive epitopes could be involved. While several important T- and B-cell epitopes have been identified, mainly in the S protein, the M and N proteins and their potential cross-reactive epitopes with other coronaviruses remain largely unexplored. AIMS To identify and map new potential B- and T-cell epitopes within the SARS-CoV-2 S, M and N proteins, as well as cross-reactive epitopes with human coronaviruses. METHODS Different bioinformatics tools were used to: i) Identify new and compile previously-reported B-and T-cell epitopes from SARS-CoV-2 S, M and N proteins; ii) Determine the mutations in S protein from VOC that affect B- and T-cell epitopes, and; iii) Identify cross-reactive epitopes with coronaviruses relevant to human health. RESULTS New, potential B- and T-cell epitopes from S, M and N proteins as well as cross-reactive epitopes with other coronaviruses were found and mapped within the proteins' structures. CONCLUSION Numerous potential B- and T-cell epitopes were found in S, M and N proteins, some of which are conserved between coronaviruses. VOCs present mutations within important epitopes in the S protein; however, a significant number of other epitopes remain unchanged. The epitopes identified here may contribute to augmenting the protective response to SARS-CoV-2 and its variants induced by infection and/or vaccination, and may also be used for the rational design of novel broad-spectrum coronavirus vaccines.
Collapse
Affiliation(s)
- Diana Laura Pacheco-Olvera
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Stephanie Saint Remy-Hernández
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - María Guadalupe García-Valeriano
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Tania Rivera-Hernández
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México; Consejo Nacional de Ciencia y Tecnología, Ciudad de México, México
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| |
Collapse
|
199
|
Bhadane R, Salo-Ahen OMH. High-Throughput Molecular Dynamics-Based Alchemical Free Energy Calculations for Predicting the Binding Free Energy Change Associated with the Selected Omicron Mutations in the Spike Receptor-Binding Domain of SARS-CoV-2. Biomedicines 2022; 10:2779. [PMID: 36359299 PMCID: PMC9687918 DOI: 10.3390/biomedicines10112779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 11/10/2023] Open
Abstract
The ongoing pandemic caused by SARS-CoV-2 has gone through various phases. Since the initial outbreak, the virus has mutated several times, with some lineages showing even stronger infectivity and faster spread than the original virus. Among all the variants, omicron is currently classified as a variant of concern (VOC) by the World Health Organization, as the previously circulating variants have been replaced by it. In this work, we have focused on the mutations observed in omicron sub lineages BA.1, BA.2, BA.4 and BA.5, particularly at the receptor-binding domain (RBD) of the spike protein that is responsible for the interactions with the host ACE2 receptor and binding of antibodies. Studying such mutations is particularly important for understanding the viral infectivity, spread of the disease and for tracking the escape routes of this virus from antibodies. Molecular dynamics (MD) based alchemical free energy calculations have been shown to be very accurate in predicting the free energy change, due to a mutation that could have a deleterious or a stabilizing effect on either the protein itself or its binding affinity to another protein. Here, we investigated the significance of five spike RBD mutations on the stability of the spike protein binding to ACE2 by free energy calculations using high throughput MD simulations. For comparison, we also used conventional MD simulations combined with a Molecular Mechanics-Generalized Born Surface Area (MM-GBSA) based approach, and compared our results with the available experimental data. Overall, the alchemical free energy calculations performed far better than the MM-GBSA approach in predicting the individual impact of the mutations. When considering the experimental variation, the alchemical free energy method was able to produce a relatively accurate prediction for N501Y, the mutant that has previously been reported to increase the binding affinity to hACE2. On the other hand, the other individual mutations seem not to have a significant effect on the spike RBD binding affinity towards hACE2.
Collapse
Affiliation(s)
- Rajendra Bhadane
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, FI-20520 Turku, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, FI-20520 Turku, Finland
| | - Outi M. H. Salo-Ahen
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, FI-20520 Turku, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, FI-20520 Turku, Finland
| |
Collapse
|
200
|
Igari H, Asano H, Murata S, Yoshida T, Kawasaki K, Kageyama T, Ikeda K, Koshikawa H, Okuda Y, Urushihara M, Chiba H, Yahaba M, Taniguchi T, Matsushita K, Yoshino I, Yokote K, Nakajima H. Antibody responses and SARS-CoV-2 infection after BNT162b2 mRNA booster vaccination among healthcare workers in Japan. J Infect Chemother 2022; 28:1483-1488. [PMID: 35870791 PMCID: PMC9296373 DOI: 10.1016/j.jiac.2022.07.010] [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: 06/02/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Vaccine effectiveness against SARS-CoV-2 infections decreases due to waning immunity, and booster vaccination was therefore introduced. We estimated the anti-spike antibody (AS-ab) recovery by booster vaccination and analyzed the risk factors for SARS-CoV-2 infections. METHODS The subjects were health care workers (HCWs) in a Chiba University Hospital vaccination cohort. They had received two doses of vaccine (BNT162b2) and a booster vaccine (BNT162b2). We retrospectively analyzed AS-ab titers and watched out for SARS-CoV-2 infection for 90 days following booster vaccination. RESULTS AS-ab titer eight months after two-dose vaccinations had decreased to as low as 587 U/mL (median, IQR (interquartile range) 360-896). AS-ab titer had then increased to 22471 U/mL (15761-32622) three weeks after booster vaccination. There were no significant differences among age groups. A total of 1708 HCWs were analyzed for SARS-CoV-2 infection, and 48 of them proved positive. SARS-CoV-2 infections in the booster-vaccinated and non-booster groups were 1.8% and 4.0%, respectively, and were not significant. However, when restricted to those 20-29 years old, SARS-CoV-2 infections in the booster-vaccinated and non-booster groups were 2.9% and 13.6%, respectively (p = 0.04). After multivariate logistic regression, COVID-19 wards (adjusted odds ratio (aOR):2.9, 95% confidence interval (CI) 1.5-5.6) and those aged 20-49 years (aOR:9.7, 95%CI 1.3-71.2) were risk factors for SARS-CoV-2 infection. CONCLUSIONS Booster vaccination induced the recovery of AS-ab titers. Risk factors for SARS-CoV-2 infection were HCWs of COVID-19 wards and those aged 20-49 years. Increased vaccination coverage, together with implementing infection control, remains the primary means of preventing HCWs from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hidetoshi Igari
- Department of Infection Control, Chiba University Hospital, Japan; COVID-19 Vaccine Center, Chiba University Hospital, Japan.
| | - Haruna Asano
- Division of Laboratory Medicine, Chiba University Hospital, Japan.
| | - Shota Murata
- Division of Laboratory Medicine, Chiba University Hospital, Japan.
| | | | - Kenji Kawasaki
- Division of Laboratory Medicine, Chiba University Hospital, Japan.
| | - Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Key Ikeda
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Hiromi Koshikawa
- Department of Infection Control, Chiba University Hospital, Japan.
| | - Yoshio Okuda
- Department of Infection Control, Chiba University Hospital, Japan.
| | - Misao Urushihara
- Department of Infection Control, Chiba University Hospital, Japan
| | - Hitoshi Chiba
- Department of Infection Control, Chiba University Hospital, Japan.
| | - Misuzu Yahaba
- Department of Infection Control, Chiba University Hospital, Japan.
| | | | | | - Ichiro Yoshino
- Department of General Thoracic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Hiroshi Nakajima
- COVID-19 Vaccine Center, Chiba University Hospital, Japan; Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|