51
|
Sahni C, Basu Roy Chowdhury P, Devadas D, Ashish A, Singh NK, Yadav A, Kaur M, Mishra S, Vishwakarma S, Singh R. SARS-CoV-2 Mutations Responsible for Immune Evasion Leading to Breakthrough Infection. Cureus 2022; 14:e29544. [PMID: 36312656 PMCID: PMC9592688 DOI: 10.7759/cureus.29544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES India had faced a devastating second outbreak of COVID-19 infection, in which a majority of the viral sequences were found to be of the B.1.617.2 lineage (Delta-variant). While India and the world focused on vaccination, reports of vaccine-immunity evasion by the virus, termed "breakthrough cases", emerged worldwide. Our study was focused on the primary objective to identify the mutations associated with breakthrough infections SARS-CoV-2. METHODS In our study, we extracted the SARS-CoV-2 RNA (ribonucleic acid) from reverse transcription-polymerase chain reaction (RT-PCR) positive COVID-19 patients, and 150 random samples were sent for sequencing to the Centre for Cellular & Molecular Biology, Hyderabad. Whole genome sequences of 150 SARS-CoV-2 viral samples were analyzed thoroughly. We mostly found B.1.617 and its sub-lineages in the genomic sequencing results. RESULTS AND INTERPRETATION On further analysis of patient data, it was seen that nine patients had been vaccinated against the SARS-CoV-2 previously. These nine patients had B.1.617/B.1 or A strains, and all of them had similar genomic variations in spike proteins as well as non-structural proteins (NSPs). The mutations seen in these sequences in the Spike (S), NSPs, and open reading frame (ORF) regions would have produced amino acid changes known to improve viral replication, confer drug resistance, influence host-cell interaction, and lead to antigenic drift. CONCLUSIONS Increased virulence culminating in vaccine immunity evasion may be inferred from these specific mutations. Our study adds to the growing body of evidence linking rapidly emerging mutations in the S (Spike) and ORF genes of the SARS-CoV-2 genome to immune evasion.
Collapse
Affiliation(s)
- Chetan Sahni
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | | | - Deepa Devadas
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | - Ashish Ashish
- Multidisciplinary Research Unit/Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | - Nitish K Singh
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | - Abhay Yadav
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | - Manpreet Kaur
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | - Shivani Mishra
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | - Shani Vishwakarma
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| | - Royana Singh
- Anatomy, Institute of Medical Sciences (IMS) Banaras Hindu University (BHU), Varanasi, IND
| |
Collapse
|
52
|
Meng B, Datir R, Choi J, Bradley JR, Smith KGC, Lee JH, Gupta RK. SARS-CoV-2 spike N-terminal domain modulates TMPRSS2-dependent viral entry and fusogenicity. Cell Rep 2022; 40:111220. [PMID: 35963244 PMCID: PMC9346021 DOI: 10.1016/j.celrep.2022.111220] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/30/2022] [Accepted: 07/22/2022] [Indexed: 11/28/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike N-terminal domain (NTD) remains poorly characterized despite enrichment of mutations in this region across variants of concern (VOCs). Here, we examine the contribution of the NTD to infection and cell-cell fusion by constructing chimeric spikes bearing B.1.617 lineage (Delta and Kappa variants) NTDs and generating spike pseudotyped lentivirus. We find that the Delta NTD on a Kappa or wild-type (WT) background increases S1/S2 cleavage efficiency and virus entry, specifically in lung cells and airway organoids, through use of TMPRSS2. Delta exhibits increased cell-cell fusogenicity that could be conferred to WT and Kappa spikes by Delta NTD transfer. However, chimeras of Omicron BA.1 and BA.2 spikes with a Delta NTD do not show more efficient TMPRSS2 use or fusogenicity. We conclude that the NTD allosterically modulates S1/S2 cleavage and spike-mediated functions in a spike context-dependent manner, and allosteric interactions may be lost when combining regions from more distantly related VOCs.
Collapse
Affiliation(s)
- Bo Meng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Rawlings Datir
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jinwook Choi
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - John R Bradley
- Department of Medicine, University of Cambridge, Cambridge, UK; NIHR Bioresource, Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Joo Hyeon Lee
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Africa Health Research Institute, Durban, South Africa.
| |
Collapse
|
53
|
Tsuchiya K, Yamamoto N, Hosaka Y, Wakita M, Hiki M, Matsushita Y, Mori H, Hori S, Misawa S, Miida T, Nojiri S, Takahashi K, Naito T, Tabe Y. Molecular characterization of SARS-CoV-2 detected in Tokyo, Japan during five waves: Identification of the amino acid substitutions associated with transmissibility and severity. Front Microbiol 2022; 13:912061. [PMID: 35966679 PMCID: PMC9363898 DOI: 10.3389/fmicb.2022.912061] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/29/2022] [Indexed: 11/15/2022] Open
Abstract
Many variants of SARS-CoV-2 have emerged around the world. It is therefore important to understand its global viral evolution and the corresponding mutations associated with transmissibility and severity. In this study, we analyzed 112 whole genome sequences of SARS-CoV-2 collected from patients at Juntendo University Hospital in Tokyo and the genome data from entire Japan deposited in Global Initiative on Sharing Avian Influenza Data (GISAID) to examine the relationship of amino acid changes with the transmissibility and the severity of each strain/lineage. We identified 12 lineages, including B.1.1.284, B.1.1.214, R.1, AY.29, and AY.29.1, which were prevalent specifically in Japan. B.1.1.284 was most frequently detected in the second wave, but B.1.1.214 became the predominant lineage in the third wave, indicating that B.1.1.214 has a higher transmissibility than B.1.1.284. The most prevalent lineage during the fourth and fifth wave was B.1.1.7 and AY.29, respectively. In regard to the severity of identified lineages, B.1.1.214 was significantly lower than the reference lineage, B.1.1.284. Analysis of the genome sequence and other traits of each lineage/strain revealed the mutations in S, N, and NSPs that increase the transmissibility and/or severity. These mutations include S: M153T, N: P151L, NSP3: S543P, NSP5: P108S, and NSP12: A423V in B.1.1.284; S: W152L and E484K in R.1; S: H69del, V70del, and N501Y in the Alpha strain; S: L452R, T478K, and P681R in the Delta strain. Furthermore, it is suggested that the transmissibility of B.1.1.214 could be enhanced by the mutations N: M234I, NSP14: P43L, and NSP16: R287I. To address the issue of the virus evolution, it is necessary to continuously monitor the genomes of SARS-CoV-2 and analyze the effects of mutations for developing vaccines and antiviral drugs effective against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Koji Tsuchiya
- Department of Clinical Laboratory, Juntendo University Hospital, Bunkyo, Tokyo, Japan
| | - Norio Yamamoto
- Department of General Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
- Department of Microbiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
- Department of Microbiology, Tokai University School of Medicine, Isehara, Japan
- *Correspondence: Norio Yamamoto, ;
| | - Yoshie Hosaka
- Department of Clinical Laboratory, Juntendo University Hospital, Bunkyo, Tokyo, Japan
| | - Mitsuru Wakita
- Department of Clinical Laboratory, Juntendo University Hospital, Bunkyo, Tokyo, Japan
| | - Makoto Hiki
- Department of Emergency Medicine, Juntendo University Faculty of Medicine, Bunkyo-ku, Japan
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Yasushi Matsushita
- Department of Internal Medicine and Rheumatology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Hirotake Mori
- Department of General Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Satoshi Hori
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
- Department of Infection Control Science, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Shigeki Misawa
- Department of Clinical Laboratory, Juntendo University Hospital, Bunkyo, Tokyo, Japan
| | - Takashi Miida
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Bunkyo-ku, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Bunkyo-ku, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Bunkyo-ku, Japan
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| |
Collapse
|
54
|
Grikscheit K, Rabenau HF, Ghodratian Z, Widera M, Wilhelm A, Toptan Grabmair T, Hoehl S, Layer E, Helfritz F, Ciesek S. Characterization of the Antibody and Interferon-Gamma Release Response after a Second COVID-19 Booster Vaccination. Vaccines (Basel) 2022; 10:vaccines10071163. [PMID: 35891326 PMCID: PMC9323888 DOI: 10.3390/vaccines10071163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/28/2022] [Accepted: 07/14/2022] [Indexed: 11/26/2022] Open
Abstract
The emergence of SARS-CoV-2 Omicron subvariants prompted countries to call for accelerated booster vaccinations to limit disease and transmission. Here, we characterized correlates of protection over time after the second booster or after Omicron BA.1 infection comparing variants of concern (VOCs). Sera from subjects before and two and seven weeks after the second booster or after Omicron infection were examined for the level of Spike receptor-binding-domain (RBD)-specific antibodies. Furthermore, neutralizing antibodies (nABs) were characterized in in vitro neutralization assays comparing the variants of concern Alpha, Beta, Delta, and Omicron BA.1 and BA.2 against the ancestral strain B.1. Here, the second booster resulted in an increase in anti-RBD-IgG-antibodies, remaining nearly constant over time, accompanied by an increase in nABs against B.1 and the VOCs Alpha, Beta, Delta, and Omicron BA.1 and BA.2. However, compared to B.1, the neutralizing capacity against the Omicron subvariants remained low and was limited after the second booster vaccination. This indicates that antibody-mediated protection against infection with this VOC is unlikely, as evidenced by the fact that three individuals of our study cohort became infected with Omicron BA.1 after the second booster. T cell activation was measured by interferon-gamma release assays in a subgroup of subjects and was increased in all subjects tested after the second booster vaccination, correlating with the amount of Spike-specific antibodies. In subjects with Omicron BA.1 breakthrough infection, a significant increase in nABs to all VOCs studied was observed independently of booster vaccinations. Taken together, our data indicate that a second booster or Omicron BA.1 infection mediate a substantial increase in anti-Spike IgG antibodies; however, infection with Omicron BA.1 induced a stronger increase in neutralizing antibodies against the different VOCs
Collapse
Affiliation(s)
- Katharina Grikscheit
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
| | - Holger F. Rabenau
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
| | - Zahra Ghodratian
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
| | - Alexander Wilhelm
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
| | - Tuna Toptan Grabmair
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
| | - Sebastian Hoehl
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
| | - Emily Layer
- Health Protection Authority of the City of Frankfurt am Main, 60313 Frankfurt am Main, Germany;
| | - Fabian Helfritz
- Bürgerhospital Frankfurt, Nibelungenallee 37-41, 60318 Frankfurt am Main, Germany;
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany; (K.G.); (H.F.R.); (Z.G.); (M.W.); (A.W.); (T.T.G.); (S.H.)
- German Centre for Infection Research (DZIF), Partner Site Frankfurt, 60596 Frankfurt, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmcology, 60596 Frankfurt am Main, Germany
- Correspondence:
| |
Collapse
|
55
|
Jiang W, Zhang Z, Zhu Y, Chen B, Gu C, Liu Z, Zhang X, Xiong H, Zhang Y, Zheng B, Wang R, Jiao S, Wang A, Zhang T, Zhang J, Wang S, Zhang B, Li G, Gui X. Pre-Clinical Development of a Potent Neutralizing Antibody MW3321 With Extensive SARS-CoV-2 Variants Coverage. Front Pharmacol 2022; 13:926750. [PMID: 35873586 PMCID: PMC9304585 DOI: 10.3389/fphar.2022.926750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Since the outbreak of the coronavirus disease 2019 (COVID-19) pandemic, several variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged and have consistently replaced the previous dominant variant. Therapeutics against variants of SARS-CoV-2 are urgently needed. Ideal SARS-CoV-2 therapeutic antibodies would have high potency in viral neutralization against several emerging variants. Neutralization antibodies targeting SARS-CoV-2 could provide immediate protection after SARS-CoV-2 infection, especially for the most vulnerable populations. In this work, we comprehensively characterize the breadth and efficacy of SARS-CoV-2 RBD-targeting fully human monoclonal antibody (mAb) MW3321. MW3321 retains full neutralization activity to all tested 12 variants that have arisen in the human population, which are assigned as VOC (Variants of Concern) and VOI (Variants of Interest) due to their impacts on public health. Escape mutation experiments using replicating SARS-CoV-2 pseudovirus show that escape mutants were not generated until passage 6 for MW3321, which is much more resistant to escape mutation compared with another clinical staged SARS-CoV-2 neutralizing mAb MW3311. MW3321 could effectively reduce viral burden in hACE2-transgenic mice challenged with either wild-type or Delta SARS-CoV-2 strains through viral neutralization and Fc-mediated effector functions. Moreover, MW3321 exhibits a typical hIgG1 pharmacokinetic and safety profile in cynomolgus monkeys. These data support the development of MW3321 as a monotherapy or cocktail against SARS-CoV-2-related diseases.
Collapse
Affiliation(s)
- Wen Jiang
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Zherui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yuhe Zhu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| | - Ben Chen
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Chunying Gu
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Zhiyan Liu
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Xukai Zhang
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Hualong Xiong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| | - Yanan Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Bin Zheng
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Rongjuan Wang
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
- Beijing Kohnoor Science and Technology Co., Ltd., Beijing, China
| | - Shasha Jiao
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
- Beijing Kohnoor Science and Technology Co., Ltd., Beijing, China
| | - An Wang
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Tianying Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| | - Jinchao Zhang
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
| | - Shuang Wang
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
- Beijing Kohnoor Science and Technology Co., Ltd., Beijing, China
- *Correspondence: Shuang Wang, ; Bo Zhang, ; Gang Li, ; Xun Gui,
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Shuang Wang, ; Bo Zhang, ; Gang Li, ; Xun Gui,
| | - Gang Li
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
- *Correspondence: Shuang Wang, ; Bo Zhang, ; Gang Li, ; Xun Gui,
| | - Xun Gui
- Mabwell (Shanghai) Bioscience Co., Ltd., Shanghai, China
- *Correspondence: Shuang Wang, ; Bo Zhang, ; Gang Li, ; Xun Gui,
| |
Collapse
|
56
|
Pondé RAA. Physicochemical effect of the N501Y, E484K/Q, K417N/T, L452R and T478K mutations on the SARS-CoV-2 spike protein RBD and its influence on agent fitness and on attributes developed by emerging variants of concern. Virology 2022; 572:44-54. [PMID: 35580380 PMCID: PMC9096574 DOI: 10.1016/j.virol.2022.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/26/2022] [Accepted: 05/06/2022] [Indexed: 01/17/2023]
Abstract
The spike protein comprises one of the main structural components of SARS-CoV-2 because it is directly involved in the infection process and viral transmission, and also because of its immunogenic properties, as an inducer of the protective antibodies production and as a vaccine component. The occurrence of mutations in this region or in other the virus genome regions, comprises a natural phenomenon in its evolution. However, they also occur due to the selective immune pressure, to which the agent is continuously subjected, especially in the spike protein immunodominant regions, such as the RBD. Mutations in the spike protein can change the virus' fitness, increasing its affinity for target cells, its transmissibility and its virulence. In addition, these mutations can giving it the potential ability to evade the protective antibodies action obtained from convalescent sera or vaccine origin, as well as those used in therapy, which may favor the virus expansion and compromise the infection control. Five mutations N501Y, E484K/Q, K417N/T, L452R and T478K, located in the spike protein RBD, have had a greater impact because they are associated with new attributes developed by the virus, which characterize the emerging variants of concern (VOCs) of SARS-Cov-2 identified so far. The occurrence of these mutations induces complex physicochemical effects that can alter the spike protein's structure and its function, which in turn, lead to changes in the agents' fitness. This manuscript discusses the attributes of VOCs associated with the physicochemical effects caused by the aforementioned mutations.
Collapse
Affiliation(s)
- R A A Pondé
- State Department of Health SES/Superintendence of Health Surveillance SUVISA/GO, Management of Epidemiological Surveillance-GVE/Coordination of Analysis and Research-CAP, Goiânia, Goiás, Brazil; Laboratory of Human Virology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
57
|
Neutralisation sensitivity of SARS-CoV-2 omicron subvariants to therapeutic monoclonal antibodies. THE LANCET. INFECTIOUS DISEASES 2022; 22:942-943. [PMID: 35690075 PMCID: PMC9179126 DOI: 10.1016/s1473-3099(22)00365-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 01/01/2023]
|
58
|
Mishra T, Dalavi R, Joshi G, Kumar A, Pandey P, Shukla S, Mishra RK, Chande A. SARS-CoV-2 spike E156G/Δ157-158 mutations contribute to increased infectivity and immune escape. Life Sci Alliance 2022; 5:e202201415. [PMID: 35296517 PMCID: PMC8927725 DOI: 10.26508/lsa.202201415] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Breakthrough infections by emerging SARS-CoV-2 variants raise significant concerns. Here, we sequence-characterized the spike gene from breakthrough infections that corresponded to B.1.617 sublineage. Delineating the functional impact of spike mutations revealed that N-terminal domain (NTD)-specific E156G/Δ157-158 contributed to increased infectivity and reduced sensitivity to vaccine-induced antibodies. A six-nucleotide deletion (467-472) in the spike-coding region introduced this change in the NTD. We confirmed the presence of E156G/Δ157-158 from cases concurrently screened, in addition to other circulating spike (S1) mutations such as T19R, T95I, L452R, E484Q, and D614G. Notably, E156G/Δ157-158 was present in more than 90% of the sequences reported from the USA and UK in October 2021. The spike-pseudotyped viruses bearing a combination of E156G/Δ157-158 and L452R exhibited higher infectivity and reduced sensitivity to neutralization. Notwithstanding, the post-recovery plasma robustly neutralized viral particles bearing the mutant spike. When the spike harbored E156G/Δ157-158 along with L452R and E484Q, increased cell-to-cell fusion was also observed, suggesting a combinatorial effect of these mutations. Our study underscores the importance of non-RBD changes in determining infectivity and immune escape.
Collapse
Affiliation(s)
- Tarun Mishra
- Molecular Virology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| | - Rishikesh Dalavi
- Molecular Virology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| | - Garima Joshi
- Sumo and Nuclear Pore Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| | - Atul Kumar
- Structural Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
- COVID-19 Testing Centre, Indian Institute of Science Education and Research, Bhopal, India
| | - Pankaj Pandey
- COVID-19 Testing Centre, Indian Institute of Science Education and Research, Bhopal, India
| | - Sanjeev Shukla
- COVID-19 Testing Centre, Indian Institute of Science Education and Research, Bhopal, India
- Epigenetics and RNA Processing Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| | - Ram K Mishra
- Sumo and Nuclear Pore Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
- COVID-19 Testing Centre, Indian Institute of Science Education and Research, Bhopal, India
| | - Ajit Chande
- Molecular Virology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
- COVID-19 Testing Centre, Indian Institute of Science Education and Research, Bhopal, India
| |
Collapse
|
59
|
Markarian NM, Galli G, Patel D, Hemmings M, Nagpal P, Berghuis AM, Abrahamyan L, Vidal SM. Identifying Markers of Emerging SARS-CoV-2 Variants in Patients With Secondary Immunodeficiency. Front Microbiol 2022; 13:933983. [PMID: 35847101 PMCID: PMC9283111 DOI: 10.3389/fmicb.2022.933983] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 12/03/2022] Open
Abstract
Since the end of 2019, the world has been challenged by the coronavirus disease 2019 (COVID-19) pandemic. With COVID-19 cases rising globally, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve, resulting in the emergence of variants of interest (VOI) and of concern (VOC). Of the hundreds of millions infected, immunodeficient patients are one of the vulnerable cohorts that are most susceptible to this virus. These individuals include those with preexisting health conditions and/or those undergoing immunosuppressive treatment (secondary immunodeficiency). In these cases, several researchers have reported chronic infections in the presence of anti-COVID-19 treatments that may potentially lead to the evolution of the virus within the host. Such variations occurred in a variety of viral proteins, including key structural ones involved in pathogenesis such as spike proteins. Tracking and comparing such mutations with those arisen in the general population may provide information about functional sites within the SARS-CoV-2 genome. In this study, we reviewed the current literature regarding the specific features of SARS-CoV-2 evolution in immunocompromised patients and identified recurrent de novo amino acid changes in virus isolates of these patients that can potentially play an important role in SARS-CoV-2 pathogenesis and evolution.
Collapse
Affiliation(s)
- Nathan M. Markarian
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
- Swine and Poultry Infectious Diseases Research Center and Research Group on Infectious Diseases in Production Animals, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Gaël Galli
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- CNRS, ImmunoConcEpT, UMR 5164, Université de Bordeaux, Bordeaux, France
- CHU de Bordeaux, FHU ACRONIM, Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares Est/Sud-Ouest, Bordeaux, France
| | - Dhanesh Patel
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
| | - Mark Hemmings
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Priya Nagpal
- Department of Pharmacology, McGill University, Montréal, QC, Canada
| | | | - Levon Abrahamyan
- Swine and Poultry Infectious Diseases Research Center and Research Group on Infectious Diseases in Production Animals, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Silvia M. Vidal
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
| |
Collapse
|
60
|
Matsui Y, Li L, Prahl M, Cassidy AG, Ozarslan N, Golan Y, Gonzalez VJ, Lin CY, Jigmeddagva U, Chidboy MA, Montano M, Taha TY, Khalid MM, Sreekumar B, Hayashi JM, Chen PY, Kumar GR, Warrier L, Wu AH, Song D, Jegatheesan P, Rai DS, Govindaswami B, Needens J, Rincon M, Myatt L, Asiodu IV, Flaherman VJ, Afshar Y, Jacoby VL, Murtha AP, Robinson JF, Ott M, Greene WC, Gaw SL. Neutralizing antibody activity against SARS-CoV-2 variants in gestational age-matched mother-infant dyads after infection or vaccination. JCI Insight 2022; 7:e157354. [PMID: 35579965 PMCID: PMC9309042 DOI: 10.1172/jci.insight.157354] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Pregnancy confers unique immune responses to infection and vaccination across gestation. To date, there are limited data comparing vaccine- and infection-induced neutralizing Abs (nAbs) against COVID-19 variants in mothers during pregnancy. We analyzed paired maternal and cord plasma samples from 60 pregnant individuals. Thirty women vaccinated with mRNA vaccines (from December 2020 through August 2021) were matched with 30 naturally infected women (from March 2020 through January 2021) by gestational age of exposure. Neutralization activity against the 5 SARS-CoV-2 spike sequences was measured by a SARS-CoV-2-pseudotyped spike virion assay. Effective nAbs against SARS-CoV-2 were present in maternal and cord plasma after both infection and vaccination. Compared with WT spike protein, these nAbs were less effective against the Delta and Mu spike variants. Vaccination during the third trimester induced higher cord-nAb levels at delivery than did infection during the third trimester. In contrast, vaccine-induced nAb levels were lower at the time of delivery compared with infection during the first trimester. The transfer ratio (cord nAb level divided by maternal nAb level) was greatest in mothers vaccinated in the second trimester. SARS-CoV-2 vaccination or infection in pregnancy elicits effective nAbs with differing neutralization kinetics that are influenced by gestational time of exposure.
Collapse
Affiliation(s)
- Yusuke Matsui
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
| | - Lin Li
- Division of Maternal-Fetal Medicine and
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, California, USA
| | - Mary Prahl
- Department of Pediatrics
- Division of Pediatric Infectious Diseases and Global Health
| | | | - Nida Ozarslan
- Division of Maternal-Fetal Medicine and
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, California, USA
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences
| | | | | | - Unurzul Jigmeddagva
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, California, USA
| | - Megan A. Chidboy
- Division of Maternal-Fetal Medicine and
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, California, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
| | - Taha Y. Taha
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
| | - Mir M. Khalid
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
| | - Bharath Sreekumar
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
| | - Jennifer M. Hayashi
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
| | - Pei-Yi Chen
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
| | - G. Renuka Kumar
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
| | | | - Alan H.B. Wu
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Dongli Song
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Priya Jegatheesan
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Daljeet S. Rai
- Stanford-O’Connor Family Medicine Residency Program, Division of Family Medicine, Stanford University, Palo Alto, California, USA
| | | | - Jordan Needens
- Department of Obstetrics and Gynecology, Marshall University Joan C. Edwards School of Medicine, Huntington, West Virginia, USA
| | - Monica Rincon
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | - Yalda Afshar
- Department of Obstetrics and Gynecology, UCLA, Los Angeles, California, USA
| | | | | | - Joshua F. Robinson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, California, USA
| | - Melanie Ott
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Department of Medicine, and
| | - Warner C. Greene
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, California, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Department of Medicine, and
- Department of Microbiology and Immunology, UCSF, San Francisco, California, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine and
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, California, USA
| |
Collapse
|
61
|
Kemp SA, Cheng MTK, Hamilton WL, Kamelian K, Singh S, Rakshit P, Agrawal A, Illingworth CJR, Gupta RK. Transmission of B.1.617.2 Delta variant between vaccinated healthcare workers. Sci Rep 2022; 12:10492. [PMID: 35729228 PMCID: PMC9212198 DOI: 10.1038/s41598-022-14411-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/07/2022] [Indexed: 01/01/2023] Open
Abstract
Breakthrough infections with SARS-CoV-2 Delta variant have been reported in doubly-vaccinated recipients and as re-infections. Studies of viral spread within hospital settings have highlighted the potential for transmission between doubly-vaccinated patients and health care workers and have highlighted the benefits of high-grade respiratory protection for health care workers. However the extent to which vaccination is preventative of viral spread in health care settings is less well studied. Here, we analysed data from 118 vaccinated health care workers (HCW) across two hospitals in India, constructing two probable transmission networks involving six HCWs in Hospital A and eight HCWs in Hospital B from epidemiological and virus genome sequence data, using a suite of computational approaches. A maximum likelihood reconstruction of transmission involving known cases of infection suggests a high probability that doubly vaccinated HCWs transmitted SARS-CoV-2 between each other and highlights potential cases of virus transmission between individuals who had received two doses of vaccine. Our findings show firstly that vaccination may reduce rates of transmission, supporting the need for ongoing infection control measures even in highly vaccinated populations, and secondly we have described a novel approach to identifying transmissions that is scalable and rapid, without the need for an infection control infrastructure.
Collapse
Affiliation(s)
- Steven A Kemp
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Mark T K Cheng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
| | | | - Kimia Kamelian
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | | | - Sujit Singh
- National Centre for Disease Control, Delhi, India
| | | | - Anurag Agrawal
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Christopher J R Illingworth
- Garscube Campus, MRC - University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow, UK. .,MRC Biostatistics Unit, University of Cambridge, East Forvie Building, Forvie Site, Robinson Way, Cambridge, UK. .,Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK. .,Department of Medicine, University of Cambridge, Cambridge, UK. .,Africa Health Research Institute, Durban, South Africa. .,Jeffrey Cheah Biomedical Centre, Cambridge, CB5 8UB, UK.
| |
Collapse
|
62
|
Dutta D, Naiyer S, Mansuri S, Soni N, Singh V, Bhat KH, Singh N, Arora G, Mansuri MS. COVID-19 Diagnosis: A Comprehensive Review of the RT-qPCR Method for Detection of SARS-CoV-2. Diagnostics (Basel) 2022; 12:diagnostics12061503. [PMID: 35741313 PMCID: PMC9221722 DOI: 10.3390/diagnostics12061503] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022] Open
Abstract
The world is grappling with the coronavirus disease 2019 (COVID-19) pandemic, the causative agent of which is severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 symptoms are similar to the common cold, including fever, sore throat, cough, muscle and chest pain, brain fog, dyspnoea, anosmia, ageusia, and headache. The manifestation of the disease can vary from being asymptomatic to severe life-threatening conditions warranting hospitalization and ventilation support. Furthermore, the emergence of mutecated variants of concern (VOCs) is paramount to the devastating effect of the pandemic. This highly contagious virus and its emergent variants challenge the available advanced viral diagnostic methods for high-accuracy testing with faster result yields. This review is to shed light on the natural history, pathology, molecular biology, and efficient diagnostic methods of COVID-19, detecting SARS-CoV-2 in collected samples. We reviewed the gold standard RT-qPCR method for COVID-19 diagnosis to confer a better understanding and application to combat the COVID-19 pandemic. This comprehensive review may further develop awareness about the management of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Debashis Dutta
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: (D.D.); (M.S.M.)
| | - Sarah Naiyer
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60616, USA;
| | | | - Neeraj Soni
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Vandana Singh
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Khalid Hussain Bhat
- SKUAST Kashmir, Division of Basic Science and Humanities, Faculty of Agriculture, Wadura Sopore 193201, JK, India;
| | - Nishant Singh
- Cell and Gene Therapy Absorption System, Exton, PA 19335, USA;
| | - Gunjan Arora
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - M. Shahid Mansuri
- Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Correspondence: (D.D.); (M.S.M.)
| |
Collapse
|
63
|
Ortega MA, García-Montero C, Fraile-Martinez O, Colet P, Baizhaxynova A, Mukhtarova K, Alvarez-Mon M, Kanatova K, Asúnsolo A, Sarría-Santamera A. Recapping the Features of SARS-CoV-2 and Its Main Variants: Status and Future Paths. J Pers Med 2022; 12:995. [PMID: 35743779 PMCID: PMC9225183 DOI: 10.3390/jpm12060995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/06/2022] [Accepted: 06/16/2022] [Indexed: 12/14/2022] Open
Abstract
Over the two years that we have been experiencing the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) pandemic, our challenges have been the race to develop vaccines and the difficulties in fighting against new variants due to the rapid ability of the virus to evolve. In this sense, different organizations have identified and classified the different variants that have been emerging, distinguishing between variants of concern (VOC), variants of interest (VOI), or variants under monitoring (VUM). The following review aims to describe the latest updates focusing on VOC and already de-escalated variants, as well as to describe the impact these have had on the global situation. Understanding the intrinsic properties of SARS-CoV-2 and its interaction with the immune system and vaccination is essential to make out the underlying mechanisms that have led to the appearance of these variants, helping to determine the next steps for better public management of this pandemic.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Paolo Colet
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Ardak Baizhaxynova
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Kymbat Mukhtarova
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Kaznagul Kanatova
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Angel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Antonio Sarría-Santamera
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| |
Collapse
|
64
|
Development of a simple genotyping method based on indel mutations to rapidly screen SARS-CoV-2 circulating variants: Delta, Omicron BA.1 and BA.2. J Virol Methods 2022; 307:114570. [PMID: 35724698 PMCID: PMC9212420 DOI: 10.1016/j.jviromet.2022.114570] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/24/2022]
Abstract
The high need of rapid and flexible tools that facilitate the identification of circulating SARS-CoV-2 Variants of Concern (VOCs) remains crucial for public health system monitoring. Here, we develop allele-specific (AS)-qPCR assays targeting three recurrent indel mutations, ΔEF156–157, Ins214EPE and ΔLPP24–26, in spike (S) gene to identify the Delta VOC and the Omicron sublineages BA.1 and BA.2, respectively. After verification of the analytical specificity of each primer set, two duplex qPCR assays with melting curve analysis were performed to screen 129 COVID-19 cases confirmed between December 31, 2021 and February 01, 2022 in Sfax, Tunisia. The first duplex assay targeting ΔEF156–157 and Ins214EPE mutations successfully detected the Delta VOC in 39 cases and Omicron BA.1 in 83 cases. All the remaining cases (n = 7) were identified as Omicron BA.2, by the second duplex assay targeting Ins214EPE and ΔLPP24–26 mutations. The results of the screening method were in perfect concordance with those of S gene partial sequencing. In conclusion, our findings provide a simple and flexible screening method for more rapid and reliable monitoring of circulating VOCs. We highly recommend its implementation to guide public health policies.
Collapse
|
65
|
Yamasoba D, Kimura I, Nasser H, Morioka Y, Nao N, Ito J, Uriu K, Tsuda M, Zahradnik J, Shirakawa K, Suzuki R, Kishimoto M, Kosugi Y, Kobiyama K, Hara T, Toyoda M, Tanaka YL, Butlertanaka EP, Shimizu R, Ito H, Wang L, Oda Y, Orba Y, Sasaki M, Nagata K, Yoshimatsu K, Asakura H, Nagashima M, Sadamasu K, Yoshimura K, Kuramochi J, Seki M, Fujiki R, Kaneda A, Shimada T, Nakada TA, Sakao S, Suzuki T, Ueno T, Takaori-Kondo A, Ishii KJ, Schreiber G, Sawa H, Saito A, Irie T, Tanaka S, Matsuno K, Fukuhara T, Ikeda T, Sato K. Virological characteristics of the SARS-CoV-2 Omicron BA.2 spike. Cell 2022; 185:2103-2115.e19. [PMID: 35568035 DOI: 10.1101/2022.02.14.480335] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 04/26/2022] [Indexed: 05/23/2023]
Abstract
Soon after the emergence and global spread of the SARS-CoV-2 Omicron lineage BA.1, another Omicron lineage, BA.2, began outcompeting BA.1. The results of statistical analysis showed that the effective reproduction number of BA.2 is 1.4-fold higher than that of BA.1. Neutralization experiments revealed that immunity induced by COVID vaccines widely administered to human populations is not effective against BA.2, similar to BA.1, and that the antigenicity of BA.2 is notably different from that of BA.1. Cell culture experiments showed that the BA.2 spike confers higher replication efficacy in human nasal epithelial cells and is more efficient in mediating syncytia formation than the BA.1 spike. Furthermore, infection experiments using hamsters indicated that the BA.2 spike-bearing virus is more pathogenic than the BA.1 spike-bearing virus. Altogether, the results of our multiscale investigations suggest that the risk of BA.2 to global health is potentially higher than that of BA.1.
Collapse
Affiliation(s)
- Daichi Yamasoba
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Faculty of Medicine, Kobe University, Kobe, Japan
| | - Izumi Kimura
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hesham Nasser
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan; Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Yuhei Morioka
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naganori Nao
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Jumpei Ito
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Keiya Uriu
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Jiri Zahradnik
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Kotaro Shirakawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mai Kishimoto
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yusuke Kosugi
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Laboratory of Systems Virology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kouji Kobiyama
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Teppei Hara
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mako Toyoda
- Division of Infection and immunity, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan
| | - Yuri L Tanaka
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Erika P Butlertanaka
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Ryo Shimizu
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan
| | - Hayato Ito
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Kayoko Nagata
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | - Mami Nagashima
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | - Kenji Sadamasu
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | | | | | - Motoaki Seki
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryoji Fujiki
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tadanaga Shimada
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Taka-Aki Nakada
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takamasa Ueno
- Division of Infection and immunity, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken J Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Hirofumi Sawa
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; One Health Research Center, Hokkaido University, Sapporo, Japan; Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Chiba, Japan; Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan.
| | - Takashi Irie
- Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan.
| | - Keita Matsuno
- One Health Research Center, Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan.
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; CREST, Japan Science and Technology Agency, Kawaguchi, Japan.
| |
Collapse
|
66
|
Yamasoba D, Kimura I, Nasser H, Morioka Y, Nao N, Ito J, Uriu K, Tsuda M, Zahradnik J, Shirakawa K, Suzuki R, Kishimoto M, Kosugi Y, Kobiyama K, Hara T, Toyoda M, Tanaka YL, Butlertanaka EP, Shimizu R, Ito H, Wang L, Oda Y, Orba Y, Sasaki M, Nagata K, Yoshimatsu K, Asakura H, Nagashima M, Sadamasu K, Yoshimura K, Kuramochi J, Seki M, Fujiki R, Kaneda A, Shimada T, Nakada TA, Sakao S, Suzuki T, Ueno T, Takaori-Kondo A, Ishii KJ, Schreiber G, Sawa H, Saito A, Irie T, Tanaka S, Matsuno K, Fukuhara T, Ikeda T, Sato K. Virological characteristics of the SARS-CoV-2 Omicron BA.2 spike. Cell 2022; 185:2103-2115.e19. [PMID: 35568035 PMCID: PMC9057982 DOI: 10.1016/j.cell.2022.04.035] [Citation(s) in RCA: 199] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 04/26/2022] [Indexed: 12/30/2022]
Abstract
Soon after the emergence and global spread of the SARS-CoV-2 Omicron lineage BA.1, another Omicron lineage, BA.2, began outcompeting BA.1. The results of statistical analysis showed that the effective reproduction number of BA.2 is 1.4-fold higher than that of BA.1. Neutralization experiments revealed that immunity induced by COVID vaccines widely administered to human populations is not effective against BA.2, similar to BA.1, and that the antigenicity of BA.2 is notably different from that of BA.1. Cell culture experiments showed that the BA.2 spike confers higher replication efficacy in human nasal epithelial cells and is more efficient in mediating syncytia formation than the BA.1 spike. Furthermore, infection experiments using hamsters indicated that the BA.2 spike-bearing virus is more pathogenic than the BA.1 spike-bearing virus. Altogether, the results of our multiscale investigations suggest that the risk of BA.2 to global health is potentially higher than that of BA.1.
Collapse
Affiliation(s)
- Daichi Yamasoba
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Faculty of Medicine, Kobe University, Kobe, Japan
| | - Izumi Kimura
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hesham Nasser
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan; Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Yuhei Morioka
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naganori Nao
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Jumpei Ito
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Keiya Uriu
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Jiri Zahradnik
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Kotaro Shirakawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mai Kishimoto
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yusuke Kosugi
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Laboratory of Systems Virology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kouji Kobiyama
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Teppei Hara
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mako Toyoda
- Division of Infection and immunity, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan
| | - Yuri L Tanaka
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Erika P Butlertanaka
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Ryo Shimizu
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan
| | - Hayato Ito
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Kayoko Nagata
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | - Mami Nagashima
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | - Kenji Sadamasu
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | | | | | - Motoaki Seki
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryoji Fujiki
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tadanaga Shimada
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Taka-Aki Nakada
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takamasa Ueno
- Division of Infection and immunity, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken J Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Hirofumi Sawa
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; One Health Research Center, Hokkaido University, Sapporo, Japan; Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Chiba, Japan; Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan.
| | - Takashi Irie
- Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan.
| | - Keita Matsuno
- One Health Research Center, Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto, Japan.
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; CREST, Japan Science and Technology Agency, Kawaguchi, Japan.
| |
Collapse
|
67
|
Chakraborty C, Sharma AR, Bhattacharya M, Agoramoorthy G, Lee SS. A Paradigm Shift in the Combination Changes of SARS-CoV-2 Variants and Increased Spread of Delta Variant (B.1.617.2) across the World. Aging Dis 2022; 13:927-942. [PMID: 35656100 PMCID: PMC9116911 DOI: 10.14336/ad.2021.1117] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Since September 2020, the SARS-CoV-2 variants have gained their dominance worldwide, especially in Kenya, Italy, France, the UK, Turkey, Indonesia, India, Finland, Ireland, Singapore, Denmark, Germany, and Portugal. In this study, we developed a model on the frequency of delta variants across 28 countries (R2= 0.1497), displaying the inheritance of mutations during the generation of the delta variants with 123,526 haplotypes. The country-wise haplotype network showed the distribution of haplotypes in USA (10,174), Denmark (5,637), India (4,089), Germany (2,350), Netherlands (1,899), Sweden (1,791), Italy (1,720), France (1,293), Ireland (1,257), Belgium (1,207), Singapore (1,193), Portugal (1,184) and Spain (1,133). Our analysis shows the highest haplotype in Europe with 84% and the lowest in Australia with 0.00001%. A model of scatter plot was generated with a regression line which provided the estimated rate of mutation, including 24.048 substitutions yearly. Our study concluded that the high global prevalence of the delta variants is due to a high frequency of infectivity, supporting the paradigm shift of the viral variants.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- 1Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Ashish Ranjan Sharma
- 2Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Gangwon-do, Korea
| | | | | | - Sang-Soo Lee
- 2Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Gangwon-do, Korea
| |
Collapse
|
68
|
Greaney AJ, Eguia RT, Starr TN, Khan K, Franko N, Logue JK, Lord SM, Speake C, Chu HY, Sigal A, Bloom JD. The SARS-CoV-2 Delta variant induces an antibody response largely focused on class 1 and 2 antibody epitopes. PLoS Pathog 2022; 18:e1010592. [PMID: 35767821 PMCID: PMC9275729 DOI: 10.1371/journal.ppat.1010592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/12/2022] [Accepted: 05/15/2022] [Indexed: 12/23/2022] Open
Abstract
Exposure histories to SARS-CoV-2 variants and vaccinations will shape the specificity of antibody responses. To understand the specificity of Delta-elicited antibody immunity, we characterize the polyclonal antibody response elicited by primary or mRNA vaccine-breakthrough Delta infections. Both types of infection elicit a neutralizing antibody response focused heavily on the receptor-binding domain (RBD). We use deep mutational scanning to show that mutations to the RBD's class 1 and class 2 epitopes, including sites 417, 478, and 484-486 often reduce binding of these Delta-elicited antibodies. The anti-Delta antibody response is more similar to that elicited by early 2020 viruses than the Beta variant, with mutations to the class 1 and 2, but not class 3 epitopes, having the largest effects on polyclonal antibody binding. In addition, mutations to the class 1 epitope (e.g., K417N) tend to have larger effects on antibody binding and neutralization in the Delta spike than in the D614G spike, both for vaccine- and Delta-infection-elicited antibodies. These results help elucidate how the antigenic impacts of SARS-CoV-2 mutations depend on exposure history.
Collapse
Affiliation(s)
- Allison J Greaney
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Genome Sciences & Medical Scientist Training Program, University of Washington, Seattle, Washington, United States of America
| | - Rachel T Eguia
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Tyler N Starr
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Khadija Khan
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Nicholas Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Jennifer K Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Sandra M Lord
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - Helen Y Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jesse D Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| |
Collapse
|
69
|
Feng W, Xiang Y, Wu L, Chen Z, Li Q, Chen J, Guo Y, Xia D, Chen N, Zhang L, Zhu S, Zhao K. Nucleocapsid protein of SARS-CoV-2 is a potential target for developing new generation of vaccine. J Clin Lab Anal 2022; 36:e24479. [PMID: 35527696 PMCID: PMC9169192 DOI: 10.1002/jcla.24479] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/28/2022] [Accepted: 04/23/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND SARS-CoV-2 has spread worldwide causing more than 400 million people with virus infections since early 2020. Currently, the existing vaccines targeting the spike glycoprotein (S protein) of SARS-CoV-2 are facing great challenge from the infection of SARS-CoV-2 virus and its multiple S protein variants. Thus, we need to develop a new generation of vaccines to prevent infection of the SARS-CoV-2 variants. Compared with the S protein, the nucleocapsid protein (N protein) of SARS-CoV-2 is more conservative and less mutations, which also plays a vital role in viral infection. Therefore, the N protein may have the great potential for developing new vaccines. METHODS The N protein of SARS-CoV-2 was recombinantly expressed and purified in Escherichia coli. Western Blot and ELISA assays were used to demonstrate the immunoreactivity of the recombinant N protein with the serum of 22 COVID-19 patients. We investigated further the response of the specific serum antibodies and cytokine production in BALB/c mice immunized with recombinant N protein by Western Blot and ELISA. RESULTS The N protein had good immunoreactivity and the production of IgG antibody against N protein in COVID-19 patients was tightly correlated with disease severity. Furthermore, the N protein was used to immunize BALB/c mice to have elicited strong immune responses. Not only high levels of IgG antibody, but also cytokine-IFN-γ were produced in the N protein-immunized mice. Importantly, the N protein immunization induced a high level of IgM antibody produced in the mice. CONCLUSION SARS-CoV-2 N protein shows a great big bundle of potentiality for developing a new generation of vaccines in fighting infection of SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Weixu Feng
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Yunru Xiang
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Lianpeng Wu
- Department of Laboratory MedicineThe Sixth People Hospital of WenzhouWenzhouChina
| | - Zhuo Chen
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Qingfeng Li
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Jun Chen
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Yanru Guo
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Dandan Xia
- Department of Laboratory MedicineThe Sixth People Hospital of WenzhouWenzhouChina
| | - Na Chen
- Department of Laboratory MedicineThe Sixth People Hospital of WenzhouWenzhouChina
| | - Lifang Zhang
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Shanli Zhu
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Kong‐Nan Zhao
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
- Department of Obstetrics and GynaecologyThe Second Affiliated Hospital and Yuyin Children Hospital of Wenzhou Medical UniversityWenzhouChina
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt LuciaQueenslandAustralia
| |
Collapse
|
70
|
Hoter A, Naim HY. Biochemical Characterization of SARS-CoV-2 Spike RBD Mutations and Their Impact on ACE2 Receptor Binding. Front Mol Biosci 2022; 9:893843. [PMID: 35677879 PMCID: PMC9168323 DOI: 10.3389/fmolb.2022.893843] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Infection of mammalian cells by SARS-CoV-2 coronavirus requires primary interaction between the receptor binding domain (RBD) of the viral spike protein and the host cell surface receptor angiotensin-converting enzyme 2 (ACE2) glycoprotein. Several mutations in the RBD of SARS-CoV-2 spike protein have been reported for several variants and resulted in wide spread of the COVID pandemic. For instance, the double mutations L452R and E484Q present in the Indian B.1.617 variant have been suggested to cause evasion of the host immune response. The common RBD mutations N501Y and E484K were found to enhance the interaction with the ACE2 receptor. In the current study, we analyzed the biosynthesis and secretion of the RBD double mutants L452R and E484Q in comparison to the wild-type RBD and the individual mutations N501 and E484K in mammalian cells. Moreover, we evaluated the interaction of these variants with ACE2 by means of expression of the S protein and co-immunoprecipitation with ACE2. Our results revealed that the double RBD mutations L452R and E484Q resulted in a higher expression level and secretion of spike S1 protein than other mutations. In addition, an increased interaction of these mutant forms with ACE2 in Calu3 cells was observed. Altogether, our findings highlight the impact of continuous S1 mutations on the pathogenicity of SARS-CoV-2 and provide further biochemical evidence for the dominance and high transmissibility of the double Indian mutations.
Collapse
Affiliation(s)
- Abdullah Hoter
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hassan Y. Naim
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- *Correspondence: Hassan Y. Naim,
| |
Collapse
|
71
|
Wang Y, Wang P, Qin J. Human Organoids and Organs-on-Chips for Addressing COVID-19 Challenges. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105187. [PMID: 35107217 PMCID: PMC8981475 DOI: 10.1002/advs.202105187] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/15/2022] [Indexed: 05/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses an imminent threat to our lives. Although animal models and monolayer cell cultures are utilized for pathogenesis studies and the development of COVID-19 therapeutics, models that can more accurately reflect human-relevant responses to this novel virus are still lacking. Stem cell organoids and bioengineered organs-on-chips have emerged as two cutting-edge technologies used to construct biomimetic in vitro three-dimensional (3D) tissue or organ models. In this review, the key features of these two model systems that allow them to recapitulate organ physiology and function are introduced. The recent progress of these technologies for virology research is summarized and their utility in meeting the COVID-19 pandemic is highlighted. Future opportunities and challenges in the development of advanced human organ models and their potential to accelerate translational applications to provide vaccines and therapies for COVID-19 and other emerging epidemics are also discussed.
Collapse
Affiliation(s)
- Yaqing Wang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Peng Wang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Jianhua Qin
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- Beijing Institute For Stem Cell and Regeneration MedicineBeijing100101China
- CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
72
|
Ghosh A, Walia S, Rattan R, Kanampalliwar A, Jha A, Aggarwal S, Fatma S, Das N, Chayani N, Prasad P, Raghav SK, Parida A. Genomic profiles of vaccine breakthrough SARS-CoV-2 strains from Odisha, India. Int J Infect Dis 2022; 119:111-113. [PMID: 35358727 PMCID: PMC8958730 DOI: 10.1016/j.ijid.2022.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/07/2022] [Accepted: 03/22/2022] [Indexed: 10/26/2022] Open
Affiliation(s)
- Arup Ghosh
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | - Safal Walia
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | - Roma Rattan
- SCB medical college and hospital, Cuttack, Odisha, India
| | - Amol Kanampalliwar
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | - Atimukta Jha
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | - Shifu Aggarwal
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | - Sana Fatma
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | - Niyati Das
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | | | - Punit Prasad
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India
| | - Sunil K Raghav
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India.
| | - Ajay Parida
- Institute of Life Sciences, Autonomous Institute under Department of Biotechnology, Government of India, Bhubaneswar, Odisha, India.
| |
Collapse
|
73
|
Wong TY, Horspool AM, Russ BP, Ye C, Lee KS, Winters MT, Bevere JR, Miller OA, Rader NA, Cooper M, Kieffer T, Sourimant J, Greninger AL, Plemper RK, Denvir J, Cyphert HA, Barbier M, Torrelles JB, Martinez I, Martinez-Sobrido L, Damron FH. Evaluating Antibody Mediated Protection against Alpha, Beta, and Delta SARS-CoV-2 Variants of Concern in K18-hACE2 Transgenic Mice. J Virol 2022; 96:e0218421. [PMID: 35080423 PMCID: PMC8941865 DOI: 10.1128/jvi.02184-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 12/04/2022] Open
Abstract
SARS-CoV-2 variants of concern (VoC) are impacting responses to the COVID-19 pandemic. Here, we utilized passive immunization using human convalescent plasma (HCP) obtained from a critically ill COVID-19 patient in the early pandemic to study the efficacy of polyclonal antibodies generated to ancestral SARS-CoV-2 against the Alpha, Beta, and Delta VoC in the K18 human angiotensin converting enzyme 2 (hACE2) transgenic mouse model. HCP protected mice from challenge with the original WA-1 SARS-CoV-2 strain; however, only partially protected mice challenged with the Alpha VoC (60% survival) and failed to save Beta challenged mice from succumbing to disease. HCP treatment groups had elevated receptor binding domain (RBD) and nucleocapsid IgG titers in the serum; however, Beta VoC viral RNA burden in the lung and brain was not decreased due to HCP treatment. While mice could be protected from WA-1 or Alpha challenge with a single dose of HCP, six doses of HCP could not decrease mortality of Delta challenged mice. Overall, these data demonstrate that VoC have enhanced immune evasion and this work underscores the need for in vivo models to evaluate future emerging strains. IMPORTANCE Emerging SARS-CoV-2 VoC are posing new problems regarding vaccine and monoclonal antibody efficacy. To better understand immune evasion tactics of the VoC, we utilized passive immunization to study the effect of early-pandemic SARS-CoV-2 HCP against, Alpha, Beta, and Delta VoC. We observed that HCP from a human infected with the original SARS-CoV-2 was unable to control lethality of Alpha, Beta, or Delta VoC in the K18-hACE2 transgenic mouse model of SARS-CoV-2 infection. Our findings demonstrate that passive immunization can be used as a model to evaluate immune evasion of emerging VoC strains.
Collapse
Affiliation(s)
- Ting Y. Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Alexander M. Horspool
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Brynnan P. Russ
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Chengjin Ye
- Host-Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Katherine S. Lee
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Michael T. Winters
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Justin R. Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Olivia A. Miller
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Nathaniel A. Rader
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Melissa Cooper
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Theodore Kieffer
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Julien Sourimant
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Alexander L. Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Richard K. Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - James Denvir
- Department of Biomedical Sciences, Marshall University, Huntington, West Virginia, USA
| | - Holly A. Cyphert
- Department of Biological Sciences, Marshall University, Huntington, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Jordi B. Torrelles
- Host-Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Ivan Martinez
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Cancer Institute, Morgantown, West Virginia, USA
| | - Luis Martinez-Sobrido
- Host-Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| |
Collapse
|
74
|
Cheng MH, Krieger JM, Banerjee A, Xiang Y, Kaynak B, Shi Y, Arditi M, Bahar I. Impact of new variants on SARS-CoV-2 infectivity and neutralization: A molecular assessment of the alterations in the spike-host protein interactions. iScience 2022; 25:103939. [PMID: 35194576 PMCID: PMC8851820 DOI: 10.1016/j.isci.2022.103939] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 01/31/2022] [Accepted: 02/14/2022] [Indexed: 12/29/2022] Open
Abstract
The emergence of SARS-CoV-2 variants necessitates rational assessment of their impact on the recognition and neutralization of the virus by the host cell. We present a comparative analysis of the interactions of Alpha, Beta, Gamma, and Delta variants with cognate molecules (ACE2 and/or furin), neutralizing nanobodies (Nbs), and monoclonal antibodies (mAbs) using in silico methods, in addition to Nb-binding assays. Our study elucidates the molecular origin of the ability of Beta and Delta variants to evade selected antibodies, such as REGN10933, LY-CoV555, B38, C105, or H11-H4, while being insensitive to others including REGN10987. Experiments confirm that nanobody Nb20 retains neutralizing activity against the Delta variant. The substitutions T478K and L452R in the Delta variant enhance associations with ACE2, whereas P681R promotes recognition by proteases, thus facilitating viral entry. The Ab-specific responses of variants highlight how full-atomic structure and dynamics analyses are required for assessing the response to newly emerging variants.
Collapse
Affiliation(s)
- Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James M. Krieger
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anupam Banerjee
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yufei Xiang
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Burak Kaynak
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yi Shi
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, and Biomedical Sciences, Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
75
|
Biswas B, Chattopadhyay S, Hazra S, Hansda AK, Goswami R. COVID-19 pandemic: the delta variant, T-cell responses, and the efficacy of developing vaccines. Inflamm Res 2022; 71:377-396. [PMID: 35292834 PMCID: PMC8923340 DOI: 10.1007/s00011-022-01555-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 12/15/2022] Open
Abstract
Background The mayhem COVID-19 that was ushered by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) was declared pandemic by the World Health Organization in March 2020. Since its initial outbreak in late 2019, the virus has affected hundreds of million adults in the world and killing millions in the process. After the approval of newly developed vaccines, severe challenges remain to manufacture and administer them to the adult population globally in quick time. However, we have witnessed several mutations of the virus leading to ‘waves’ of viral spread and mortality. WHO has categorized these mutations as variants of concern (VOCs) and variants of interest (VOIs). The mortality due to COVID-19 has also been associated with various comorbidities and improper immune response. This has created further complications in understanding the nature of the SARS-CoV2–host interaction that has fuelled doubts in the efficacy of the approved vaccines. Whether there is requirement of booster dose and whether the impending wave could affect the children are some of the hotly debated topics. Materials and Methods A systematic literature review of PubMed, Medline, Scopus, Google Scholar was utilized to understand the nature of Delta variant and how it alters our T-cell responses and cytokine production and neutralizes vaccine-generated antibodies.
Conclusion In this review, we discuss the variants of SARS-CoV2 with specific focus on the Delta variant. We also specifically review the T-cell response against the virus and bring a narrative of various factors that may hold the key to fight against this marauding virus.
Collapse
Affiliation(s)
- Biswajit Biswas
- School of Bioscience, IIT Kharagpur, Kharagpur, 721302, West Bengal, India
| | | | - Sayantee Hazra
- School of Bioscience, IIT Kharagpur, Kharagpur, 721302, West Bengal, India
| | | | - Ritobrata Goswami
- School of Bioscience, IIT Kharagpur, Kharagpur, 721302, West Bengal, India.
| |
Collapse
|
76
|
Greaney AJ, Eguia RT, Starr TN, Khan K, Franko N, Logue JK, Lord SM, Speake C, Chu HY, Sigal A, Bloom JD. The SARS-CoV-2 Delta variant induces an antibody response largely focused on class 1 and 2 antibody epitopes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.12.484088. [PMID: 35313588 PMCID: PMC8936118 DOI: 10.1101/2022.03.12.484088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Exposure histories to SARS-CoV-2 variants and vaccinations will shape the specificity of antibody responses. To understand the specificity of Delta-elicited antibody immunity, we characterize the polyclonal antibody response elicited by primary or mRNA vaccine-breakthrough Delta infections. Both types of infection elicit a neutralizing antibody response focused heavily on the receptor-binding domain (RBD). We use deep mutational scanning to show that mutations to the RBD's class 1 and class 2 epitopes, including sites 417, 478, and 484-486 often reduce binding of these Delta-elicited antibodies. The anti-Delta antibody response is more similar to that elicited by early 2020 viruses than the Beta variant, with mutations to the class 1 and 2, but not class 3 epitopes, having the largest effects on polyclonal antibody binding. In addition, mutations to the class 1 epitope (e.g., K417N) tend to have larger effects on antibody binding and neutralization in the Delta spike than in the D614G spike, both for vaccine- and Delta-infection-elicited antibodies. These results help elucidate how the antigenic impacts of SARS-CoV-2 mutations depend on exposure history.
Collapse
Affiliation(s)
- Allison J. Greaney
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center; Seattle, WA, USA
- Department of Genome Sciences & Medical Scientist Training Program, University of Washington; Seattle, WA, USA
| | - Rachel T. Eguia
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center; Seattle, WA, USA
| | - Tyler N. Starr
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center; Seattle, WA, USA
- Howard Hughes Medical Institute; Chevy Chase, MD, USA
| | - Khadija Khan
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu–Natal, Durban, South Africa
| | - Nicholas Franko
- Division of Allergy and Infectious Diseases, University of Washington; Seattle, WA, USA
| | - Jennifer K. Logue
- Division of Allergy and Infectious Diseases, University of Washington; Seattle, WA, USA
| | - Sandra M. Lord
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason
| | - Helen Y. Chu
- Division of Allergy and Infectious Diseases, University of Washington; Seattle, WA, USA
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu–Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center; Seattle, WA, USA
- Howard Hughes Medical Institute; Chevy Chase, MD, USA
| |
Collapse
|
77
|
Gerdol M, Dishnica K, Giorgetti A. Emergence of a recurrent insertion in the N-terminal domain of the SARS-CoV-2 spike glycoprotein. Virus Res 2022; 310:198674. [PMID: 35021068 PMCID: PMC8743576 DOI: 10.1016/j.virusres.2022.198674] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
Tracking the evolution of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) through genomic surveillance programs is undoubtedly one of the key priorities in the current pandemic situation. Although the genome of SARS-CoV-2 acquires mutations at a slower rate compared with other RNA viruses, evolutionary pressures derived from the widespread circulation of SARS-CoV-2 in the human population have progressively favored the global emergence, though natural selection, of several variants of concern that carry multiple non-synonymous mutations in the spike glycoprotein. These are often placed in key sites within major antibody epitopes and may therefore confer resistance to neutralizing antibodies, leading to partial immune escape, or otherwise compensate infectivity deficits associated with other non-synonymous substitutions. As previously shown by other authors, several emerging variants carry recurrent deletion regions (RDRs) that display a partial overlap with antibody epitopes located in the spike N-terminal domain (NTD). Comparatively, very little attention had been directed towards spike insertion mutations prior to the emergence of the B.1.1.529 (omicron) lineage. This manuscript describes a single recurrent insertion region (RIR1) in the N-terminal domain of SARS-CoV-2 spike protein, characterized by at least 49 independent acquisitions of 1-8 additional codons between Val213 and Leu216 in different viral lineages. Even though RIR1 is unlikely to confer antibody escape, its association with two distinct formerly widespread lineages (A.2.5 and B.1.214.2), with the quickly spreading omicron and with other VOCs and VOIs warrants further investigation concerning its effects on spike structure and viral infectivity.
Collapse
Affiliation(s)
- Marco Gerdol
- University of Trieste, Department of Life Sciences, 34127 Trieste, Italy.
| | - Klevia Dishnica
- University of Verona, Department of Biotechnology, 37134 Verona, Italy
| | | |
Collapse
|
78
|
Lin L, Zhang J, Rogers J, Campbell A, Zhao J, Harding D, Sahr F, Liu Y, Wurie I. The dynamic change of SARS-CoV-2 variants in Sierra Leone. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 98:105208. [PMID: 34999288 PMCID: PMC8734169 DOI: 10.1016/j.meegid.2022.105208] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/24/2021] [Accepted: 01/03/2022] [Indexed: 10/31/2022]
Abstract
Since the beginning of the SARS-CoV-2 pandemic, the emergence of multiple new variants posed an increased risk to global public health. The aim of this study is to investigate SARS-CoV-2 variants and possible transmission of variants of concern (VOCs) in Sierra Leone. A total of 65 nasal swab samples were collected from COVID-19 cases in Sierra Leone, among which 24 samples were collected during the second wave and 41 samples were collected during the third wave. Nanopore sequencing generated 54 SARS-CoV-2 whole genomes. The second COVID-19 wave was mainly caused by R.1 lineage while the third COVID-19 wave was dominated by B.1.617.2 lineage (Delta variant). The phylogenetic analysis suggested multiple introductions of SARS-CoV-2 Delta variant into Sierra Leone and subsequent local transmission in this country. Our findings highlight the importance of genomic surveillance of SARS-CoV-2 variants and the urgent need for implementation of strengthened public health and social measures (PHSM) to control the spread of virus variants.
Collapse
Affiliation(s)
- Lei Lin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Juling Zhang
- Department of Clinical Laboratory, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - James Rogers
- College of Medicine and Allied Health Science, University of Sierra Leone, Freetown, Sierra Leone
| | - Allan Campbell
- Central Public Health Reference Laboratories, Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Jianjun Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Doris Harding
- Central Public Health Reference Laboratories, Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Foday Sahr
- College of Medicine and Allied Health Science, University of Sierra Leone, Freetown, Sierra Leone
| | - Yongjian Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China.
| | - Isata Wurie
- College of Medicine and Allied Health Science, University of Sierra Leone, Freetown, Sierra Leone
| |
Collapse
|
79
|
Meng B, Abdullahi A, Ferreira IATM, Goonawardane N, Saito A, Kimura I, Yamasoba D, Gerber PP, Fatihi S, Rathore S, Zepeda SK, Papa G, Kemp SA, Ikeda T, Toyoda M, Tan TS, Kuramochi J, Mitsunaga S, Ueno T, Shirakawa K, Takaori-Kondo A, Brevini T, Mallery DL, Charles OJ, Bowen JE, Joshi A, Walls AC, Jackson L, Martin D, Smith KGC, Bradley J, Briggs JAG, Choi J, Madissoon E, Meyer KB, Mlcochova P, Ceron-Gutierrez L, Doffinger R, Teichmann SA, Fisher AJ, Pizzuto MS, de Marco A, Corti D, Hosmillo M, Lee JH, James LC, Thukral L, Veesler D, Sigal A, Sampaziotis F, Goodfellow IG, Matheson NJ, Sato K, Gupta RK. Altered TMPRSS2 usage by SARS-CoV-2 Omicron impacts infectivity and fusogenicity. Nature 2022; 603:706-714. [PMID: 35104837 PMCID: PMC8942856 DOI: 10.1038/s41586-022-04474-x] [Citation(s) in RCA: 682] [Impact Index Per Article: 341.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/26/2022] [Indexed: 11/08/2022]
Abstract
The SARS-CoV-2 Omicron BA.1 variant emerged in 20211 and has multiple mutations in its spike protein2. Here we show that the spike protein of Omicron has a higher affinity for ACE2 compared with Delta, and a marked change in its antigenicity increases Omicron's evasion of therapeutic monoclonal and vaccine-elicited polyclonal neutralizing antibodies after two doses. mRNA vaccination as a third vaccine dose rescues and broadens neutralization. Importantly, the antiviral drugs remdesivir and molnupiravir retain efficacy against Omicron BA.1. Replication was similar for Omicron and Delta virus isolates in human nasal epithelial cultures. However, in lung cells and gut cells, Omicron demonstrated lower replication. Omicron spike protein was less efficiently cleaved compared with Delta. The differences in replication were mapped to the entry efficiency of the virus on the basis of spike-pseudotyped virus assays. The defect in entry of Omicron pseudotyped virus to specific cell types effectively correlated with higher cellular RNA expression of TMPRSS2, and deletion of TMPRSS2 affected Delta entry to a greater extent than Omicron. Furthermore, drug inhibitors targeting specific entry pathways3 demonstrated that the Omicron spike inefficiently uses the cellular protease TMPRSS2, which promotes cell entry through plasma membrane fusion, with greater dependency on cell entry through the endocytic pathway. Consistent with suboptimal S1/S2 cleavage and inability to use TMPRSS2, syncytium formation by the Omicron spike was substantially impaired compared with the Delta spike. The less efficient spike cleavage of Omicron at S1/S2 is associated with a shift in cellular tropism away from TMPRSS2-expressing cells, with implications for altered pathogenesis.
Collapse
Affiliation(s)
- Bo Meng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Niluka Goonawardane
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Izumi Kimura
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Daichi Yamasoba
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Saman Fatihi
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Surabhi Rathore
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Samantha K Zepeda
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Guido Papa
- MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Mako Toyoda
- Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Toong Seng Tan
- Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | | | - Shigeki Mitsunaga
- Human Genetics Laboratory, National Institute of Genetics, Mishima, Japan
| | - Takamasa Ueno
- Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kotaro Shirakawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Teresa Brevini
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - John E Bowen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Anshu Joshi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Department of Virology, University of Cambridge, Cambridge, UK
| | | | | | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - John Bradley
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Jinwook Choi
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - Elo Madissoon
- Welcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, UK
| | - Kerstin B Meyer
- Welcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lourdes Ceron-Gutierrez
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Rainer Doffinger
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Sarah A Teichmann
- Welcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Andrew J Fisher
- Transplant and Regenerative Medicine Laboratory, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Matteo S Pizzuto
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Anna de Marco
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Myra Hosmillo
- Department of Virology, University of Cambridge, Cambridge, UK
| | - Joo Hyeon Lee
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Leo C James
- MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Lipi Thukral
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Fotios Sampaziotis
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Kei Sato
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- CREST, Japan Science and Technology Agency, Saitama, Japan.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Africa Health Research Institute, Durban, South Africa.
| |
Collapse
|
80
|
Tsai TI, Khalili JS, Gilchrist M, Waight AB, Cohen D, Zhuo S, Zhang Y, Ding M, Zhu H, Mak ANS, Zhu Y, Goulet DR. ACE2-Fc fusion protein overcomes viral escape by potently neutralizing SARS-CoV-2 variants of concern. Antiviral Res 2022; 199:105271. [PMID: 35240221 PMCID: PMC8882475 DOI: 10.1016/j.antiviral.2022.105271] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/06/2022] [Accepted: 02/23/2022] [Indexed: 01/01/2023]
Abstract
COVID-19, an infectious disease caused by the SARS-CoV-2 virus, emerged globally in early 2020 and has remained a serious public health issue. To date, although several preventative vaccines have been approved by FDA and EMA, vaccinated individuals increasingly suffer from breakthrough infections. Therapeutic antibodies may provide an alternative strategy to neutralize viral infection and treat serious cases; however, the clinical data and our experiments show that some FDA-approved monoclonal antibodies lose function against COVID-19 variants such as Omicron. Therefore, in this study, we present a novel therapeutic agent, SI-F019, an ACE2-Fc fusion protein whose neutralization efficiency is not compromised, but actually strengthened, by the mutations of dominant variants including Omicron. Comprehensive biophysical analyses revealed the mechanism of increased inhibition to be enhanced interaction of SI-F019 with all the tested spike variants, in contrast to monoclonal antibodies which tended to show weaker binding to some variants. The results imply that SI-F019 may be a broadly useful agent for treatment of COVID-19.
Collapse
Affiliation(s)
- Tsung-I Tsai
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | | | - Mark Gilchrist
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | | | - Daniella Cohen
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | - Shi Zhuo
- Chengdu Strait Technology Industry Development Park, Wenjiang Dist., Chengdu, Sichuan, 611130, China
| | - Yong Zhang
- Chengdu Strait Technology Industry Development Park, Wenjiang Dist., Chengdu, Sichuan, 611130, China
| | - Muran Ding
- Chengdu Strait Technology Industry Development Park, Wenjiang Dist., Chengdu, Sichuan, 611130, China
| | - Hai Zhu
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | | | - Yi Zhu
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | - Dennis R Goulet
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA.
| |
Collapse
|
81
|
Zhang X, Zheng M, Wang H, Zhou H, Liang T, Zhang J, Ren J, Peng H, Li S, Bian H, Wei C, Yin S, He C, Han Y, Li M, Hou X, Zhang J, Xie L, Lv J, Kan B, Wang Y, Yu X. Inhibitor screening using microarray identifies the high capacity of neutralizing antibodies to Spike variants in SARS-CoV-2 infection and vaccination. Theranostics 2022; 12:2519-2534. [PMID: 35401825 PMCID: PMC8965487 DOI: 10.7150/thno.67038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/10/2022] [Indexed: 11/18/2022] Open
Abstract
Rationale: Mutations of SARS-CoV-2, which is responsible for coronavirus disease 2019 (COVID-19), could impede drug development and reduce the efficacy of COVID-19 vaccines. Here, we developed a multiplexed Spike-ACE2 Inhibitor Screening (mSAIS) assay that can measure the neutralizing effect of antibodies across numerous variants of the coronavirus's Spike (S) protein simultaneously. Methods: The SARS-CoV-2 spike variant protein microarrays were prepared by printing 72 S variants onto a chemically-modified glass slides. The neutralization potential of purified anti-S antibodies and serum from convalescent COVID-19 patients and vaccinees to S variants were assessed with the mSAIS assay. Results: We identified new S mutations that are sensitive and resistant to neutralization. Serum from both infected and vaccinated groups with a high titer of neutralizing antibodies (NAbs) displayed a broader capacity to neutralize S variants than serum with low titer NAbs. These data were validated using serum from a large vaccinated cohort (n = 104) with a tiled S peptide microarray. In addition, similar results were obtained using a SARS-CoV-2 pseudovirus neutralization assay specific for wild-type S and five prevalent S variants (D614G, B.1.1.7, B.1.351, P.1, B.1.617.2), thus demonstrating that high antibody diversity is associated with high NAb titers. Conclusions: Our results demonstrate the utility of the mSAIS platform in screening NAbs. Moreover, we show that heterogeneous antibody populations provide a more protective effect against S variants, which may help direct COVID-19 vaccine and drug development.
Collapse
Affiliation(s)
- Xiaomei Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Mei Zheng
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Department of Research Ward, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Hongye Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Haijian Zhou
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Te Liang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jiahui Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jing Ren
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Huoying Peng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Siping Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Haodong Bian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Chundi Wei
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Shangqi Yin
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Department of Research Ward, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Chaonan He
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Department of Research Ward, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Ying Han
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Department of Research Ward, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Minghui Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Xuexin Hou
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Jie Zhang
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological, Inc., Beijing, 100176, China
| | - Liangzhi Xie
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological, Inc., Beijing, 100176, China
| | - Jing Lv
- Gobond Testing Technology (Beijing) Co., Ltd., Beijing, 102629, China
| | - Biao Kan
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Department of Research Ward, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
- Joint Laboratory for Pathogen Identification of ICDC and Ditan Hospital, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| |
Collapse
|
82
|
Martinell M, Andersson T, Mannsverk SS, Bergholm J, Ellström P, Hill A, Lindh J, Kaden R. In-Flight Transmission of a SARS-CoV-2 Lineage B.1.617.2 Harbouring the Rare S:E484Q Immune Escape Mutation. Viruses 2022; 14:504. [PMID: 35336908 PMCID: PMC8953675 DOI: 10.3390/v14030504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/04/2022] Open
Abstract
We describe a flight-associated infection scenario of seven individuals with a B.1.617.2 (Delta) lineage, harbouring an S:E484Q point mutation. In Sweden, at least 10% of all positive SARS-CoV-2 samples were sequenced in each county; the B.1.717.2 + S:E484Q combination was not detected in Sweden before and was imported within the scenario described in this report. The high transmission rate of the delta lineage combined with the S:E484Q mutation, associated with immune escape in other lineages, makes this specific genetic combination a possible threat to the global fight against the COVID-19 pandemic. Even within the Omicron wave, the B.1.617.2 + S:E484Q variant appeared in community samples in Sweden, as it seems that this combination has an evolutionary gain compared to other B.1.617.2 lineages. The here described genomic combination was not detectable with the common fasta file-based Pango-lineage analysis, hence increasing the probability of the true global prevalence to be higher.
Collapse
Affiliation(s)
- Mats Martinell
- Primary Care and Health, Uppsala County Council, 75320 Uppsala, Sweden; (M.M.); (T.A.)
- Department of Public Health and Caring Sciences, Uppsala University, 75237 Uppsala, Sweden
| | - Tanja Andersson
- Primary Care and Health, Uppsala County Council, 75320 Uppsala, Sweden; (M.M.); (T.A.)
- Department of Public Health and Caring Sciences, Uppsala University, 75237 Uppsala, Sweden
| | - Steinar Smørholm Mannsverk
- Clinical Microbiology and Hospital Hygiene, Uppsala University Hospital, 75237 Uppsala, Sweden; (S.S.M.); (J.B.); (P.E.); (A.H.); (J.L.)
| | - Julia Bergholm
- Clinical Microbiology and Hospital Hygiene, Uppsala University Hospital, 75237 Uppsala, Sweden; (S.S.M.); (J.B.); (P.E.); (A.H.); (J.L.)
| | - Patrik Ellström
- Clinical Microbiology and Hospital Hygiene, Uppsala University Hospital, 75237 Uppsala, Sweden; (S.S.M.); (J.B.); (P.E.); (A.H.); (J.L.)
- Department of Medical Sciences, Clinical Microbiology, Uppsala University, 75236 Uppsala, Sweden
| | - Anna Hill
- Clinical Microbiology and Hospital Hygiene, Uppsala University Hospital, 75237 Uppsala, Sweden; (S.S.M.); (J.B.); (P.E.); (A.H.); (J.L.)
| | - Johan Lindh
- Clinical Microbiology and Hospital Hygiene, Uppsala University Hospital, 75237 Uppsala, Sweden; (S.S.M.); (J.B.); (P.E.); (A.H.); (J.L.)
- Department of Medical Sciences, Clinical Microbiology, Uppsala University, 75236 Uppsala, Sweden
- Science for Life Laboratory, Clinical Genomics Uppsala, 75237 Uppsala, Sweden
| | - Rene Kaden
- Department of Medical Sciences, Clinical Microbiology, Uppsala University, 75236 Uppsala, Sweden
- Science for Life Laboratory, Clinical Genomics Uppsala, 75237 Uppsala, Sweden
| |
Collapse
|
83
|
Kalnin KV, Plitnik T, Kishko M, Huang D, Raillard A, Piolat J, Anosova NG, Tibbitts T, DiNapoli J, Karve S, Goldman R, Gopani H, Dias A, Tran K, Zacharia M, Gu X, Boeglin L, Abysalh J, Vargas J, Beaulieu A, Shah M, Jeannotte T, Gillis K, Chivukula S, Swearingen R, Landolfi V, Fu TM, DeRosa F, Casimiro D. Pan-SARS neutralizing responses after third boost vaccination in non-human primate immunogenicity model. Vaccine 2022; 40:1289-1298. [PMID: 35101265 PMCID: PMC8801978 DOI: 10.1016/j.vaccine.2022.01.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/08/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022]
Abstract
The emergence of SARS-CoV-2 variants, especially Beta and Delta, has raised concerns about the reduced protection from previous infection or vaccination based on the original Wuhan-Hu-1 (D614) virus. To identify promising regimens for inducing neutralizing titers towards new variants, we evaluated monovalent and bivalent mRNA vaccines either as primary vaccination or as a booster in nonhuman primates (NHPs). Two mRNA vaccines, D614-based MRT5500 and Beta-based MRT5500β, tested in sequential regimens or as a bivalent combination in naïve NHPs produced modest neutralizing titers to heterologous variants. However, when mRNA vaccines were administered as a booster to pre-immune NHPs, we observed a robust increase in neutralizing titers with expanded breadth towards all tested variants, and notably SARS-CoV-1. The breadth of the neutralizing response was independent of vaccine sequence or modality, as we further showed either MRT5500 or recombinant subunit Spike protein (with adjuvant) can serve as boosters to induce broadly neutralizing antibodies in the NHPs primed with MRT5500. The data support the notion that a third vaccination is key to boosting existing titers and improving the breadth of antibodies to address variants of concern, including those with an E484K mutation in the Receptor Binding Domain (RBD) (Beta, Gamma).
Collapse
Affiliation(s)
- Kirill V Kalnin
- Emergent BioSolutions, 3985-A Sorrento Valley Blvd, San Diego, CA 92121, United States
| | - Timothy Plitnik
- Yoh Services LLC, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Michael Kishko
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Dean Huang
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Alice Raillard
- Sanofi Pasteur, 1541 AV Marcel Mérieux, 69280 Marcy l'Etoile, France
| | - Julie Piolat
- Sanofi Pasteur, 1541 AV Marcel Mérieux, 69280 Marcy l'Etoile, France
| | - Natalie G Anosova
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States.
| | - Timothy Tibbitts
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Joshua DiNapoli
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Shrirang Karve
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Rebecca Goldman
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Hardip Gopani
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Anusha Dias
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Khang Tran
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Minnie Zacharia
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Xiaobo Gu
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Lianne Boeglin
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Jonathan Abysalh
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Jorel Vargas
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Angela Beaulieu
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Monic Shah
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Travis Jeannotte
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Kimberly Gillis
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Sudha Chivukula
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Ron Swearingen
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | | | - Tong-Ming Fu
- UT Health Science Center at Houston, 7000 Fannin St #1200, Houston, TX 77030, United States
| | - Frank DeRosa
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Danilo Casimiro
- Sanofi Pasteur, 1541 AV Marcel Mérieux, 69280 Marcy l'Etoile, France
| |
Collapse
|
84
|
Pecetta S, Kratochvil S, Kato Y, Vadivelu K, Rappuoli R. Immunology and Technology of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccines. Pharmacol Rev 2022; 74:313-339. [PMID: 35101964 DOI: 10.1124/pharmrev.120.000285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We have experienced an enormous cohesive effort of the scientific community to understand how the immune system reacts to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and how to elicit protective immunity via vaccination. This effort resulted in the development of vaccines in record time with high levels of safety, efficacy, and real-life effectiveness. However, the rapid diffusion of viral variants that escape protective antibodies prompted new studies to understand SARS-CoV-2 vulnerabilities and strategies to guide follow-up actions to increase, and maintain, the protection offered by vaccines. In this review, we report the main findings on human immunity to SARS-CoV-2 after natural infection and vaccination; we dissect the immunogenicity and efficacy of the different vaccination strategies that resulted in products widely used in the population; and we describe the impact of viral variants on vaccine-elicited immunity, summarizing the main discoveries and challenges to stay ahead of SARS-CoV-2 evolution. SIGNIFICANCE STATEMENT: This study reviewed findings on human immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), analyzed the immunogenicity and efficacy of the various vaccines currently used in large vaccination campaigns or candidates in advanced clinical development, and discussed the challenging task to ensure high protective efficacy against the rapidly evolving SARS-CoV-2 virus. This manuscript was completed prior to the emergence of the Omicron variant and to global vaccine boosting efforts.
Collapse
Affiliation(s)
- Simone Pecetta
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Sven Kratochvil
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Yu Kato
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Kumaran Vadivelu
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Rino Rappuoli
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| |
Collapse
|
85
|
Kovacech B, Fialova L, Filipcik P, Skrabana R, Zilkova M, Paulenka-Ivanovova N, Kovac A, Palova D, Rolkova GP, Tomkova K, Csokova NT, Markova K, Skrabanova M, Sinska K, Basheer N, Majerova P, Hanes J, Parrak V, Prcina M, Cehlar O, Cente M, Piestansky J, Fresser M, Novak M, Slavikova M, Borsova K, Cabanova V, Brejova B, Vinař T, Nosek J, Klempa B, Eyer L, Hönig V, Palus M, Ruzek D, Vyhlidalova T, Strakova P, Mrazkova B, Zudova D, Koubkova G, Novosadova V, Prochazka J, Sedlacek R, Zilka N, Kontsekova E. Monoclonal antibodies targeting two immunodominant epitopes on the Spike protein neutralize emerging SARS-CoV-2 variants of concern. EBioMedicine 2022; 76:103818. [PMID: 35078012 PMCID: PMC8782626 DOI: 10.1016/j.ebiom.2022.103818] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The emergence of new SARS-CoV-2 variants of concern B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma) and B.1.617.2 (Delta) that harbor mutations in the viral S protein raised concern about activity of current vaccines and therapeutic antibodies. Independent studies have shown that mutant variants are partially or completely resistant against some of the therapeutic antibodies authorized for emergency use. METHODS We employed hybridoma technology, ELISA-based and cell-based S-ACE2 interaction assays combined with authentic virus neutralization assays to develop second-generation antibodies, which were specifically selected for their ability to neutralize the new variants of SARS-CoV-2. FINDINGS AX290 and AX677, two monoclonal antibodies with non-overlapping epitopes, exhibit subnanomolar or nanomolar affinities to the receptor binding domain of the viral Spike protein carrying amino acid substitutions N501Y, N439K, E484K, K417N, and a combination N501Y/E484K/K417N found in the circulating virus variants. The antibodies showed excellent neutralization of an authentic SARS-CoV-2 virus representing strains circulating in Europe in spring 2020 and also the variants of concern B.1.1.7 (Alpha), B.1.351 (Beta) and B.1.617.2 (Delta). In addition, AX677 is able to bind Omicron Spike protein just like the wild type Spike. The combination of the two antibodies prevented the appearance of escape mutations of the authentic SARS-CoV-2 virus. Prophylactic administration of AX290 and AX677, either individually or in combination, effectively reduced viral burden and inflammation in the lungs, and prevented disease in a mouse model of SARS-CoV-2 infection. INTERPRETATION The virus-neutralizing properties were fully reproduced in chimeric mouse-human versions of the antibodies, which may represent a promising tool for COVID-19 therapy. FUNDING The study was funded by AXON Neuroscience SE and AXON COVIDAX a.s.
Collapse
Affiliation(s)
- Branislav Kovacech
- AXON COVIDAX a. s.; Bratislava, 811 02, Slovakia; AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia.
| | - Lubica Fialova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Peter Filipcik
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | | | - Monika Zilkova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | | | - Andrej Kovac
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Denisa Palova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | | | | | - Natalia Turic Csokova
- Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Karina Markova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Michaela Skrabanova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Kristina Sinska
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Neha Basheer
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Petra Majerova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Jozef Hanes
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Vojtech Parrak
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Michal Prcina
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Ondrej Cehlar
- Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Martin Cente
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | | | - Michal Fresser
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | | | - Monika Slavikova
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia
| | - Kristina Borsova
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia; Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava; Bratislava, 842 15, Slovakia
| | - Viktoria Cabanova
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia
| | - Bronislava Brejova
- Department of Computer Science, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava; Bratislava, 842 48, Slovakia
| | - Tomas Vinař
- Department of Applied Informatics, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava; Bratislava, 842 48, Slovakia
| | - Jozef Nosek
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava; Bratislava, 842 15, Slovakia
| | - Boris Klempa
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia
| | - Ludek Eyer
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Vaclav Hönig
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Martin Palus
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Daniel Ruzek
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-62500 Brno, Czech Republic
| | - Tereza Vyhlidalova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic
| | - Petra Strakova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Blanka Mrazkova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Dagmar Zudova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Gizela Koubkova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Vendula Novosadova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Jan Prochazka
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Radislav Sedlacek
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Norbert Zilka
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia.
| | - Eva Kontsekova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| |
Collapse
|
86
|
Xiang Q, Li L, Wu J, Tian M, Fu Y. Application of pseudovirus system in the development of vaccine, antiviral-drugs, and neutralizing antibodies. Microbiol Res 2022; 258:126993. [PMID: 35240544 PMCID: PMC8848573 DOI: 10.1016/j.micres.2022.126993] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/16/2022]
Abstract
Pseudoviruses are viral particles coated with a heterologous envelope protein, which mediates the entry of pseudoviruses as efficiently as that of the live viruses possessing high pathogenicity and infectivity. Due to the deletion of the envelope protein gene and the absence of pathogenic genes, pseudoviruses have no autonomous replication ability and can infect host cells for only a single cycle. In addition, pseudoviruses have the desired characteristics of high safety, strong operability, and can be easily used to perform rapid throughput detection. Therefore, pseudoviruses are widely employed in the mechanistic investigation of viral infection, the screening and evaluation of monoclonal antibodies and antiviral drugs, and the detection of neutralizing antibody titers in serum after vaccination. In this review, we will discuss the construction of pseudoviruses based on different packaging systems, their current applications especially in the research of SARS-CoV-2, limitations, and further directions.
Collapse
Affiliation(s)
- Qi Xiang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Linhao Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jie Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Miao Tian
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yang Fu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
87
|
da Silva SJR, de Lima SC, da Silva RC, Kohl A, Pena L. Viral Load in COVID-19 Patients: Implications for Prognosis and Vaccine Efficacy in the Context of Emerging SARS-CoV-2 Variants. Front Med (Lausanne) 2022; 8:836826. [PMID: 35174189 PMCID: PMC8841511 DOI: 10.3389/fmed.2021.836826] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/29/2021] [Indexed: 12/14/2022] Open
Abstract
The worldwide spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused an unprecedented public health crisis in the 21st century. As the pandemic evolves, the emergence of SARS-CoV-2 has been characterized by the emergence of new variants of concern (VOCs), which resulted in a catastrophic impact on SARS-CoV-2 infection. In light of this, research groups around the world are unraveling key aspects of the associated illness, coronavirus disease 2019 (COVID-19). A cumulative body of data has indicated that the SARS-CoV-2 viral load may be a determinant of the COVID-19 severity. Here we summarize the main characteristics of the emerging variants of SARS-CoV-2, discussing their impact on viral transmissibility, viral load, disease severity, vaccine breakthrough, and lethality among COVID-19 patients. We also provide a rundown of the rapidly expanding scientific evidence from clinical studies and animal models that indicate how viral load could be linked to COVID-19 prognosis and vaccine efficacy among vaccinated individuals, highlighting the differences compared to unvaccinated individuals.
Collapse
Affiliation(s)
- Severino Jefferson Ribeiro da Silva
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Suelen Cristina de Lima
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Ronaldo Celerino da Silva
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Lindomar Pena
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| |
Collapse
|
88
|
Hu Z, Huang X, Zhang J, Fu S, Ding D, Tao Z. Differences in Clinical Characteristics Between Delta Variant and Wild-Type SARS-CoV-2 Infected Patients. Front Med (Lausanne) 2022; 8:792135. [PMID: 35047533 PMCID: PMC8761722 DOI: 10.3389/fmed.2021.792135] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/17/2021] [Indexed: 01/08/2023] Open
Abstract
Background: As delta variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) prevailed in the current coronavirus disease 2019 (COVID-19) pandemic, its clinical characteristics with the difference from those of wild-type strains have been little studied. Methods: We reported one cohort of 341 wild-type patients with COVID-19 admitted at Wuhan, China in 2020 and the other cohort of 336 delta variant patients with COVID-19 admitted at Yangzhou, China in 2021, with comparisons of their demographic information, medical history, clinical manifestation, and hematological data. Furthermore, within the delta variant cohort, patients with none, partial, and full vaccination were also compared to assess vaccine effectiveness. Findings: For a total of 677 patients with COVID-19 included in this study, their median age was 53.0 years [interquartile range (IQR): 38.0–66.0] and 46.8% were men. No difference was found in age, gender, and percentage of patients with the leading comorbidity between wild-type and delta variant cohorts, but delta variant cohort showed a lessened time interval between disease onset to hospitalization, a reduced portion of patients with smoking history, and a lowered frequency of clinical symptoms. For hematological parameters, most values demonstrated significant differences between wild-type and delta variant cohorts, while full vaccination rather than partial vaccination alleviated the disease condition. This reflected the viremic effect of delta variant when vaccination succeeds or fails to protect. Interpretation: Delta variant of SARS-CoV-2 may cause severe disease profiles, but timely diagnosis and full vaccination could protect patients with COVID-19 from worsened disease progression.
Collapse
Affiliation(s)
- Zhenkui Hu
- Department of Critical Care Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xing Huang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianguo Zhang
- Department of Critical Care Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Emergency Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shixiang Fu
- Department of Hepatology, The Third People's Hospital of Yangzhou City, Yangzhou, China
| | - Daoyin Ding
- Department of Critical Care Medicine, The First People's Hospital of Jiangxia District, Wuhan, China
| | - Zhimin Tao
- Department of Emergency Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
89
|
Santiago GA, Flores B, Gonzalez GL, Charriez KN, Cora-Huertas L, Volkman HR, Van Belleghem S, Rivera-Amill V, Adams LE, Marzan M, Hernandez L, Cardona I, O'Neill E, Paz-Bailey G, Papa R, Munoz-Jordan JL. Genomic surveillance of SARS-CoV-2 in Puerto Rico reveals emergence of an autochthonous lineage and early detection of variants. RESEARCH SQUARE 2022:rs.3.rs-1277781. [PMID: 35075454 PMCID: PMC8786232 DOI: 10.21203/rs.3.rs-1277781/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Puerto Rico has experienced the full impact of the COVID-19 pandemic. Since SARS-CoV-2, the virus that causes COVID-19, was first detected on the island in March of 2020, it spread rapidly though the island’s population and became a critical threat to public health. We conducted a genomic surveillance study through a partnership with health agencies and academic institutions to understand the emergence and molecular epidemiology of the virus on the island. We sampled COVID-19 cases monthly over 19 months and sequenced a total of 753 SARS-CoV-2 genomes between March 2020 and September 2021 to reconstruct the local epidemic in a regional context using phylogenetic inference. Our analyses revealed that multiple importation events propelled the emergence and spread of the virus throughout the study period, including the introduction and spread of most SARS-CoV-2 variants detected world-wide. Lineage turnover cycles through various phases of the local epidemic were observed, where the predominant lineage was replaced by the next competing lineage or variant after approximately 4 months of circulation locally. We also identified the emergence of lineage B.1.588, an autochthonous lineage that predominated circulation in Puerto Rico from September to December 2020 and subsequently spread to the United States. The results of this collaborative approach highlight the importance of timely collection and analysis of SARS-CoV-2 genomic surveillance data to inform public health responses.
Collapse
|
90
|
Abstract
The SARS-CoV-2 Omicron BA.1 variant emerged in late 2021 and is characterised by multiple spike mutations across all spike domains. Here we show that Omicron BA.1 has higher affinity for ACE2 compared to Delta, and confers very significant evasion of therapeutic monoclonal and vaccine-elicited polyclonal neutralising antibodies after two doses. mRNA vaccination as a third vaccine dose rescues and broadens neutralisation. Importantly, antiviral drugs remdesevir and molnupiravir retain efficacy against Omicron BA.1. We found that in human nasal epithelial 3D cultures replication was similar for both Omicron and Delta. However, in lower airway organoids, Calu-3 lung cells and gut adenocarcinoma cell lines live Omicron virus demonstrated significantly lower replication in comparison to Delta. We noted that despite presence of mutations predicted to favour spike S1/S2 cleavage, the spike protein is less efficiently cleaved in live Omicron virions compared to Delta virions. We mapped the replication differences between the variants to entry efficiency using spike pseudotyped virus (PV) entry assays. The defect for Omicron PV in specific cell types correlated with higher cellular RNA expression of TMPRSS2, and accordingly knock down of TMPRSS2 impacted Delta entry to a greater extent as compared to Omicron. Furthermore, drug inhibitors targeting specific entry pathways demonstrated that the Omicron spike inefficiently utilises the cellular protease TMPRSS2 that mediates cell entry via plasma membrane fusion. Instead, we demonstrate that Omicron spike has greater dependency on cell entry via the endocytic pathway requiring the activity of endosomal cathepsins to cleave spike. Consistent with suboptimal S1/S2 cleavage and inability to utilise TMPRSS2, syncytium formation by the Omicron spike was dramatically impaired compared to the Delta spike. Overall, Omicron appears to have gained significant evasion from neutralising antibodies whilst maintaining sensitivity to antiviral drugs targeting the polymerase. Omicron has shifted cellular tropism away from TMPRSS2 expressing cells that are enriched in cells found in the lower respiratory and GI tracts, with implications for altered pathogenesis.
Collapse
|
91
|
Abstract
The spike protein (S-protein) of SARS-CoV-2, the protein that enables the virus to infect human cells, is the basis for many vaccines and a hotspot of concerning virus evolution. Here, we discuss the outstanding progress in structural characterization of the S-protein and how these structures facilitate analysis of virus function and evolution. We emphasize the differences in reported structures and that analysis of structure-function relationships is sensitive to the structure used. We show that the average residue solvent exposure in nearly complete structures is a good descriptor of open vs closed conformation states. Because of structural heterogeneity of functionally important surface-exposed residues, we recommend using averages of a group of high-quality protein structures rather than a single structure before reaching conclusions on specific structure-function relationships. To illustrate these points, we analyze some significant chemical tendencies of prominent S-protein mutations in the context of the available structures. In the discussion of new variants, we emphasize the selectivity of binding to ACE2 vs prominent antibodies rather than simply the antibody escape or ACE2 affinity separately. We note that larger chemical changes, in particular increased electrostatic charge or side-chain volume of exposed surface residues, are recurring in mutations of concern, plausibly related to adaptation to the negative surface potential of human ACE2. We also find indications that the fixated mutations of the S-protein in the main variants are less destabilizing than would be expected on average, possibly pointing toward a selection pressure on the S-protein. The richness of available structures for all of these situations provides an enormously valuable basis for future research into these structure-function relationships.
Collapse
Affiliation(s)
- Rukmankesh Mehra
- Department of Chemistry, Indian Institute
of Technology Bhilai, Sejbahar, Raipur 492015, Chhattisgarh,
India
| | - Kasper P. Kepp
- DTU Chemistry, Technical University of
Denmark, Building 206, 2800 Kongens Lyngby,
Denmark
| |
Collapse
|
92
|
Tran BM, Grimley SL, McAuley JL, Hachani A, Earnest L, Wong SL, Caly L, Druce J, Purcell DFJ, Jackson DC, Catton M, Nowell CJ, Leonie L, Deliyannis G, Waters SA, Torresi J, Vincan E. Air-Liquid-Interface Differentiated Human Nose Epithelium: A Robust Primary Tissue Culture Model of SARS-CoV-2 Infection. Int J Mol Sci 2022; 23:835. [PMID: 35055020 PMCID: PMC8776210 DOI: 10.3390/ijms23020835] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
The global urgency to uncover medical countermeasures to combat the COVID-19 pandemic caused by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has revealed an unmet need for robust tissue culture models that faithfully recapitulate key features of human tissues and disease. Infection of the nose is considered the dominant initial site for SARS-CoV-2 infection and models that replicate this entry portal offer the greatest potential for examining and demonstrating the effectiveness of countermeasures designed to prevent or manage this highly communicable disease. Here, we test an air-liquid-interface (ALI) differentiated human nasal epithelium (HNE) culture system as a model of authentic SARS-CoV-2 infection. Progenitor cells (basal cells) were isolated from nasal turbinate brushings, expanded under conditionally reprogrammed cell (CRC) culture conditions and differentiated at ALI. Differentiated cells were inoculated with different SARS-CoV-2 clinical isolates. Infectious virus release into apical washes was determined by TCID50, while infected cells were visualized by immunofluorescence and confocal microscopy. We demonstrate robust, reproducible SARS-CoV-2 infection of ALI-HNE established from different donors. Viral entry and release occurred from the apical surface, and infection was primarily observed in ciliated cells. In contrast to the ancestral clinical isolate, the Delta variant caused considerable cell damage. Successful establishment of ALI-HNE is donor dependent. ALI-HNE recapitulate key features of human SARS-CoV-2 infection of the nose and can serve as a pre-clinical model without the need for invasive collection of human respiratory tissue samples.
Collapse
Affiliation(s)
- Bang M. Tran
- Department of Infectious Diseases, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia;
| | - Samantha L. Grimley
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Julie L. McAuley
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Linda Earnest
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Sharon L. Wong
- Molecular and Integrative Cystic Fibrosis Research Centre, School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; (S.L.W.); (S.A.W.)
| | - Leon Caly
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
| | - Julian Druce
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
| | - Damian F. J. Purcell
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - David C. Jackson
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Mike Catton
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
| | - Cameron J. Nowell
- Imaging, FACS and Analysis Core, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia;
| | - Laura Leonie
- Melbourne Histology Platform, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Georgia Deliyannis
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Shafagh A. Waters
- Molecular and Integrative Cystic Fibrosis Research Centre, School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; (S.L.W.); (S.A.W.)
- School of Women’s and Children’s Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children’s Hospital, Randwick, NSW 2031, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (S.L.G.); (J.L.M.); (A.H.); (L.E.); (D.F.J.P.); (D.C.J.); (G.D.)
| | - Elizabeth Vincan
- Department of Infectious Diseases, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia;
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (J.D.); (M.C.)
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| |
Collapse
|
93
|
Kimura I, Kosugi Y, Wu J, Zahradnik J, Yamasoba D, Butlertanaka EP, Tanaka YL, Uriu K, Liu Y, Morizako N, Shirakawa K, Kazuma Y, Nomura R, Horisawa Y, Tokunaga K, Ueno T, Takaori-Kondo A, Schreiber G, Arase H, Motozono C, Saito A, Nakagawa S, Sato K. The SARS-CoV-2 Lambda variant exhibits enhanced infectivity and immune resistance. Cell Rep 2022; 38:110218. [PMID: 34968415 DOI: 10.1101/2021.07.28.454085] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/24/2021] [Accepted: 12/14/2021] [Indexed: 05/22/2023] Open
Abstract
SARS-CoV-2 Lambda, a variant of interest, has spread in some South American countries; however, its virological features and evolutionary traits remain unclear. In this study, we use pseudoviruses and reveal that the spike protein of the Lambda variant is more infectious than that of other variants due to the T76I and L452Q mutations. The RSYLTPGD246-253N mutation, a unique 7-amino acid deletion in the N-terminal domain of the Lambda spike protein, is responsible for evasion from neutralizing antibodies and further augments antibody-mediated enhancement of infection. Although this mutation generates a nascent N-linked glycosylation site, the additional N-linked glycan is dispensable for the virological property conferred by this mutation. Since the Lambda variant has dominantly spread according to the increasing frequency of the isolates harboring the RSYLTPGD246-253N mutation, our data suggest that the RSYLTPGD246-253N mutation is closely associated with the substantial spread of the Lambda variant in South America.
Collapse
Affiliation(s)
- Izumi Kimura
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo 1088639, Japan
| | - Yusuke Kosugi
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo 1088639, Japan; Laboratory of Systems Virology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 6068501, Japan
| | - Jiaqi Wu
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Kanagawa 2591193, Japan; CREST, Japan Science and Technology Agency, Saitama 3220012, Japan
| | - Jiri Zahradnik
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Daichi Yamasoba
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo 1088639, Japan; Faculty of Medicine, Kobe University, Hyogo 6500017, Japan
| | - Erika P Butlertanaka
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 8892192, Japan
| | - Yuri L Tanaka
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 8892192, Japan
| | - Keiya Uriu
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo 1088639, Japan; Graduate School of Medicine, The University of Tokyo, 1130033 Tokyo, Japan
| | - Yafei Liu
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 5650871, Japan; Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 5650871, Japan
| | - Nanami Morizako
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 8892192, Japan
| | - Kotaro Shirakawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 6068507, Japan
| | - Yasuhiro Kazuma
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 6068507, Japan
| | - Ryosuke Nomura
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 6068507, Japan
| | - Yoshihito Horisawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 6068507, Japan
| | - Kenzo Tokunaga
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 1628640, Japan
| | - Takamasa Ueno
- Division of Infection and immunity, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto 8600811, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 6068507, Japan
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hisashi Arase
- Graduate School of Medicine, The University of Tokyo, 1130033 Tokyo, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 5650871, Japan; Center for Infectious Disease Education and Research, Osaka University, Osaka 5650871, Japan
| | - Chihiro Motozono
- Division of Infection and immunity, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto 8600811, Japan
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 8892192, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki 8892192, Japan; Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 8892192, Japan
| | - So Nakagawa
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Kanagawa 2591193, Japan; CREST, Japan Science and Technology Agency, Saitama 3220012, Japan; Bioinformation and DDBJ Center, National Institute of Genetics, Mishima, Shizuoka 4118540, Japan.
| | - Kei Sato
- Division of Systems Virology, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo 1088639, Japan; CREST, Japan Science and Technology Agency, Saitama 3220012, Japan; Graduate School of Medicine, The University of Tokyo, 1130033 Tokyo, Japan.
| |
Collapse
|
94
|
Tang J, Grubbs G, Lee Y, Wu H, Luke TC, Egland KA, Bausch CL, Sullivan EJ, Khurana S. OUP accepted manuscript. J Infect Dis 2022; 226:655-663. [PMID: 35106573 PMCID: PMC8903330 DOI: 10.1093/infdis/jiac031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 12/01/2022] Open
Abstract
Passive antibody immunotherapeutics directed against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are promising countermeasures for protection and treatment of coronavirus disease 2019 (COVID-19). SARS-CoV-2 variants of concern (VOCs) and variants of interest (VOIs) can impact the clinical efficacy of immunotherapeutics. A fully human polyclonal antibody immunotherapeutic purified from plasma of transchromosomic (Tc) bovines hyperimmunized with SARS-CoV-2 WA-1 spike (SAB-185) is being assessed for efficacy in a phase 2/3 clinical trial when different circulating SARS-CoV-2 variants predominated. We evaluated antibody binding, avidity maturation, and SARS-CoV-2 VOCs/VOIs virus-neutralizing capacity of convalescent plasma compared with different lots of SAB-185 and individual Tc bovine sera sequentially obtained after each vaccination against Alpha, Epsilon, Iota, Gamma, Beta, Kappa, and Delta variants. In contrast to convalescent plasma, sera and SAB-185 derived from hyperimmunized Tc bovines demonstrated higher antibody avidity and more potent cross-neutralizing activity of VOCs/VOIs. Thus, SAB-185 is a potential promising therapeutic candidate for the treatment of patients infected with SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Youri Lee
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hua Wu
- SAB Biotherapeutics, Sioux Falls, South Dakota, USA
| | | | | | | | | | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
- Correspondence: Surender Khurana, PhD, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA ()
| |
Collapse
|
95
|
Abstract
The unprecedented public health and economic impact of the COVID-19 pandemic caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been met with an equally unprecedented scientific response. Much of this response has focused, appropriately, on the mechanisms of SARS-CoV-2 entry into host cells, and in particular the binding of the spike (S) protein to its receptor, angiotensin-converting enzyme 2 (ACE2), and subsequent membrane fusion. This Review provides the structural and cellular foundations for understanding the multistep SARS-CoV-2 entry process, including S protein synthesis, S protein structure, conformational transitions necessary for association of the S protein with ACE2, engagement of the receptor-binding domain of the S protein with ACE2, proteolytic activation of the S protein, endocytosis and membrane fusion. We define the roles of furin-like proteases, transmembrane protease, serine 2 (TMPRSS2) and cathepsin L in these processes, and delineate the features of ACE2 orthologues in reservoir animal species and S protein adaptations that facilitate efficient human transmission. We also examine the utility of vaccines, antibodies and other potential therapeutics targeting SARS-CoV-2 entry mechanisms. Finally, we present key outstanding questions associated with this critical process.
Collapse
Affiliation(s)
- Cody B Jackson
- Department of Immunology and Microbiology, Scripps Research, Jupiter, FL, USA
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Michael Farzan
- Department of Immunology and Microbiology, Scripps Research, Jupiter, FL, USA
| | - Bing Chen
- Division of Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Hyeryun Choe
- Department of Immunology and Microbiology, Scripps Research, Jupiter, FL, USA.
| |
Collapse
|
96
|
Jeong BS, Cha JS, Hwang I, Kim U, Adolf-Bryfogle J, Coventry B, Cho HS, Kim KD, Oh BH. Computational design of a neutralizing antibody with picomolar binding affinity for all concerning SARS-CoV-2 variants. MAbs 2022; 14:2021601. [PMID: 35030983 PMCID: PMC8765073 DOI: 10.1080/19420862.2021.2021601] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/17/2021] [Indexed: 11/29/2022] Open
Abstract
Coronavirus disease 2019, caused by SARS-CoV-2, remains an on-going pandemic, partly due to the emergence of variant viruses that can "break-through" the protection of the current vaccines and neutralizing antibodies (nAbs), highlighting the needs for broadly nAbs and next-generation vaccines. We report an antibody that exhibits breadth and potency in binding the receptor-binding domain (RBD) of the virus spike glycoprotein across SARS coronaviruses. Initially, a lead antibody was computationally discovered and crystallographically validated that binds to a highly conserved surface of the RBD of wild-type SARS-CoV-2. Subsequently, through experimental affinity enhancement and computational affinity maturation, it was further developed to bind the RBD of all concerning SARS-CoV-2 variants, SARS-CoV-1 and pangolin coronavirus with pico-molar binding affinities, consistently exhibited strong neutralization activity against wild-type SARS-CoV-2 and the Alpha and Delta variants. These results identify a vulnerable target site on coronaviruses for development of pan-sarbecovirus nAbs and vaccines.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/chemistry
- Angiotensin-Converting Enzyme 2/metabolism
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/metabolism
- Antibody Affinity
- Antibody Specificity
- Antigen-Antibody Reactions
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Broadly Neutralizing Antibodies/genetics
- Broadly Neutralizing Antibodies/immunology
- Broadly Neutralizing Antibodies/metabolism
- COVID-19/immunology
- Crystallography, X-Ray
- Epitopes/chemistry
- Epitopes/immunology
- Humans
- Immunoglobulin Fragments/immunology
- Molecular Docking Simulation
- Monte Carlo Method
- Neutralization Tests
- Peptide Fragments/chemistry
- Peptide Fragments/metabolism
- Protein Domains
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
Collapse
Affiliation(s)
- Bo-Seong Jeong
- Department of Biological Sciences, Kaist Institute for the Biocentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jeong Seok Cha
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Insu Hwang
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Uijin Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jared Adolf-Bryfogle
- Institute for Protein Innovation, Boston, MA, USA
- Division of Hematology-Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Coventry
- Molecular Engineering & Sciences Institute & Institute for Protein Design, University of Washington, Seattle, Washington, USA
| | - Hyun-Soo Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Kyun-Do Kim
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Byung-Ha Oh
- Department of Biological Sciences, Kaist Institute for the Biocentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
97
|
Augusto G, Mohsen MO, Zinkhan S, Liu X, Vogel M, Bachmann MF. In vitro data suggest that Indian delta variant B.1.617 of SARS-CoV-2 escapes neutralization by both receptor affinity and immune evasion. Allergy 2022; 77:111-117. [PMID: 34453338 PMCID: PMC8652796 DOI: 10.1111/all.15065] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Emerged mutations can be attributed to increased transmissibility of the B.1.617 and B.1.36 Indian delta variants of SARS-CoV-2, most notably substitutions L452R/E484Q and N440K, respectively, which occur in the receptor-binding domain (RBD) of the Spike (S) fusion glycoprotein. OBJECTIVE We aimed to assess the effects of mutations L452R/E484Q and N440K (as well as the previously studied mutation E484K present in variants B.1.351 and P.1) on the affinity of RBD for ACE2, SARS-CoV-2 main receptor. We also aimed to assess the ability of antibodies induced by natural infection or by immunization with BNT162b2 mRNA vaccine to recognize the mutated versions of the RBD, as well as blocking the interaction RBD-ACE2, an important surrogate readout for virus neutralization. METHODS To this end, we produced recombinant wild-type RBD, as well as RBD containing each of the mutations L452R/E484Q, N440K, or E484K (the latest present in variants of concern B.1.351 and P.1), as well as the ectodomain of ACE2. Using Biolayer Interferometry (BLI), we measured the binding affinity of RBD for ACE2 and the ability of sera from COVID-19 convalescent donors or subjects immunized with BNT162b2 mRNA vaccine to block this interaction. Finally, we correlated these results with total anti-RBD IgG titers measured from the same sera by direct ELISA. RESULTS The binding assays showed L452R/E484Q double-mutant RBD to interact with ACE2 with higher affinity (KD = 4.6 nM) than wild-type (KD = 21.3 nM) or single mutants N440K (KD = 9.9 nM) and E484K (KD = 19.7 nM) RBDs. Meanwhile, the anti-RBD IgG titration resulted in lower recognition of mutants E484K and L452R/E484Q by infection-induced antibodies, whereas only mutant E484K was recognized less by antibodies induced by vaccination. More interestingly, sera from convalescent as well as immunized subjects showed reduced ability to block the interaction between ACE2 and RBD mutants E484K and L452R/E484Q, as shown by the inhibition assays. CONCLUSION Our data suggest that the newly emerged SARS-CoV-2 variant B.1.617, as well as the better-studied variants B.1.351 and P.1 (all containing a mutation at position E484) display increased transmissibility both due to their higher affinity for the cell receptor ACE2 and their ability to partially bypass immunity generated against the wild-type virus. For variant B.1.36 (with a point mutation at position N440), only increased affinity seems to play a role.
Collapse
Affiliation(s)
- Gilles Augusto
- Department of ImmunologyUniversity clinic of Rheumatology and Immunology, InselspitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
- The Jenner InstituteUniversity of OxfordOxfordUK
| | - Mona O. Mohsen
- Department of ImmunologyUniversity clinic of Rheumatology and Immunology, InselspitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Simon Zinkhan
- Department of ImmunologyUniversity clinic of Rheumatology and Immunology, InselspitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Xuelan Liu
- Department of ImmunologyUniversity clinic of Rheumatology and Immunology, InselspitalBernSwitzerland
- International Immunology CentreAnhui Agricultural UniversityHefeiChina
| | - Monique Vogel
- Department of ImmunologyUniversity clinic of Rheumatology and Immunology, InselspitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Martin F. Bachmann
- Department of ImmunologyUniversity clinic of Rheumatology and Immunology, InselspitalBernSwitzerland
- Department of BioMedical ResearchUniversity of BernBernSwitzerland
- The Jenner InstituteUniversity of OxfordOxfordUK
- International Immunology CentreAnhui Agricultural UniversityHefeiChina
| |
Collapse
|
98
|
Santiago GA, Flores B, González GL, Charriez KN, Huertas LC, Volkman HR, Van Belleghem SM, Rivera-Amill V, Adams LE, Marzán M, Hernández L, Cardona I, O’Neill E, Paz-Bailey G, Papa R, Muñoz-Jordan JL. Genomic surveillance of SARS-CoV-2 in Puerto Rico enabled early detection and tracking of variants. COMMUNICATIONS MEDICINE 2022; 2:100. [PMID: 35968047 PMCID: PMC9366129 DOI: 10.1038/s43856-022-00168-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022] Open
Abstract
Background Puerto Rico has experienced the full impact of the COVID-19 pandemic. Since SARS-CoV-2, the virus that causes COVID-19, was first detected on the island in March of 2020, it spread rapidly though the island's population and became a critical threat to public health. Methods We conducted a genomic surveillance study through a partnership with health agencies and academic institutions to understand the emergence and molecular epidemiology of the virus on the island. We sampled COVID-19 cases monthly over 19 months and sequenced a total of 753 SARS-CoV-2 genomes between March 2020 and September 2021 to reconstruct the local epidemic in a regional context using phylogenetic inference. Results Our analyses reveal that multiple importation events propelled the emergence and spread of the virus throughout the study period, including the introduction and spread of most SARS-CoV-2 variants detected world-wide. Lineage turnover cycles through various phases of the local epidemic were observed, where the predominant lineage was replaced by the next competing lineage or variant after ~4 months of circulation locally. We also identified the emergence of lineage B.1.588, an autochthonous lineage that predominated in Puerto Rico from September to December 2020 and subsequently spread to the United States. Conclusions The results of this collaborative approach highlight the importance of timely collection and analysis of SARS-CoV-2 genomic surveillance data to inform public health responses.
Collapse
Affiliation(s)
- Gilberto A. Santiago
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| | - Betzabel Flores
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| | - Glenda L. González
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| | - Keyla N. Charriez
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| | - Limari Cora Huertas
- grid.280412.dUniversity of Puerto Rico—Río Piedras, Department of Biology, Molecular Sciences and Research Center, San Juan, Puerto Rico
| | - Hannah R. Volkman
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| | - Steven M. Van Belleghem
- grid.280412.dUniversity of Puerto Rico—Río Piedras, Department of Biology, Molecular Sciences and Research Center, San Juan, Puerto Rico
| | - Vanessa Rivera-Amill
- grid.262009.f0000 0004 0455 6268Ponce Health Sciences University, Ponce Research Institute, Department of Basic Sciences, Ponce, Puerto Rico
| | - Laura E. Adams
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| | - Melissa Marzán
- grid.280499.ePuerto Rico Department of Health, Epidemiology Office, San Juan, Puerto Rico
| | - Lorena Hernández
- grid.280499.ePuerto Rico Department of Health, Epidemiology Office, San Juan, Puerto Rico
| | - Iris Cardona
- grid.280499.ePuerto Rico Department of Health, Epidemiology Office, San Juan, Puerto Rico
| | - Eduardo O’Neill
- grid.416738.f0000 0001 2163 0069Centers for Disease Control and Prevention, Office of Island Affairs, Center for State, Tribal, Local, and Territorial Support, Atlanta, GA USA
| | - Gabriela Paz-Bailey
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| | - Riccardo Papa
- grid.280412.dUniversity of Puerto Rico—Río Piedras, Department of Biology, Molecular Sciences and Research Center, San Juan, Puerto Rico
| | - Jorge L. Muñoz-Jordan
- grid.470962.eCenters for Disease Control and Prevention, National Centers for Emerging and Zoonotic Infectious Diseases, Division of Vector Borne Diseases, Dengue Branch, San Juan, Puerto Rico
| |
Collapse
|
99
|
E484K and N501Y SARS-CoV 2 spike mutants Increase ACE2 recognition but reduce affinity for neutralizing antibody. Int Immunopharmacol 2021; 102:108424. [PMID: 34915409 PMCID: PMC8641390 DOI: 10.1016/j.intimp.2021.108424] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022]
Abstract
SARS-CoV2 mutants B.1.1.7, B.1.351, and P.1 contain a key mutation N501Y. B.1.135 and P.1 lineages have another mutation, E484K. Here, we decode the effect of these two mutations on the host receptor, ACE2, and neutralizing antibody (B38) recognition. The N501Y RBD mutant binds to ACE2 with higher affinity due to improved π-π stacking and π-cation interactions. The higher binding affinity of the E484K mutant is caused due to the formation of additional hydrogen bond and salt-bridge interactions with ACE2. Both the mutants bind to the B38 antibody with reduced affinity due to the loss of several hydrogen-bonding interactions. The insights obtained from the study are crucial to interpret the increased transmissibility and reduced neutralization efficacy of rapidly emerging SARS-CoV2 VOCs.
Collapse
|
100
|
Nathan N, Prevost B, Lambert S, Schnuriger A, Corvol H. Severe Acute Respiratory Syndrome Coronavirus 2 Variant Delta Infects All 6 Siblings but Spares Comirnaty (BNT162b2, BioNTech/Pfizer)-Vaccinated Parents. J Infect Dis 2021; 224:1984-1986. [PMID: 34409999 PMCID: PMC8499735 DOI: 10.1093/infdis/jiab410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/18/2021] [Indexed: 01/31/2023] Open
Affiliation(s)
- Nadia Nathan
- Paediatric Pulmonology Department and Reference Centre for Rare Lung Disease RespiRare, Trousseau Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Blandine Prevost
- Paediatric Pulmonology Department and Reference Centre for Rare Lung Disease RespiRare, Trousseau Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Sidonie Lambert
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP), Paris, France.,Virology Department, Trousseau and Saint Antoine Hospitals, AP-HP, Paris, France
| | - Aurélie Schnuriger
- Virology Department, Trousseau and Saint Antoine Hospitals, AP-HP, Paris, France.,Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Harriet Corvol
- Paediatric Pulmonology Department and Reference Centre for Rare Lung Disease RespiRare, Trousseau Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|