151
|
Serna García G, Al Khalaf R, Invernici F, Ceri S, Bernasconi A. CoVEffect: interactive system for mining the effects of SARS-CoV-2 mutations and variants based on deep learning. Gigascience 2022; 12:giad036. [PMID: 37222749 PMCID: PMC10205000 DOI: 10.1093/gigascience/giad036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Literature about SARS-CoV-2 widely discusses the effects of variations that have spread in the past 3 years. Such information is dispersed in the texts of several research articles, hindering the possibility of practically integrating it with related datasets (e.g., millions of SARS-CoV-2 sequences available to the community). We aim to fill this gap, by mining literature abstracts to extract-for each variant/mutation-its related effects (in epidemiological, immunological, clinical, or viral kinetics terms) with labeled higher/lower levels in relation to the nonmutated virus. RESULTS The proposed framework comprises (i) the provisioning of abstracts from a COVID-19-related big data corpus (CORD-19) and (ii) the identification of mutation/variant effects in abstracts using a GPT2-based prediction model. The above techniques enable the prediction of mutations/variants with their effects and levels in 2 distinct scenarios: (i) the batch annotation of the most relevant CORD-19 abstracts and (ii) the on-demand annotation of any user-selected CORD-19 abstract through the CoVEffect web application (http://gmql.eu/coveffect), which assists expert users with semiautomated data labeling. On the interface, users can inspect the predictions and correct them; user inputs can then extend the training dataset used by the prediction model. Our prototype model was trained through a carefully designed process, using a minimal and highly diversified pool of samples. CONCLUSIONS The CoVEffect interface serves for the assisted annotation of abstracts, allowing the download of curated datasets for further use in data integration or analysis pipelines. The overall framework can be adapted to resolve similar unstructured-to-structured text translation tasks, which are typical of biomedical domains.
Collapse
Affiliation(s)
- Giuseppe Serna García
- Dipartimento di Informazione, Elettronica e Bioingegneria, 20133 Milano Country: Italy, Italy
| | - Ruba Al Khalaf
- Dipartimento di Informazione, Elettronica e Bioingegneria, 20133 Milano Country: Italy, Italy
| | - Francesco Invernici
- Dipartimento di Informazione, Elettronica e Bioingegneria, 20133 Milano Country: Italy, Italy
| | - Stefano Ceri
- Dipartimento di Informazione, Elettronica e Bioingegneria, 20133 Milano Country: Italy, Italy
| | - Anna Bernasconi
- Dipartimento di Informazione, Elettronica e Bioingegneria, 20133 Milano Country: Italy, Italy
| |
Collapse
|
152
|
Feng Y, Zhao X, Yin Z, Wu C, Chen Z, Nie K, A R, Li L, Niu P, Wang J, Wu Y, Wang S, Wang D, Tan W, Wang H, Ma X, Gao GF, Chen C, Xu W, Xu W, National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China. Surveillance and Analysis of SARS-CoV-2 Variant Importation - China, January-June 2022. China CDC Wkly 2022; 4:1136-1142. [PMID: 36751558 PMCID: PMC9897968 DOI: 10.46234/ccdcw2022.229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant is the dominant circulating strain worldwide. To assess the importation of SARS-CoV-2 variants in the mainland of China during the Omicron epidemic, the genomic surveillance data of SARS-CoV-2 from imported coronavirus disease 2019 (COVID-19) cases in the mainland of China during the first half of 2022 were analyzed. Methods Sequences submitted from January to July 2022, with a collection date before June 30, 2022, were incorporated. The proportions of SARS-CoV-2 variants as well as the relationships between the origin and destination of each Omicron imported case were analyzed. Results 4,946 sequences of imported cases were submitted from 27 provincial-level administrative divisions (PLADs), and the median submission interval was within 1 month after collection. In 3,851 Omicron sequences with good quality, 1 recombinant (XU) and 4 subvariants under monitoring (BA.4, BA.5, BA.2.12.1, and BA.2.13) were recorded, and 3 of them (BA.4, BA.5, and BA.2.12.1) caused local transmissions in the mainland of China later than that recorded in the surveillance. Omicron subvariants dominated in the first half of 2022 and shifted from BA.1 to BA.2 then to BA.4 and BA.5. The percentage of BA.2 in the imported SARS-CoV-2 surveillance data was far higher than that in the Global Initiative on Sharing All Influenza Data (GISAID). The imported cases from Hong Kong Special Administrative Region, China, accounted for 32.30% of Omicron cases sampled, and 98.71% of them were BA.2. Conclusions The Omicron variant showed the intra-Omicron evolution in the first half of 2022, and all of the Omicron subvariants were introduced into the mainland of China multiple times from multiple different locations.
Collapse
Affiliation(s)
- Yenan Feng
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China
| | - Xiang Zhao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zeyuan Yin
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Changcheng Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhixiao Chen
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kai Nie
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China
| | - Ruhan A
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lili Li
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peihua Niu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China
| | - Ji Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China
| | - Yuchao Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shiwen Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China
| | - Dayan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wenjie Tan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Huanyu Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xuejun Ma
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China
| | - George F. Gao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China
| | - Cao Chen
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,Cao Chen,
| | - Wenbo Xu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China,Wenbo Xu,
| | | | | |
Collapse
|
153
|
Bader W, Delerce J, Aherfi S, La Scola B, Colson P. Quasispecies Analysis of SARS-CoV-2 of 15 Different Lineages during the First Year of the Pandemic Prompts Scratching under the Surface of Consensus Genome Sequences. Int J Mol Sci 2022; 23:15658. [PMID: 36555300 PMCID: PMC9779826 DOI: 10.3390/ijms232415658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
The tremendous majority of SARS-CoV-2 genomic data so far neglected intra-host genetic diversity. Here, we studied SARS-CoV-2 quasispecies based on data generated by next-generation sequencing (NGS) of complete genomes. SARS-CoV-2 raw NGS data had been generated for nasopharyngeal samples collected between March 2020 and February 2021 by the Illumina technology on a MiSeq instrument, without prior PCR amplification. To analyze viral quasispecies, we designed and implemented an in-house Excel file (“QuasiS”) that can characterize intra-sample nucleotide diversity along the genomes using data of the mapping of NGS reads. We compared intra-sample genetic diversity and global genetic diversity available from Nextstrain. Hierarchical clustering of all samples based on the intra-sample genetic diversity was performed and visualized with the Morpheus web application. NGS mapping data from 110 SARS-CoV-2-positive respiratory samples characterized by a mean depth of 169 NGS reads/nucleotide position and for which consensus genomes that had been obtained were classified into 15 viral lineages were analyzed. Mean intra-sample nucleotide diversity was 0.21 ± 0.65%, and 5357 positions (17.9%) exhibited significant (>4%) diversity, in ≥2 genomes for 1730 (5.8%) of them. ORF10, spike, and N genes had the highest number of positions exhibiting diversity (0.56%, 0.34%, and 0.24%, respectively). Nine hot spots of intra-sample diversity were identified in the SARS-CoV-2 NSP6, NSP12, ORF8, and N genes. Hierarchical clustering delineated a set of six genomes of different lineages characterized by 920 positions exhibiting intra-sample diversity. In addition, 118 nucleotide positions (0.4%) exhibited diversity at both intra- and inter-patient levels. Overall, the present study illustrates that the SARS-CoV-2 consensus genome sequences are only an incomplete and imperfect representation of the entire viral population infecting a patient, and that quasispecies analysis may allow deciphering more accurately the viral evolutionary pathways.
Collapse
Affiliation(s)
- Wahiba Bader
- IHU Méditerranée Infection, 19–21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
| | - Jeremy Delerce
- IHU Méditerranée Infection, 19–21 Boulevard Jean Moulin, 13005 Marseille, France
| | - Sarah Aherfi
- IHU Méditerranée Infection, 19–21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille (AP-HM), 264 rue Saint-Pierre, 13005 Marseille, France
| | - Bernard La Scola
- IHU Méditerranée Infection, 19–21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille (AP-HM), 264 rue Saint-Pierre, 13005 Marseille, France
| | - Philippe Colson
- IHU Méditerranée Infection, 19–21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement (IRD), Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille (AP-HM), 264 rue Saint-Pierre, 13005 Marseille, France
| |
Collapse
|
154
|
Moen LV, Vollan HS, Bråte J, Hungnes O, Bragstad K. Molecular Epidemiology of the Norwegian SARS-CoV-2 Delta Lineage AY.63. Viruses 2022; 14:2734. [PMID: 36560738 PMCID: PMC9781678 DOI: 10.3390/v14122734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Extensive genomic surveillance has given great insights into the evolution of the SARS-CoV-2 virus and emerging variants. During the summer months of 2021, Norway was dominated by the Pango lineage AY.63 which is a sub-lineage of the highly transmissible Delta variant. Strikingly, AY.63 did not spread in other countries to any significant extent. AY.63 carried a key mutation, A222V, in the spike protein, as well as the deletion of three residues in nsp1. Although these mutations are close to functionally important areas, we did not find any evidence that they induced higher fitness compared to other Delta lineages. This variant was introduced to Norway at a time when there were low levels of SARS-CoV-2 and contact-reducing measures were relaxed, which probably explains why the lineage rose so quickly. Furthermore, we found that the lack of imports of AY.63 from other countries probably led to the eventual demise of the lineage in Norway.
Collapse
Affiliation(s)
| | | | | | | | - Karoline Bragstad
- Division of Infectious Disease Control and Environmental Health, Norwegian Institute of Public Health, 0213 Oslo, Norway
| |
Collapse
|
155
|
Zhang M, Chen Z, Zhou J, Zhao X, Chen Y, Sun Y, Liu Z, Gu W, Luo C, Fu X, Zhao X. An imported human case with the SARS-CoV-2 Omicron subvariant BA.2.75 in Yunnan Province, China. BIOSAFETY AND HEALTH 2022; 4:406-409. [PMID: 36320663 PMCID: PMC9613801 DOI: 10.1016/j.bsheal.2022.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
The Omicron variants spread rapidly worldwide after being initially detected in South Africa in November 2021. It showed increased transmissibility and immune evasion with far more amino acid mutations in the spike (S) protein than the previously circulating variants of concern (VOCs). Notably, on 15 July 2022, we monitored the first VOC / Omicron subvariant BA.2.75 in China from an imported case. Moreover, nowadays, this subvariant still is predominant in India. It has nine additional mutations in the S protein compared to BA.2, three of which (W152R, G446S, and R493Q reversion) might contribute to higher transmissibility and immune escape. This subvariant could cause wider spread and pose a threat to the global situation. Our timely reporting and continuous genomic analysis are essential to fully elucidate the characteristics of the subvariant BA.2.75 in the future.
Collapse
Affiliation(s)
- Meiling Zhang
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China,Corresponding authors: Yunnan Center for Disease Control and Prevention, Kunming 650022, China (M. Zhang and X. Fu); National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China (X. Zhao)
| | - Zhixiao Chen
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jienan Zhou
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China
| | - Xiaonan Zhao
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China
| | - Yaoyao Chen
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China
| | - Yanhong Sun
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China
| | - Zhaosheng Liu
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China
| | - Wenpeng Gu
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China
| | - Chunrui Luo
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China
| | - Xiaoqing Fu
- Yunnan Center for Disease Control and Prevention, Kunming 650022, China,Corresponding authors: Yunnan Center for Disease Control and Prevention, Kunming 650022, China (M. Zhang and X. Fu); National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China (X. Zhao)
| | - Xiang Zhao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China,Corresponding authors: Yunnan Center for Disease Control and Prevention, Kunming 650022, China (M. Zhang and X. Fu); National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China (X. Zhao)
| |
Collapse
|
156
|
Arora P, Zhang L, Nehlmeier I, Kempf A, Cossmann A, Dopfer-Jablonka A, Schulz SR, Jäck HM, Behrens GMN, Pöhlmann S, Hoffmann M. The effect of cilgavimab and neutralisation by vaccine-induced antibodies in emerging SARS-CoV-2 BA.4 and BA.5 sublineages. THE LANCET. INFECTIOUS DISEASES 2022; 22:1665-1666. [PMID: 36327999 PMCID: PMC9621698 DOI: 10.1016/s1473-3099(22)00693-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Prerna Arora
- Infection Biology Unit, German Primate Center, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Lu Zhang
- Infection Biology Unit, German Primate Center, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center, 37077 Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Anne Cossmann
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany; Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Alexandra Dopfer-Jablonka
- Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Hannover, Germany
| | - Sebastian R Schulz
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany; Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Georg M N Behrens
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany; Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Hannover, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany.
| |
Collapse
|
157
|
Vattiatio G, Lustig A, Maclaren OJ, Plank MJ. Modelling the dynamics of infection, waning of immunity and re-infection with the Omicron variant of SARS-CoV-2 in Aotearoa New Zealand. Epidemics 2022; 41:100657. [PMID: 36427472 PMCID: PMC9677563 DOI: 10.1016/j.epidem.2022.100657] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/15/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Aotearoa New Zealand experienced a wave of the Omicron variant of SARS-CoV-2 in 2022 with around 200 confirmed cases per 1000 people between January and May. Waning of infection-derived immunity means people become increasingly susceptible to re-infection with SARS-CoV-2 over time. We investigated a model that included waning of vaccine-derived and infection-derived immunity under scenarios representing different levels of behavioural change relative to the first Omicron wave. Because the durability of infection-derived immunity is a key uncertainty in epidemiological models, we investigated outcomes under different assumptions about the speed of waning. The model was used to provide scenarios to the New Zealand Government, helping to inform policy response and healthcare system preparedness ahead of the winter respiratory illness season. In all scenarios investigated, a second Omicron wave was projected to occur in the second half of 2022. The timing of the peak depended primarily on the speed of waning and was typically between August and November. The peak number of daily infections in the second Omicron wave was smaller than in the first Omicron wave. Peak hospital occupancy was also generally lower than in the first wave but was sensitive to the age distribution of infections. A scenario with increased contact rates in older groups had higher peak hospital occupancy than the first wave. Scenarios with relatively high transmission, whether a result of relaxation of control measures or voluntary behaviour change, did not necessarily lead to higher peaks. However, they generally resulted in more sustained healthcare demand (>250 hospital beds throughout the winter period). The estimated health burden of Covid-19 in the medium term is sensitive to the strength and durability of infection-derived and hybrid immunity against reinfection and severe illness, which are uncertain.
Collapse
Affiliation(s)
- Giorgia Vattiatio
- Department of Physics, University of Auckland, Auckland, New Zealand; School of Mathematics and Statistics, University of Canterbury, Christchurch, New Zealand
| | | | - Oliver J Maclaren
- Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | - Michael J Plank
- School of Mathematics and Statistics, University of Canterbury, Christchurch, New Zealand.
| |
Collapse
|
158
|
Chia TRT, Young BE, Chia PY. The Omicron-transformer: Rise of the subvariants in the age of vaccines. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2022. [DOI: 10.47102/annals-acadmedsg.2022294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Introduction: Omicron is the latest SARS-CoV-2 variant of concern, the pathogen that causes COVID-19. Since its emergence in late 2021, Omicron has displaced other circulating variants and caused successive waves of infection worldwide throughout 2022. Omicron is characterised by the rapid emergence of many subvariants and high rates of infection in people with vaccine- and/or infection-induced immunity. This review article will consolidate current knowledge regarding Omicron subvariants, the role of boosters, and future vaccine development. Method: This narrative review is based on a literature search using PubMed. Search terms related to Omicron were used and priority was given to published peer-reviewed articles over pre-prints. Results: Studies indicate that vaccinations and boosters are important to reduce disease severity, hospitalisation and death from Omicron. A variety of factors, such as differing host factors, circulating variants, and forces of infection, can influence the benefit of repeated booster administration. Next-generation bivalent vaccines have now been approved in some countries including Singapore and have demonstrated the ability to induce broad variant protection. Future third-generation vaccines involving mucosal vaccines and/or pan-sarbecovirus vaccines may provide broader and longer-lasting protection. Conclusion: Due to current high levels of vaccine- and infection-induced immunity, it is likely that rates of severe illness, hospitalisation, and death due to Omicron will continue to moderate. Nevertheless, the virus is ever-changing, and public health policies, especially those related to vaccinations, will also have to continually evolve and adapt as COVID-19 transitions to endemicity.
Keywords: Booster, COVID-19, infectious diseases, Omicron, vaccine
Collapse
Affiliation(s)
| | | | - Po Ying Chia
- National Centre for Infectious Diseases, Singapore
| |
Collapse
|
159
|
Jeong BS, Jeon JY, Lai CJ, Yun HY, Jung JU, Oh BH. Structural basis for the broad and potent cross-reactivity of an N501Y-centric antibody against sarbecoviruses. Front Immunol 2022; 13:1049867. [PMID: 36466915 PMCID: PMC9714666 DOI: 10.3389/fimmu.2022.1049867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022] Open
Abstract
More than 80% of SARS-CoV-2 variants, including Alpha and Omicron, contain an N501Y mutation in the receptor-binding domain (RBD) of the spike protein. The N501Y change is an adaptive mutation enabling tighter interaction with the human ACE2 receptor. We have developed a broadly neutralizing antibody (nAb), D27LEY, whose binding affinity was intentionally optimized for Y501. This N501Y-centric antibody not only interacts with the Y501-containing RBDs of SARS-CoV-2 variants, including Omicron, with pico- or subnanomolar binding affinity, but also binds tightly to the RBDs with a different amino acid at residue 501. The crystal structure of the Fab fragment of D27LEY bound to the RBD of the Alpha variant reveals that the Y501-containing loop adopts a ribbon-like topology and serves as a small but major epitope in which Y501 is a part of extensive intermolecular interactions. A hydrophobic cleft on the most conserved surface of the RBD core serves as another major binding epitope. These data explain the broad and potent cross-reactivity of this N501Y-centric antibody, and suggest that a vaccine antigenic component composed of the RBD core and a part of receptor-binding motif (RBM) containing tyrosine at residue 501 might elicit broad and potent humoral responses across sarbecoviruses.
Collapse
Affiliation(s)
- Bo-Seong Jeong
- Department of Biological Sciences, KAIST Institute for the Biocentury, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Joon Young Jeon
- Department of Protein Design, Therazyne, lnc., Daejeon, South Korea
| | - Chih-Jen Lai
- Cancer Biology Department, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | | | - Jae U. Jung
- Cancer Biology Department, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Byung-Ha Oh
- Department of Biological Sciences, KAIST Institute for the Biocentury, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
- Department of Protein Design, Therazyne, lnc., Daejeon, South Korea
| |
Collapse
|
160
|
Sanderson T. Taxonium, a web-based tool for exploring large phylogenetic trees. eLife 2022; 11:e82392. [PMID: 36377483 PMCID: PMC9704803 DOI: 10.7554/elife.82392] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
The COVID-19 pandemic has resulted in a step change in the scale of sequencing data, with more genomes of SARS-CoV-2 having been sequenced than any other organism on earth. These sequences reveal key insights when represented as a phylogenetic tree, which captures the evolutionary history of the virus, and allows the identification of transmission events and the emergence of new variants. However, existing web-based tools for exploring phylogenies do not scale to the size of datasets now available for SARS-CoV-2. We have developed Taxonium, a new tool that uses WebGL to allow the exploration of trees with tens of millions of nodes in the browser for the first time. Taxonium links each node to associated metadata and supports mutation-annotated trees, which are able to capture all known genetic variation in a dataset. It can either be run entirely locally in the browser, from a server-based backend, or as a desktop application. We describe insights that analysing a tree of five million sequences can provide into SARS-CoV-2 evolution, and provide a tool at cov2tree.org for exploring a public tree of more than five million SARS-CoV-2 sequences. Taxonium can be applied to any tree, and is available at taxonium.org, with source code at github.com/theosanderson/taxonium.
Collapse
|
161
|
Kopsidas I, Karagiannidou S, Kostaki EG, Kousi D, Douka E, Sfikakis PP, Moustakidis S, Kokkotis C, Tsaopoulos D, Tseti I, Zaoutis T, Paraskevis D. Global Distribution, Dispersal Patterns, and Trend of Several Omicron Subvariants of SARS-CoV-2 across the Globe. Trop Med Infect Dis 2022; 7:373. [PMID: 36422924 PMCID: PMC9698960 DOI: 10.3390/tropicalmed7110373] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 08/27/2023] Open
Abstract
Our study aims to describe the global distribution and dispersal patterns of the SARS-CoV-2 Omicron subvariants. Genomic surveillance data were extracted from the CoV-Spectrum platform, searching for BA.1*, BA.2*, BA.3*, BA.4*, and BA.5* variants by geographic region. BA.1* increased in November 2021 in South Africa, with a similar increase across all continents in early December 2021. BA.1* did not reach 100% dominance in all continents. The spread of BA.2*, first described in South Africa, differed greatly by geographic region, in contrast to BA.1*, which followed a similar global expansion, firstly occurring in Asia and subsequently in Africa, Europe, Oceania, and North and South America. BA.4* and BA.5* followed a different pattern, where BA.4* reached high proportions (maximum 60%) only in Africa. BA.5* is currently, by Mid-August 2022, the dominant strain, reaching almost 100% across Europe, which is the first continent aside from Africa to show increasing proportions, and Asia, the Americas, and Oceania are following. The emergence of new variants depends mostly on their selective advantage, translated as enhanced transmissibility and ability to invade people with existing immunity. Describing these patterns is useful for a better understanding of the epidemiology of the VOCs' transmission and for generating hypotheses about the future of emerging variants.
Collapse
Affiliation(s)
- Ioannis Kopsidas
- Center for Clinical Epidemiology and Outcomes Research (CLEO), 15451 Athens, Greece
| | | | - Evangelia Georgia Kostaki
- Department of Hygiene Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitra Kousi
- Center for Clinical Epidemiology and Outcomes Research (CLEO), 15451 Athens, Greece
| | - Eirini Douka
- National Public Health Organisation (NPHO), 15123 Athens, Greece
| | - Petros P. Sfikakis
- First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Christos Kokkotis
- Department of Physical Education and Sport Science, Democritus University of Thrace, 69100 Komotini, Greece
| | - Dimitrios Tsaopoulos
- Center for Research and Technology Hellas, Institute for Bio-Economy & Agri-Technology, 38333 Volos, Greece
| | | | - Theoklis Zaoutis
- National Public Health Organisation (NPHO), 15123 Athens, Greece
| | - Dimitrios Paraskevis
- National Public Health Organisation (NPHO), 15123 Athens, Greece
- Department of Hygiene Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
162
|
Halfmann PJ, Minor NR, Haddock III LA, Maddox R, Moreno GK, Braun KM, Baker DA, Riemersa KK, Prasad A, Alman KJ, Lambert MC, Florek K, Bateman A, Westergaard R, Safdar N, Andes DR, Kawaoka Y, Fida M, Yao JD, Friedrich TC, O’Connor DH. Evolution of a globally unique SARS-CoV-2 Spike E484T monoclonal antibody escape mutation in a persistently infected, immunocompromised individual. Virus Evol 2022; 9:veac104. [PMID: 37692895 PMCID: PMC10491860 DOI: 10.1093/ve/veac104] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/29/2022] [Accepted: 11/04/2022] [Indexed: 09/12/2023] Open
Abstract
Prolonged infections in immunocompromised individuals may be a source for novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) variants, particularly when both the immune system and antiviral therapy fail to clear the infection and enable within-host evolution. Here we describe a 486-day case of SARS-CoV-2 infection in an immunocompromised individual. Following monotherapy with the monoclonal antibody Bamlanivimab, the individual's virus acquired resistance, likely via the earliest known occurrence of Spike amino acid variant E484T. Recently, E484T has arisen again as a derivative of E484A in the Omicron Variant of Concern, supporting the hypothesis that prolonged infections can give rise to novel variants long before they become prevalent in the human population.
Collapse
Affiliation(s)
- Peter J Halfmann
- Department of Pathobiological Sciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53706, USA
| | - Nicholas R Minor
- Department of Pathology and Laboratory Medicine, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA
| | - Luis A Haddock III
- Department of Pathobiological Sciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53706, USA
| | - Robert Maddox
- Department of Pathology and Laboratory Medicine, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA
| | - Gage K Moreno
- Department of Pathology and Laboratory Medicine, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA
| | - Katarina M Braun
- Department of Pathobiological Sciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53706, USA
| | - David A Baker
- Department of Pathology and Laboratory Medicine, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA
| | - Kasen K Riemersa
- Department of Pathobiological Sciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53706, USA
| | - Ankur Prasad
- Division of Allergy, Pulmonary and Critical Care Medicine, School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281, USA
| | - Kirsten J Alman
- University of Wisconsin Division of Infectious Disease, Room 5275-07C, 1685 Highland Avenue, Madison, WI 53705, USA
| | - Matthew C Lambert
- University of Wisconsin Division of Infectious Disease, Room 5275-07C, 1685 Highland Avenue, Madison, WI 53705, USA
| | - Kelsey Florek
- Wisconsin State Laboratory of Hygiene, 2601 Agriculture Drive, PO Box 7996, Madison, WI 53707, USA
| | - Allen Bateman
- Wisconsin State Laboratory of Hygiene, 2601 Agriculture Drive, PO Box 7996, Madison, WI 53707, USA
| | - Ryan Westergaard
- Department of Medicine, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, USA
| | - Nasia Safdar
- Department of Medicine, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, USA
| | - David R Andes
- Department of Medicine, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53706, USA
| | - Madiha Fida
- Division of Infectious Diseases, Mayo Clinic, 200 First St. SW, Rochester, Rochester, Minnesota 55905, USA
| | - Joseph D Yao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53706, USA
| | - David H O’Connor
- Department of Pathology and Laboratory Medicine, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
163
|
Dadonaite B, Crawford KHD, Radford CE, Farrell AG, Yu TC, Hannon WW, Zhou P, Andrabi R, Burton DR, Liu L, Ho DD, Neher RA, Bloom JD. A pseudovirus system enables deep mutational scanning of the full SARS-CoV-2 spike. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.10.13.512056. [PMID: 36263061 PMCID: PMC9580381 DOI: 10.1101/2022.10.13.512056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A major challenge in understanding SARS-CoV-2 evolution is interpreting the antigenic and functional effects of emerging mutations in the viral spike protein. Here we describe a new deep mutational scanning platform based on non-replicative pseudotyped lentiviruses that directly quantifies how large numbers of spike mutations impact antibody neutralization and pseudovirus infection. We demonstrate this new platform by making libraries of the Omicron BA.1 and Delta spikes. These libraries each contain ~7000 distinct amino-acid mutations in the context of up to ~135,000 unique mutation combinations. We use these libraries to map escape mutations from neutralizing antibodies targeting the receptor binding domain, N-terminal domain, and S2 subunit of spike. Overall, this work establishes a high-throughput and safe approach to measure how ~10 5 combinations of mutations affect antibody neutralization and spike-mediated infection. Notably, the platform described here can be extended to the entry proteins of many other viruses.
Collapse
Affiliation(s)
- Bernadeta Dadonaite
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
| | - Katharine H D Crawford
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
- Department of Genome Sciences & Medical Scientist Training Program, University of Washington, Seattle, Washington, 98109, USA
| | - Caelan E Radford
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, and Basic Sciences Division, Fred Hutch Cancer Center, Seattle, Washington, 98109, USA
| | - Ariana G Farrell
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
| | - Timothy C Yu
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, and Basic Sciences Division, Fred Hutch Cancer Center, Seattle, Washington, 98109, USA
| | - William W Hannon
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, and Basic Sciences Division, Fred Hutch Cancer Center, Seattle, Washington, 98109, USA
| | - Panpan Zhou
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of MGH, MIT & Harvard, Cambridge, MA 02139, USA
| | - Lihong Liu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - David D. Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Richard A. Neher
- Biozentrum, University of Basel, Basel, Switzerland, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jesse D Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
- Howard Hughes Medical Institute, Seattle, WA, 98195, USA
| |
Collapse
|
164
|
Characterization of the enhanced infectivity and antibody evasion of Omicron BA.2.75. Cell Host Microbe 2022; 30:1527-1539.e5. [PMID: 36270286 PMCID: PMC9531665 DOI: 10.1016/j.chom.2022.09.018] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 01/09/2023]
Abstract
Recently emerged SARS-CoV-2 Omicron subvariant, BA.2.75, displayed a growth advantage over circulating BA.2.38, BA.2.76, and BA.5 in India. However, the underlying mechanisms for enhanced infectivity, especially compared with BA.5, remain unclear. Here, we show that BA.2.75 exhibits substantially higher affinity for host receptor angiotensin-converting enzyme 2 (ACE2) than BA.5 and other variants. Structural analyses of BA.2.75 spike shows its decreased thermostability and increased frequency of the receptor binding domain (RBD) in the "up" conformation under acidic conditions, suggesting enhanced low-pH-endosomal cell entry. Relative to BA.4/BA.5, BA.2.75 exhibits reduced evasion of humoral immunity from BA.1/BA.2 breakthrough-infection convalescent plasma but greater evasion of Delta breakthrough-infection convalescent plasma. BA.5 breakthrough-infection plasma also exhibits weaker neutralization against BA.2.75 than BA.5, mainly due to BA.2.75's distinct neutralizing antibody (NAb) escape pattern. Antibody therapeutics Evusheld and Bebtelovimab remain effective against BA.2.75. These results suggest BA.2.75 may prevail after BA.4/BA.5, and its increased receptor-binding capability could support further immune-evasive mutations.
Collapse
|
165
|
Gruell H, Vanshylla K, Tober-Lau P, Hillus D, Sander LE, Kurth F, Klein F. Neutralisation sensitivity of the SARS-CoV-2 omicron BA.2.75 sublineage. THE LANCET. INFECTIOUS DISEASES 2022; 22:1422-1423. [PMID: 36084664 PMCID: PMC9448327 DOI: 10.1016/s1473-3099(22)00580-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 01/19/2023]
Affiliation(s)
- Henning Gruell
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Kanika Vanshylla
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Pinkus Tober-Lau
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Hillus
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leif Erik Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Center for Regenerative Therapies, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner site Bonn-Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
166
|
Wang L, Zhou H, Li J, Cheng Y, Zhang S, Aliyari S, Wu A, Cheng G. Potential intervariant and intravariant recombination of Delta and Omicron variants. J Med Virol 2022; 94:4830-4838. [PMID: 35705528 PMCID: PMC9350351 DOI: 10.1002/jmv.27939] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/12/2022]
Abstract
Among numerous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concerns, Omicron is more infectious and immune-escaping, while Delta is more pathogenic. Here, we provide evidence for both intervariant and intravariant recombination of the rapidly evolving new SARS-CoV-2 genomes, including XD/XE/XF and BA.3, raising concerns of potential more infectious, immune-escaping, and disease-causing Omicron and Delta-Omicron variants.
Collapse
Affiliation(s)
- Lulan Wang
- Department of Microbiology, Immunology, and Molecular GeneticsUniversity of California, Los AngelesLos AngelesUSA
| | - Hang‐Yu Zhou
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- Suzhou Institute of Systems MedicineSuzhouJiangsuChina
| | - Jia‐Ying Li
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- Suzhou Institute of Systems MedicineSuzhouJiangsuChina
| | - Ye‐Xiao Cheng
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- Suzhou Institute of Systems MedicineSuzhouJiangsuChina
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Shilei Zhang
- Department of Microbiology, Immunology, and Molecular GeneticsUniversity of California, Los AngelesLos AngelesUSA
| | - Saba Aliyari
- Department of Microbiology, Immunology, and Molecular GeneticsUniversity of California, Los AngelesLos AngelesUSA
| | - Aiping Wu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- Suzhou Institute of Systems MedicineSuzhouJiangsuChina
| | - Genhong Cheng
- Department of Microbiology, Immunology, and Molecular GeneticsUniversity of California, Los AngelesLos AngelesUSA
| |
Collapse
|
167
|
Kim M, Opsasnick L, Batio S, Benavente JY, Zheng P, Lovett RM, Bailey SC, Kwasny MJ, Ladner DP, Chou SH, Linder JA, Weintraub S, Luo Y, Zee PC, Wolf MS. Prevalence and risk factors of sleep disturbance in adults with underlying health conditions during the ongoing COVID-19 pandemic. Medicine (Baltimore) 2022; 101:e30637. [PMID: 36123887 PMCID: PMC9477708 DOI: 10.1097/md.0000000000030637] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To determine the prevalence of sleep disturbance during the coronavirus disease 2019 (COVID-19) pandemic among US adults who are more vulnerable to complications because of age and co-morbid conditions, and to identify associated sociodemographic and psychosocial factors. Cross-sectional survey linked to 3 active clinical trials and 2 cohort studies, conducted between 11/30/2020 and 3/3/2021. Five academic internal medicine practices and 2 federally qualified health centers. A total of 715 adults ages 23 to 91 years living with one or more chronic conditions. A fifth (20%) of participants reported poor sleep. Black adults were twice as likely to report poor sleep compared to Whites. Self-reported poor physical function (51%), stress (42%), depression (28%), and anxiety (36%) were also common and all significantly associated with poor sleep. Age ≥70 years and having been vaccinated for COVID-19 were protective against poor sleep. Sex, education, income, alcohol use, and employment status were not significantly associated with sleep quality. In this diverse sample of adults with chronic conditions, by race, ethnicity, and socioeconomic status, disparities in sleep health amid the ongoing pandemic were apparent. Worse physical function and mental health were associated with poor sleep and should be considered targets for health system interventions to prevent the many subsequent consequences of disturbed sleep on health outcomes. Measurements: self-reported sleep quality, physical function, stress, depression, and anxiety.
Collapse
Affiliation(s)
- Minjee Kim
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- Center for Circadian and Sleep Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- * Correspondence: Minjee Kim, Department of Neurology, Feinberg School of Medicine, Northwestern University, 625 N. Michigan Avenue Suite 1150, Chicago IL 60611, USA (e-mail: )
| | - Lauren Opsasnick
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Stephanie Batio
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Julia Y. Benavente
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Pauline Zheng
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Rebecca M. Lovett
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Stacy C. Bailey
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Mary J. Kwasny
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Daniela P. Ladner
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Sherry H.Y. Chou
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Jeffrey A. Linder
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Sandra Weintraub
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Phyllis C. Zee
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
- Center for Circadian and Sleep Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Michael S. Wolf
- Division of General Internal Medicine and Geriatrics, Center for Applied Health Research on Aging (CAHRA), Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| |
Collapse
|
168
|
Pastorio C, Zech F, Noettger S, Jung C, Jacob T, Sanderson T, Sparrer KMJ, Kirchhoff F. Determinants of Spike infectivity, processing, and neutralization in SARS-CoV-2 Omicron subvariants BA.1 and BA.2. Cell Host Microbe 2022; 30:1255-1268.e5. [PMID: 35931073 PMCID: PMC9289044 DOI: 10.1016/j.chom.2022.07.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/29/2022] [Accepted: 07/12/2022] [Indexed: 12/02/2022]
Abstract
SARS-CoV-2 Omicron rapidly outcompeted other variants and currently dominates the COVID-19 pandemic. Its enhanced transmission and immune evasion are thought to be driven by numerous mutations in the Omicron Spike protein. Here, we systematically introduced BA.1 and/or BA.2 Omicron Spike mutations into the ancestral Spike protein and examined the impacts on Spike function, processing, and susceptibility to neutralization. Individual mutations of S371F/L, S375F, and T376A in the ACE2-receptor-binding domain as well as Q954H and N969K in the hinge region 1 impaired infectivity, while changes to G339D, D614G, N764K, and L981F moderately enhanced it. Most mutations in the N-terminal region and receptor-binding domain reduced the sensitivity of the Spike protein to neutralization by sera from individuals vaccinated with the BNT162b2 vaccine and by therapeutic antibodies. Our results represent a systematic functional analysis of Omicron Spike adaptations that have allowed this SARS-CoV-2 variant to dominate the current pandemic.
Collapse
Affiliation(s)
- Chiara Pastorio
- Institute of Molecular Virology, Ulm University Medical Centre, 89081 Ulm, Germany
| | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Centre, 89081 Ulm, Germany
| | - Sabrina Noettger
- Institute of Molecular Virology, Ulm University Medical Centre, 89081 Ulm, Germany
| | - Christoph Jung
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany; Electrochemical Energy Storage, Helmholtz-Institute-Ulm (HIU), 89081 Ulm, Germany; Karlsruhe Institute of Technology (KIT), 76344 Karlsruhe, Germany
| | - Timo Jacob
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany
| | | | | | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Centre, 89081 Ulm, Germany.
| |
Collapse
|
169
|
Wittig A, Miranda F, Hölzer M, Altenburg T, Bartoszewicz JM, Beyvers S, Dieckmann MA, Genske U, Giese SH, Nowicka M, Richard H, Schiebenhoefer H, Schmachtenberg AJ, Sieben P, Tang M, Tembrockhaus J, Renard BY, Fuchs S. CovRadar: continuously tracking and filtering SARS-CoV-2 mutations for genomic surveillance. Bioinformatics 2022; 38:4223-4225. [PMID: 35799354 DOI: 10.1093/bioinformatics/btac411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/13/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
SUMMARY The ongoing pandemic caused by SARS-CoV-2 emphasizes the importance of genomic surveillance to understand the evolution of the virus, to monitor the viral population, and plan epidemiological responses. Detailed analysis, easy visualization and intuitive filtering of the latest viral sequences are powerful for this purpose. We present CovRadar, a tool for genomic surveillance of the SARS-CoV-2 Spike protein. CovRadar consists of an analytical pipeline and a web application that enable the analysis and visualization of hundreds of thousand sequences. First, CovRadar extracts the regions of interest using local alignment, then builds a multiple sequence alignment, infers variants and consensus and finally presents the results in an interactive app, making accessing and reporting simple, flexible and fast. AVAILABILITY AND IMPLEMENTATION CovRadar is freely accessible at https://covradar.net, its open-source code is available at https://gitlab.com/dacs-hpi/covradar. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Alice Wittig
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany.,Methods Development, Research Infrastructure and Information Technology (MFI), Bioinformatics and Systems Biology, Robert Koch Institute, Berlin, Germany
| | - Fábio Miranda
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Martin Hölzer
- Methods Development, Research Infrastructure and Information Technology (MFI), Bioinformatics and Systems Biology, Robert Koch Institute, Berlin, Germany
| | - Tom Altenburg
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Jakub M Bartoszewicz
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany.,Methods Development, Research Infrastructure and Information Technology (MFI), Bioinformatics and Systems Biology, Robert Koch Institute, Berlin, Germany
| | - Sebastian Beyvers
- Department of Biology and Chemistry, Justus-Liebig-University Gießen, Gießen 35390, Germany
| | - Marius A Dieckmann
- Department of Biology and Chemistry, Justus-Liebig-University Gießen, Gießen 35390, Germany
| | - Ulrich Genske
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Sven H Giese
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Melania Nowicka
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Hugues Richard
- Methods Development, Research Infrastructure and Information Technology (MFI), Bioinformatics and Systems Biology, Robert Koch Institute, Berlin, Germany
| | - Henning Schiebenhoefer
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | | | - Paul Sieben
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Ming Tang
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany.,Department of Human Genetics, Hannover Medical School, Hannover 30625, Germany
| | - Julius Tembrockhaus
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Bernhard Y Renard
- Digital Engineering Faculty, Hasso Plattner Institute, University of Potsdam, Potsdam 14482, Germany
| | - Stephan Fuchs
- Methods Development, Research Infrastructure and Information Technology (MFI), Bioinformatics and Systems Biology, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
170
|
Merhi G, Koweyes J, Salloum T, Khoury CA, Haidar S, Tokajian S. SARS-CoV-2 genomic epidemiology: data and sequencing infrastructure. Future Microbiol 2022; 17:1001-1007. [PMID: 35899481 PMCID: PMC9332909 DOI: 10.2217/fmb-2021-0207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 06/15/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Genomic surveillance of SARS-CoV-2 is critical in monitoring viral lineages. Available data reveal a significant gap between low- and middle-income countries and the rest of the world. Methods: The SARS-CoV-2 sequencing costs using the Oxford Nanopore MinION device and hardware prices for data computation in Lebanon were estimated and compared with those in developed countries. SARS-CoV-2 genomes deposited on the Global Initiative on Sharing All Influenza Data per 1000 COVID-19 cases were determined per country. Results: Sequencing costs in Lebanon were significantly higher compared with those in developed countries. Low- and middle-income countries showed limited sequencing capabilities linked to the lack of support, high prices, long delivery delays and limited availability of trained personnel. Conclusion: The authors recommend the mobilization of funds to develop whole-genome sequencing-based surveillance platforms and the implementation of genomic epidemiology to better identify and track outbreaks, leading to appropriate and mindful interventions.
Collapse
Affiliation(s)
- Georgi Merhi
- Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Byblos, Lebanon
| | - Jad Koweyes
- Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Byblos, Lebanon
| | - Tamara Salloum
- Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Byblos, Lebanon
| | - Charbel Al Khoury
- Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Byblos, Lebanon
| | - Siwar Haidar
- Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Byblos, Lebanon
| | - Sima Tokajian
- Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
171
|
Jian F, Yu Y, Song W, Yisimayi A, Yu L, Gao Y, Zhang N, Wang Y, Shao F, Hao X, Xu Y, Jin R, Wang Y, Xie XS, Cao Y. Further humoral immunity evasion of emerging SARS-CoV-2 BA.4 and BA.5 subvariants. THE LANCET INFECTIOUS DISEASES 2022; 22:1535-1537. [PMID: 36179744 PMCID: PMC9514837 DOI: 10.1016/s1473-3099(22)00642-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 10/31/2022]
|
172
|
Arora P, Nehlmeier I, Kempf A, Cossmann A, Schulz SR, Dopfer-Jablonka A, Baier E, Tampe B, Moerer O, Dickel S, Winkler MS, Jäck HM, Behrens GMN, Pöhlmann S, Hoffmann M. Lung cell entry, cell–cell fusion capacity, and neutralisation sensitivity of omicron sublineage BA.2.75. THE LANCET INFECTIOUS DISEASES 2022; 22:1537-1538. [PMID: 36116462 PMCID: PMC9477470 DOI: 10.1016/s1473-3099(22)00591-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/12/2022]
|
173
|
Loucera C, Perez-Florido J, Casimiro-Soriguer CS, Ortuño FM, Carmona R, Bostelmann G, Martínez-González LJ, Muñoyerro-Muñiz D, Villegas R, Rodriguez-Baño J, Romero-Gomez M, Lorusso N, Garcia-León J, Navarro-Marí JM, Camacho-Martinez P, Merino-Diaz L, de Salazar A, Viñuela L, The Andalusian COVID-19 Sequencing Initiative, Lepe JA, Garcia F, Dopazo J. Assessing the Impact of SARS-CoV-2 Lineages and Mutations on Patient Survival. Viruses 2022; 14:1893. [PMID: 36146700 PMCID: PMC9500738 DOI: 10.3390/v14091893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES More than two years into the COVID-19 pandemic, SARS-CoV-2 still remains a global public health problem. Successive waves of infection have produced new SARS-CoV-2 variants with new mutations for which the impact on COVID-19 severity and patient survival is uncertain. METHODS A total of 764 SARS-CoV-2 genomes, sequenced from COVID-19 patients, hospitalized from 19th February 2020 to 30 April 2021, along with their clinical data, were used for survival analysis. RESULTS A significant association of B.1.1.7, the alpha lineage, with patient mortality (log hazard ratio (LHR) = 0.51, C.I. = [0.14,0.88]) was found upon adjustment by all the covariates known to affect COVID-19 prognosis. Moreover, survival analysis of mutations in the SARS-CoV-2 genome revealed 27 of them were significantly associated with higher mortality of patients. Most of these mutations were located in the genes coding for the S, ORF8, and N proteins. CONCLUSIONS This study illustrates how a combination of genomic and clinical data can provide solid evidence for the impact of viral lineage on patient survival.
Collapse
Affiliation(s)
- Carlos Loucera
- Bioinformatics Area, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Sevilla, Spain
| | - Javier Perez-Florido
- Bioinformatics Area, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Sevilla, Spain
| | - Carlos S. Casimiro-Soriguer
- Bioinformatics Area, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Sevilla, Spain
| | - Francisco M. Ortuño
- Bioinformatics Area, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
- Department of Computer Architecture and Computer Technology, University of Granada, 18011 Granada, Spain
| | - Rosario Carmona
- Bioinformatics Area, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
| | - Gerrit Bostelmann
- Bioinformatics Area, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
| | - L. Javier Martínez-González
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain
| | - Dolores Muñoyerro-Muñiz
- Subdirección Técnica Asesora de Gestión de la Información, Servicio Andaluz de Salud, 41001 Sevilla, Spain
| | - Román Villegas
- Subdirección Técnica Asesora de Gestión de la Información, Servicio Andaluz de Salud, 41001 Sevilla, Spain
| | - Jesus Rodriguez-Baño
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Sevilla, Spain
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain
- Departamento de Medicina, Universidad de Sevilla, C. San Fernando, 4, 41004 Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), ISCIII, 28029 Madrid, Spain
| | - Manuel Romero-Gomez
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Sevilla, Spain
- Departamento de Medicina, Universidad de Sevilla, C. San Fernando, 4, 41004 Sevilla, Spain
- Servicio de Aparato Digestivo, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Nicola Lorusso
- Dirección General de Salud Pública, Consejería de Salud y Familias, Junta de Andalucía, 41020 Sevilla, Spain
| | - Javier Garcia-León
- Departamento de Metafísica y Corrientes Actuales de la Filosofía, Ética y Filosofía Política, Universidad de Sevilla, 41004 Sevilla, Spain
| | - Jose M. Navarro-Marí
- Servicio de Microbiología, Hospital Virgen de las Nieves, 18014 Granada, Spain
- Instituto de Investigación Biosanitaria, ibs.GRANADA, 18012 Granada, Spain
| | - Pedro Camacho-Martinez
- Servicio de Microbiología, Unidad Clínica Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Laura Merino-Diaz
- Servicio de Microbiología, Unidad Clínica Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Adolfo de Salazar
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), ISCIII, 28029 Madrid, Spain
- Servicio de Microbiología, Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Laura Viñuela
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), ISCIII, 28029 Madrid, Spain
- Servicio de Microbiología, Hospital Universitario San Cecilio, 18016 Granada, Spain
| | | | - Jose A. Lepe
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), ISCIII, 28029 Madrid, Spain
- Servicio de Microbiología, Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Federico Garcia
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Biosanitaria, ibs.GRANADA, 18012 Granada, Spain
- Servicio de Microbiología, Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Joaquin Dopazo
- Bioinformatics Area, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Sevilla, Spain
- FPS/ELIXIR-ES, Andalusian Public Foundation Progress and Health-FPS, 41013 Sevilla, Spain
| |
Collapse
|
174
|
Chaqroun A, Hartard C, Josse T, Taverniers A, Jeulin H, Gantzer C, Murray JM, Obepine Consortium, Bertrand I, Schvoerer E. SARS-CoV-2 Variability in Patients and Wastewaters—Potential Immuno-Modulation during the Shift from Delta to Omicron. Biomedicines 2022; 10:biomedicines10092080. [PMID: 36140181 PMCID: PMC9496010 DOI: 10.3390/biomedicines10092080] [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: 07/23/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The continuous emergence of SARS-CoV-2 variants favors potential co-infections and/or viral mutation events, leading to possible new biological properties. The aim of this work was to characterize SARS-CoV-2 genetic variability during the Delta–Omicron shift in patients and in a neighboring wastewater treatment plant (WWTP) in the same urban area. The surveillance of SARS-CoV-2 was performed by routine screening of positive samples by single nucleotide polymorphism analysis within the S gene. Moreover, additionally to national systematic whole genome sequencing (WGS) once a week in SARS-CoV-2-positive patients, WGS was also applied when mutational profiles were difficult to interpret by routine screening. Thus, WGS was performed on 414 respiratory samples and on four wastewater samples, northeastern France. This allowed us to report (i) the temporally concordant Delta to Omicron viral shift in patients and wastewaters; (ii) the characterization of 21J (Delta) and 21K (Omicron)/BA.1-21L (Omicron)/BA.2-BA.4 mixtures from humans or environmental samples; (iii) the mapping of composite mutations and the predicted impact on immune properties in the viral Spike protein.
Collapse
Affiliation(s)
- Ahlam Chaqroun
- CNRS, LCPME, Université de Lorraine, F-54000 Nancy, France
- Laboratoire de Virologie-Microbiologie, Hôpital Universitaire de Nancy, Université de Lorraine, F-54000 Nancy, France
| | - Cédric Hartard
- CNRS, LCPME, Université de Lorraine, F-54000 Nancy, France
- Laboratoire de Virologie-Microbiologie, Hôpital Universitaire de Nancy, Université de Lorraine, F-54000 Nancy, France
| | - Thomas Josse
- Laboratoire de Virologie-Microbiologie, Hôpital Universitaire de Nancy, Université de Lorraine, F-54000 Nancy, France
| | - Audrey Taverniers
- Laboratoire de Virologie-Microbiologie, Hôpital Universitaire de Nancy, Université de Lorraine, F-54000 Nancy, France
| | - Hélène Jeulin
- CNRS, LCPME, Université de Lorraine, F-54000 Nancy, France
- Laboratoire de Virologie-Microbiologie, Hôpital Universitaire de Nancy, Université de Lorraine, F-54000 Nancy, France
| | | | - John M. Murray
- School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW 2052, Australia
| | | | | | - Evelyne Schvoerer
- CNRS, LCPME, Université de Lorraine, F-54000 Nancy, France
- Laboratoire de Virologie-Microbiologie, Hôpital Universitaire de Nancy, Université de Lorraine, F-54000 Nancy, France
- Correspondence: or
| |
Collapse
|
175
|
Li YT, Polotan FGM, Sotelo GIS, Alpino APA, Dolor AYM, Tujan MAA, Gomez MRR, Onza OJT, Chang AKT, Bautista CT, Carandang JC, Yangzon MSL, Pangilinan EAR, Mantaring RJ, Telles AJE, Egana JMC, Endozo JJS, Cruz RPS, Tablizo FA, Yap JMC, Maralit BA, Ayes MEC, de la Paz EMC, Saloma CP, Lim DR, Dancel LLM, Uy-Lumandas M, Medado IAP, Dizon TJR, Hampson K, Daldry S, Hughes J, Brunker K. Lineage BA.2 dominated the Omicron SARS-CoV-2 epidemic wave in the Philippines. Virus Evol 2022; 8:veac078. [PMID: 36090771 PMCID: PMC9452094 DOI: 10.1093/ve/veac078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/11/2022] [Accepted: 08/18/2022] [Indexed: 11/14/2022] Open
Abstract
The Omicron severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant led to a dramatic global epidemic wave following detection in South Africa in November 2021. The BA.1 Omicron lineage was dominant and responsible for most SARS-CoV-2 outbreaks in countries around the world during December 2021-January 2022, while other Omicron lineages, including BA.2, accounted for the minority of global isolates. Here, we describe the Omicron wave in the Philippines by analysing genomic data. Our results identify the presence of both BA.1 and BA.2 lineages in the Philippines in December 2021, before cases surged in January 2022. We infer that only the BA.2 lineage underwent sustained transmission in the country, with an estimated emergence around 18 November 2021 (95 per cent highest posterior density: 6-28 November), while despite multiple introductions, BA.1 transmission remained limited. These results suggest that the Philippines was one of the earliest areas affected by BA.2 and reiterate the importance of whole genome sequencing for monitoring outbreaks.
Collapse
Affiliation(s)
- Yao-Tsun Li
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Francisco Gerardo M Polotan
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Gerald Ivan S Sotelo
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Anne Pauline A Alpino
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Ardiane Ysabelle M Dolor
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Ma. Angelica A Tujan
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Ma. Ricci R Gomez
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Othoniel Jan T Onza
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Angela Kae T Chang
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Criselda T Bautista
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - June C Carandang
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Maria Sofia L Yangzon
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Elcid Aaron R Pangilinan
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Renato Jacinto Mantaring
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Alyssa Joyce E Telles
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - John Michael C Egana
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Joshua Jose S Endozo
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Rianna Patricia S Cruz
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Francis A Tablizo
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Jan Michael C Yap
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Benedict A Maralit
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Marc Edsel C Ayes
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Eva Marie C de la Paz
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
- National Institutes of Health, University of the Philippines Manila, 623 Pedro Gil Street, Ermita, Manila 1000, Philippine
| | - Cynthia P Saloma
- Philippine Genome Center, National Science Complex, U.P. Campus, University of the Philippines, A. Ma. Regidor Street, Quezon City, Metro Manila 1101, Philippines
| | - Dodge R Lim
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Lei Lanna M Dancel
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Mayan Uy-Lumandas
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Inez Andrea P Medado
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Timothy John R Dizon
- Research Institute for Tropical Medicine, 9002 Research Drive, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila 1781, Philippines
| | - Katie Hampson
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Simon Daldry
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Kirstyn Brunker
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
176
|
Mushegian AA, Long SW, Olsen RJ, Christensen PA, Subedi S, Chung M, Davis J, Musser J, Ghedin E. Within-host genetic diversity of SARS-CoV-2 in the context of large-scale hospital-associated genomic surveillance. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.08.17.22278898. [PMID: 36032964 PMCID: PMC9413716 DOI: 10.1101/2022.08.17.22278898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The COVID-19 pandemic has resulted in extensive surveillance of the genomic diversity of SARS-CoV-2. Sequencing data generated as part of these efforts can also capture the diversity of the SARS-CoV-2 virus populations replicating within infected individuals. To assess this within-host diversity of SARS-CoV-2 we quantified low frequency (minor) variants from deep sequence data of thousands of clinical samples collected by a large urban hospital system over the course of a year. Using a robust analytical pipeline to control for technical artefacts, we observe that at comparable viral loads, specimens from patients hospitalized due to COVID-19 had a greater number of minor variants than samples from outpatients. Since individuals with highly diverse viral populations could be disproportionate drivers of new viral lineages in the patient population, these results suggest that transmission control should pay special attention to patients with severe or protracted disease to prevent the spread of novel variants.
Collapse
Affiliation(s)
- Alexandra A. Mushegian
- Systems Genomics Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Scott W. Long
- Laboratory of Molecular and Translational Human Infectious Diseases Research, Center for Infectious Diseases, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital Houston, Texas, 77030
| | - Randall J. Olsen
- Laboratory of Molecular and Translational Human Infectious Diseases Research, Center for Infectious Diseases, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital Houston, Texas, 77030
| | - Paul A. Christensen
- Laboratory of Molecular and Translational Human Infectious Diseases Research, Center for Infectious Diseases, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital Houston, Texas, 77030
| | - Sishir Subedi
- Laboratory of Molecular and Translational Human Infectious Diseases Research, Center for Infectious Diseases, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital Houston, Texas, 77030
| | - Matthew Chung
- Systems Genomics Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - James Davis
- Division of Data Science and Learning, Argonne National Laboratory, 9700 S. Cass Ave., Lemont, Illinois, 60439
- University of Chicago Consortium for Advanced Science and Engineering, 5801 South Ellis Avenue, Chicago, Illinois, 60637
| | - James Musser
- Laboratory of Molecular and Translational Human Infectious Diseases Research, Center for Infectious Diseases, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital Houston, Texas, 77030
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
177
|
Pham D, Maddocks S, Dwyer DE, Sintchenko V, Kok J, Rockett RJ. Development and Validation of an In-House Real-Time Reverse-Transcriptase Polymerase Chain Reaction Assay for SARS-CoV-2 Omicron Lineage Subtyping between BA.1 and BA.2. Viruses 2022; 14:v14081760. [PMID: 36016382 PMCID: PMC9416591 DOI: 10.3390/v14081760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022] Open
Abstract
In order to rapidly differentiate sublineages BA.1 and BA.2 of the SARS-CoV-2 variant of concern Omicron, we developed a real-time reverse-transcriptase polymerase chain reaction to target the discriminatory spike protein deletion at amino acid position 69–70 (S:del69–70). Compared to the gold standard of whole genome sequencing, the candidate assay was 100% sensitive and 99.4% specific. Sublineage typing by RT-PCR can provide a rapid, high throughput and cost-effective method to enhance surveillance as well as potentially guiding treatment and infection control decisions.
Collapse
Affiliation(s)
- David Pham
- Centre for Infectious Diseases & Microbiology Laboratory Services, New South Wales Health, Pathology—Institute of Clinical Pathology & Medical Research, Westmead Hospital, Westmead, NSW 2145, Australia
- Correspondence:
| | - Susan Maddocks
- Centre for Infectious Diseases & Microbiology Laboratory Services, New South Wales Health, Pathology—Institute of Clinical Pathology & Medical Research, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Dominic E. Dwyer
- Centre for Infectious Diseases & Microbiology Laboratory Services, New South Wales Health, Pathology—Institute of Clinical Pathology & Medical Research, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Centre for Infectious Diseases & Microbiology—Public Health, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Vitali Sintchenko
- Centre for Infectious Diseases & Microbiology Laboratory Services, New South Wales Health, Pathology—Institute of Clinical Pathology & Medical Research, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Centre for Infectious Diseases & Microbiology—Public Health, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Jen Kok
- Centre for Infectious Diseases & Microbiology Laboratory Services, New South Wales Health, Pathology—Institute of Clinical Pathology & Medical Research, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Centre for Infectious Diseases & Microbiology—Public Health, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Rebecca J. Rockett
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Centre for Infectious Diseases & Microbiology—Public Health, Westmead Hospital, Westmead, NSW 2145, Australia
| |
Collapse
|
178
|
Jahn K, Dreifuss D, Topolsky I, Kull A, Ganesanandamoorthy P, Fernandez-Cassi X, Bänziger C, Devaux AJ, Stachler E, Caduff L, Cariti F, Corzón AT, Fuhrmann L, Chen C, Jablonski KP, Nadeau S, Feldkamp M, Beisel C, Aquino C, Stadler T, Ort C, Kohn T, Julian TR, Beerenwinkel N. Early detection and surveillance of SARS-CoV-2 genomic variants in wastewater using COJAC. Nat Microbiol 2022; 7:1151-1160. [PMID: 35851854 PMCID: PMC9352586 DOI: 10.1038/s41564-022-01185-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 06/23/2022] [Indexed: 01/12/2023]
Abstract
The continuing emergence of SARS-CoV-2 variants of concern and variants of interest emphasizes the need for early detection and epidemiological surveillance of novel variants. We used genomic sequencing of 122 wastewater samples from three locations in Switzerland to monitor the local spread of B.1.1.7 (Alpha), B.1.351 (Beta) and P.1 (Gamma) variants of SARS-CoV-2 at a population level. We devised a bioinformatics method named COJAC (Co-Occurrence adJusted Analysis and Calling) that uses read pairs carrying multiple variant-specific signature mutations as a robust indicator of low-frequency variants. Application of COJAC revealed that a local outbreak of the Alpha variant in two Swiss cities was observable in wastewater up to 13 d before being first reported in clinical samples. We further confirmed the ability of COJAC to detect emerging variants early for the Delta variant by analysing an additional 1,339 wastewater samples. While sequencing data of single wastewater samples provide limited precision for the quantification of relative prevalence of a variant, we show that replicate and close-meshed longitudinal sequencing allow for robust estimation not only of the local prevalence but also of the transmission fitness advantage of any variant. We conclude that genomic sequencing and our computational analysis can provide population-level estimates of prevalence and fitness of emerging variants from wastewater samples earlier and on the basis of substantially fewer samples than from clinical samples. Our framework is being routinely used in large national projects in Switzerland and the UK.
Collapse
Affiliation(s)
- Katharina Jahn
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - David Dreifuss
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Ivan Topolsky
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Anina Kull
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | | | - Xavier Fernandez-Cassi
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carola Bänziger
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Alexander J Devaux
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Elyse Stachler
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Lea Caduff
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Federica Cariti
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alex Tuñas Corzón
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lara Fuhrmann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Chaoran Chen
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Kim Philipp Jablonski
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sarah Nadeau
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mirjam Feldkamp
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Catharine Aquino
- Functional Genomics Center Zurich, ETH Zurich, Zurich, Switzerland
| | - Tanja Stadler
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Christoph Ort
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Tamar Kohn
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Timothy R Julian
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
179
|
Antibody and T-cellular response to COVID-19 booster vaccine in SARS-CoV-1 survivors. Clin Immunol 2022; 244:109103. [PMID: 36049602 PMCID: PMC9423872 DOI: 10.1016/j.clim.2022.109103] [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: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022]
Abstract
The severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) survivors are more likely to produce a potent immune response to SARS-CoV-2 after booster vaccination. We assessed humoral and T cell responses against SARS-CoV-2 in previously vaccinated SARS-CoV-1 survivors and naïve healthy individuals (NHIs) after a booster Ad5-nCoV dose. Boosted SARS-CoV-1 survivors had a high neutralization of SARS-CoV-2 Wuhan-Hu-1 (WA1), Beta, and Delta but is limited to Omicron subvariants (BA.1, BA.2, BA.2.12.1, and BA.4/BA.5). Most boosted SARS-CoV-1 survivors had robust SARS-CoV-2-specific CD4+ and CD8+ T cell responses. While booster vaccination in NHIs elicited less or ineffective neutralization of WA1, Beta, and Delta, and none of them induced neutralizing antibodies against Omicron subvariants. However, they developed comparable SARS-CoV-2-specific T cell responses compared to boosted SARS-CoV-1 survivors. These findings suggest that boosted Ad5-nCoV would not elicit effective neutralizing antibodies against Omicron subvariants in SARS-CoV-1 survivors and NHIs but induced comparable robust T cell responses. Achieving a high antibody titer in SARS-CoV-1 survivors and NHIs is desirable to generate broad neutralization.
Collapse
|
180
|
Jahn K, Dreifuss D, Topolsky I, Kull A, Ganesanandamoorthy P, Fernandez-Cassi X, Bänziger C, Devaux AJ, Stachler E, Caduff L, Cariti F, Corzón AT, Fuhrmann L, Chen C, Jablonski KP, Nadeau S, Feldkamp M, Beisel C, Aquino C, Stadler T, Ort C, Kohn T, Julian TR, Beerenwinkel N. Early detection and surveillance of SARS-CoV-2 genomic variants in wastewater using COJAC. Nat Microbiol 2022. [PMID: 35851854 DOI: 10.1101/2021.01.08.21249379] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The continuing emergence of SARS-CoV-2 variants of concern and variants of interest emphasizes the need for early detection and epidemiological surveillance of novel variants. We used genomic sequencing of 122 wastewater samples from three locations in Switzerland to monitor the local spread of B.1.1.7 (Alpha), B.1.351 (Beta) and P.1 (Gamma) variants of SARS-CoV-2 at a population level. We devised a bioinformatics method named COJAC (Co-Occurrence adJusted Analysis and Calling) that uses read pairs carrying multiple variant-specific signature mutations as a robust indicator of low-frequency variants. Application of COJAC revealed that a local outbreak of the Alpha variant in two Swiss cities was observable in wastewater up to 13 d before being first reported in clinical samples. We further confirmed the ability of COJAC to detect emerging variants early for the Delta variant by analysing an additional 1,339 wastewater samples. While sequencing data of single wastewater samples provide limited precision for the quantification of relative prevalence of a variant, we show that replicate and close-meshed longitudinal sequencing allow for robust estimation not only of the local prevalence but also of the transmission fitness advantage of any variant. We conclude that genomic sequencing and our computational analysis can provide population-level estimates of prevalence and fitness of emerging variants from wastewater samples earlier and on the basis of substantially fewer samples than from clinical samples. Our framework is being routinely used in large national projects in Switzerland and the UK.
Collapse
Affiliation(s)
- Katharina Jahn
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - David Dreifuss
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Ivan Topolsky
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Anina Kull
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | | | - Xavier Fernandez-Cassi
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carola Bänziger
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Alexander J Devaux
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Elyse Stachler
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Lea Caduff
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Federica Cariti
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alex Tuñas Corzón
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lara Fuhrmann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Chaoran Chen
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Kim Philipp Jablonski
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sarah Nadeau
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mirjam Feldkamp
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Catharine Aquino
- Functional Genomics Center Zurich, ETH Zurich, Zurich, Switzerland
| | - Tanja Stadler
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Christoph Ort
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Tamar Kohn
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Timothy R Julian
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
181
|
Cao Y, Yisimayi A, Jian F, Song W, Xiao T, Wang L, Du S, Wang J, Li Q, Chen X, Yu Y, Wang P, Zhang Z, Liu P, An R, Hao X, Wang Y, Wang J, Feng R, Sun H, Zhao L, Zhang W, Zhao D, Zheng J, Yu L, Li C, Zhang N, Wang R, Niu X, Yang S, Song X, Chai Y, Hu Y, Shi Y, Zheng L, Li Z, Gu Q, Shao F, Huang W, Jin R, Shen Z, Wang Y, Wang X, Xiao J, Xie XS. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. Nature 2022; 608:593-602. [PMID: 35714668 DOI: 10.21203/rs.3.rs-1611421/v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/15/2022] [Indexed: 05/28/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron sublineages BA.2.12.1, BA.4 and BA.5 exhibit higher transmissibility than the BA.2 lineage1. The receptor binding and immune-evasion capability of these recently emerged variants require immediate investigation. Here, coupled with structural comparisons of the spike proteins, we show that BA.2.12.1, BA.4 and BA.5 (BA.4 and BA.5 are hereafter referred collectively to as BA.4/BA.5) exhibit similar binding affinities to BA.2 for the angiotensin-converting enzyme 2 (ACE2) receptor. Of note, BA.2.12.1 and BA.4/BA.5 display increased evasion of neutralizing antibodies compared with BA.2 against plasma from triple-vaccinated individuals or from individuals who developed a BA.1 infection after vaccination. To delineate the underlying antibody-evasion mechanism, we determined the escape mutation profiles2, epitope distribution3 and Omicron-neutralization efficiency of 1,640 neutralizing antibodies directed against the receptor-binding domain of the viral spike protein, including 614 antibodies isolated from people who had recovered from BA.1 infection. BA.1 infection after vaccination predominantly recalls humoral immune memory directed against ancestral (hereafter referred to as wild-type (WT)) SARS-CoV-2 spike protein. The resulting elicited antibodies could neutralize both WT SARS-CoV-2 and BA.1 and are enriched on epitopes on spike that do not bind ACE2. However, most of these cross-reactive neutralizing antibodies are evaded by spike mutants L452Q, L452R and F486V. BA.1 infection can also induce new clones of BA.1-specific antibodies that potently neutralize BA.1. Nevertheless, these neutralizing antibodies are largely evaded by BA.2 and BA.4/BA.5 owing to D405N and F486V mutations, and react weakly to pre-Omicron variants, exhibiting narrow neutralization breadths. The therapeutic neutralizing antibodies bebtelovimab4 and cilgavimab5 can effectively neutralize BA.2.12.1 and BA.4/BA.5, whereas the S371F, D405N and R408S mutations undermine most broadly sarbecovirus-neutralizing antibodies. Together, our results indicate that Omicron may evolve mutations to evade the humoral immunity elicited by BA.1 infection, suggesting that BA.1-derived vaccine boosters may not achieve broad-spectrum protection against new Omicron variants.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/metabolism
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antigenic Drift and Shift/genetics
- Antigenic Drift and Shift/immunology
- COVID-19/immunology
- COVID-19/transmission
- COVID-19/virology
- COVID-19 Vaccines/immunology
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Humans
- Immune Tolerance
- Immunity, Humoral
- Immunization, Secondary
- Mutation
- Neutralization Tests
- SARS-CoV-2/classification
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/metabolism
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
Collapse
Affiliation(s)
- Yunlong Cao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China.
- Changping Laboratory, Beijing, P. R. China.
| | - Ayijiang Yisimayi
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Fanchong Jian
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- College of Chemistry and Molecular Engineering, Peking University, Beijing, P. R. China
| | - Weiliang Song
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Tianhe Xiao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- Joint Graduate Program of Peking-Tsinghua-NIBS, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P. R. China
| | - Lei Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P. R. China
| | - Shuo Du
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Jing Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Qianqian Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China
| | - Xiaosu Chen
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | - Yuanling Yu
- Changping Laboratory, Beijing, P. R. China
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China
| | - Peng Wang
- Changping Laboratory, Beijing, P. R. China
| | - Zhiying Zhang
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Pulan Liu
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Ran An
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
| | - Xiaohua Hao
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Yao Wang
- Changping Laboratory, Beijing, P. R. China
| | - Jing Wang
- Changping Laboratory, Beijing, P. R. China
| | - Rui Feng
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P. R. China
| | - Haiyan Sun
- Changping Laboratory, Beijing, P. R. China
| | | | - Wen Zhang
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Dong Zhao
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | | | | | - Can Li
- Changping Laboratory, Beijing, P. R. China
| | - Na Zhang
- Changping Laboratory, Beijing, P. R. China
| | - Rui Wang
- Changping Laboratory, Beijing, P. R. China
| | - Xiao Niu
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- College of Chemistry and Molecular Engineering, Peking University, Beijing, P. R. China
| | - Sijie Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, P. R. China
| | | | - Yangyang Chai
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | - Ye Hu
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | - Yansong Shi
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | | | - Zhiqiang Li
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, P. R. China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P. R. China
| | | | - Fei Shao
- Changping Laboratory, Beijing, P. R. China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China
| | - Ronghua Jin
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Zhongyang Shen
- Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, P. R. China.
| | - Youchun Wang
- Changping Laboratory, Beijing, P. R. China.
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China.
| | - Xiangxi Wang
- Changping Laboratory, Beijing, P. R. China.
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P. R. China.
| | - Junyu Xiao
- Changping Laboratory, Beijing, P. R. China.
- School of Life Sciences, Peking University, Beijing, P. R. China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, P. R. China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P. R. China.
| | - Xiaoliang Sunney Xie
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China.
- Changping Laboratory, Beijing, P. R. China.
| |
Collapse
|
182
|
Cao Y, Yisimayi A, Jian F, Song W, Xiao T, Wang L, Du S, Wang J, Li Q, Chen X, Yu Y, Wang P, Zhang Z, Liu P, An R, Hao X, Wang Y, Wang J, Feng R, Sun H, Zhao L, Zhang W, Zhao D, Zheng J, Yu L, Li C, Zhang N, Wang R, Niu X, Yang S, Song X, Chai Y, Hu Y, Shi Y, Zheng L, Li Z, Gu Q, Shao F, Huang W, Jin R, Shen Z, Wang Y, Wang X, Xiao J, Xie XS. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. Nature 2022; 608:593-602. [PMID: 35714668 PMCID: PMC9385493 DOI: 10.1038/s41586-022-04980-y] [Citation(s) in RCA: 907] [Impact Index Per Article: 302.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron sublineages BA.2.12.1, BA.4 and BA.5 exhibit higher transmissibility than the BA.2 lineage1. The receptor binding and immune-evasion capability of these recently emerged variants require immediate investigation. Here, coupled with structural comparisons of the spike proteins, we show that BA.2.12.1, BA.4 and BA.5 (BA.4 and BA.5 are hereafter referred collectively to as BA.4/BA.5) exhibit similar binding affinities to BA.2 for the angiotensin-converting enzyme 2 (ACE2) receptor. Of note, BA.2.12.1 and BA.4/BA.5 display increased evasion of neutralizing antibodies compared with BA.2 against plasma from triple-vaccinated individuals or from individuals who developed a BA.1 infection after vaccination. To delineate the underlying antibody-evasion mechanism, we determined the escape mutation profiles2, epitope distribution3 and Omicron-neutralization efficiency of 1,640 neutralizing antibodies directed against the receptor-binding domain of the viral spike protein, including 614 antibodies isolated from people who had recovered from BA.1 infection. BA.1 infection after vaccination predominantly recalls humoral immune memory directed against ancestral (hereafter referred to as wild-type (WT)) SARS-CoV-2 spike protein. The resulting elicited antibodies could neutralize both WT SARS-CoV-2 and BA.1 and are enriched on epitopes on spike that do not bind ACE2. However, most of these cross-reactive neutralizing antibodies are evaded by spike mutants L452Q, L452R and F486V. BA.1 infection can also induce new clones of BA.1-specific antibodies that potently neutralize BA.1. Nevertheless, these neutralizing antibodies are largely evaded by BA.2 and BA.4/BA.5 owing to D405N and F486V mutations, and react weakly to pre-Omicron variants, exhibiting narrow neutralization breadths. The therapeutic neutralizing antibodies bebtelovimab4 and cilgavimab5 can effectively neutralize BA.2.12.1 and BA.4/BA.5, whereas the S371F, D405N and R408S mutations undermine most broadly sarbecovirus-neutralizing antibodies. Together, our results indicate that Omicron may evolve mutations to evade the humoral immunity elicited by BA.1 infection, suggesting that BA.1-derived vaccine boosters may not achieve broad-spectrum protection against new Omicron variants.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/metabolism
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antigenic Drift and Shift/genetics
- Antigenic Drift and Shift/immunology
- COVID-19/immunology
- COVID-19/transmission
- COVID-19/virology
- COVID-19 Vaccines/immunology
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Humans
- Immune Tolerance
- Immunity, Humoral
- Immunization, Secondary
- Mutation
- Neutralization Tests
- SARS-CoV-2/classification
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/metabolism
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
Collapse
Affiliation(s)
- Yunlong Cao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China.
- Changping Laboratory, Beijing, P. R. China.
| | - Ayijiang Yisimayi
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Fanchong Jian
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- College of Chemistry and Molecular Engineering, Peking University, Beijing, P. R. China
| | - Weiliang Song
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Tianhe Xiao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- Joint Graduate Program of Peking-Tsinghua-NIBS, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P. R. China
| | - Lei Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P. R. China
| | - Shuo Du
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Jing Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Qianqian Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China
| | - Xiaosu Chen
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | - Yuanling Yu
- Changping Laboratory, Beijing, P. R. China
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China
| | - Peng Wang
- Changping Laboratory, Beijing, P. R. China
| | - Zhiying Zhang
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Pulan Liu
- School of Life Sciences, Peking University, Beijing, P. R. China
| | - Ran An
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
| | - Xiaohua Hao
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Yao Wang
- Changping Laboratory, Beijing, P. R. China
| | - Jing Wang
- Changping Laboratory, Beijing, P. R. China
| | - Rui Feng
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P. R. China
| | - Haiyan Sun
- Changping Laboratory, Beijing, P. R. China
| | | | - Wen Zhang
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Dong Zhao
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | | | | | - Can Li
- Changping Laboratory, Beijing, P. R. China
| | - Na Zhang
- Changping Laboratory, Beijing, P. R. China
| | - Rui Wang
- Changping Laboratory, Beijing, P. R. China
| | - Xiao Niu
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- College of Chemistry and Molecular Engineering, Peking University, Beijing, P. R. China
| | - Sijie Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, P. R. China
| | | | - Yangyang Chai
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | - Ye Hu
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | - Yansong Shi
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, P. R. China
| | | | - Zhiqiang Li
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, P. R. China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P. R. China
| | | | - Fei Shao
- Changping Laboratory, Beijing, P. R. China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China
| | - Ronghua Jin
- Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Zhongyang Shen
- Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, P. R. China.
| | - Youchun Wang
- Changping Laboratory, Beijing, P. R. China.
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, P. R. China.
| | - Xiangxi Wang
- Changping Laboratory, Beijing, P. R. China.
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P. R. China.
| | - Junyu Xiao
- Changping Laboratory, Beijing, P. R. China.
- School of Life Sciences, Peking University, Beijing, P. R. China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, P. R. China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P. R. China.
| | - Xiaoliang Sunney Xie
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, P. R. China.
- Changping Laboratory, Beijing, P. R. China.
| |
Collapse
|
183
|
Yao L, Zhu KL, Jiang XL, Wang XJ, Zhan BD, Gao HX, Geng XY, Duan LJ, Dai EH, Ma MJ. Omicron subvariants escape antibodies elicited by vaccination and BA.2.2 infection. THE LANCET. INFECTIOUS DISEASES 2022; 22:1116-1117. [PMID: 35738299 PMCID: PMC9212811 DOI: 10.1016/s1473-3099(22)00410-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Lin Yao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Ka-Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Xiao-Lin Jiang
- Shandong Provincial Key Laboratory of Infectious Disease Control and Prevention, Shandong Provincial Center for Disease Control and Prevention, Jinan, China
| | - Xue-Jun Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Bing-Dong Zhan
- Department of Laboratory Medicine, Quzhou Center for Disease Control and Prevention, Quzhou, China
| | - Hui-Xia Gao
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, China
| | - Xing-Yi Geng
- Department of Infectious Disease Control and Prevention, Jinan Center for Disease Control and Prevention, Jinan, China
| | - Li-Jun Duan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Er-Hei Dai
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, China
| | - Mai-Juan Ma
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.
| |
Collapse
|
184
|
Focosi D, Maggi F. Recombination in Coronaviruses, with a Focus on SARS-CoV-2. Viruses 2022; 14:1239. [PMID: 35746710 PMCID: PMC9228924 DOI: 10.3390/v14061239] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023] Open
Abstract
Recombination is a common evolutionary tool for RNA viruses, and coronaviruses are no exception. We review here the evidence for recombination in SARS-CoV-2 and reconcile nomenclature for recombinants, discuss their origin and fitness, and speculate how recombinants could make a difference in the future of the COVID-19 pandemics.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
185
|
Saad M, Lee SJ, Tan AC, El Naqa IM, Hodi FS, Butterfield LH, LaFramboise WA, Storkus W, Karunamurthy AD, Conejo-Garcia J, Hwu P, Streicher H, Sondak VK, Kirkwood JM, Tarhini AA. Enhanced immune activation within the tumor microenvironment and circulation of female high-risk melanoma patients and improved survival with adjuvant CTLA4 blockade compared to males. J Transl Med 2022; 20:253. [PMID: 35659704 PMCID: PMC9164320 DOI: 10.1186/s12967-022-03450-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/19/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND We hypothesized that a gender difference in clinical response may exist to adjuvant CTLA4 blockade with ipilimumab versus high-dose IFNα (HDI). We investigated differences in candidate immune biomarkers in the circulation and tumor microenvironment (TME). PATIENTS AND METHODS This gender-based analysis was nested within the E1609 trial that tested adjuvant therapy with ipilimumab 3 mg/kg (ipi3) and 10 mg/kg (ipi10) versus HDI in high risk resected melanoma. We investigated gender differences in treatment efficacy with ipi3 and ipi10 versus HDI while adjusting for age, stage, ECOG performance (PS), ulceration, primary tumor status and lymph node number. Forest plots were created to compare overall survival (OS) and relapse free survival (RFS) between ipi and HDI. Gene expression profiling (GEP) was performed on tumors of 718 (454 male, 264 female) patients. Similarly, serum and peripheral blood mononuclear cells (PBMC) samples were tested for soluble and cellular biomarkers (N = 321 patients; 109 female and 212 male). RESULTS The subgroups of female, stage IIIC, PS = 1, ulcerated primary, in-transit metastasis demonstrated significant improvement in RFS and/or OS with ipi3 versus HDI. Female gender was significant for both OS and RFS and was further explored. In the RFS comparison, a multivariate Cox regression model including significant variables indicated a significant interaction between gender and treatment (P = 0.024). In peripheral blood, percentages of CD3+ T cells (P = 0.024) and CD3+ CD4+ helper T cells (P = 0.0001) were higher in females compared to males. Trends toward higher circulating levels of IL1β (P = 0.07) and IL6 (P = 0.06) were also found in females. Males had higher percentages of monocytes (P = 0.03) with trends toward higher percentages of regulatory T cells (T-reg). Tumor GEP analysis supported enhanced infiltration with immune cells including gammadelta T cells (P = 0.005), NK cells (P = 0.01), dendritic cells (P = 0.01), CD4+ T cells (P = 0.03), CD8+ T cells (P = 0.03) and T-reg (P = 0.008) in the tumors of females compared to males and a higher T-effector and IFNγ gene signature score (P = 0.0244). CONCLUSION Female gender was associated with adjuvant CTLA4 blockade clinical benefits and female patients were more likely to have evidence of type1 immune activation within the TME and the circulation. Trial registration ClinicalTrials.gov NCT01274338. Registered 11 January 2011, https://www. CLINICALTRIALS gov/ct2/show/NCT01274338.
Collapse
Affiliation(s)
- Mariam Saad
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA, 10920 McKinley Dr
| | - Sandra J Lee
- Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Aik Choon Tan
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA, Florida
| | - Issam M El Naqa
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | | | - Lisa H Butterfield
- Univ. California San Francisco and The Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Walter Storkus
- University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | | | - Jose Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Florida, Tampa, USA
| | - Patrick Hwu
- Administration, Cutaneous Oncology, Immunology, H. Lee Moffitt Cancer Center and Research Institute, Florida, Tampa, USA
| | | | - Vernon K Sondak
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Florida, Tampa, USA
| | - John M Kirkwood
- University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ahmad A Tarhini
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA, 10920 McKinley Dr..
| |
Collapse
|
186
|
Vattiato G, Maclaren O, Lustig A, Binny RN, Hendy SC, Plank MJ. An assessment of the potential impact of the Omicron variant of SARS-CoV-2 in Aotearoa New Zealand. Infect Dis Model 2022; 7:94-105. [PMID: 35434431 PMCID: PMC8993704 DOI: 10.1016/j.idm.2022.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 01/02/2023] Open
Abstract
New Zealand delayed the introduction of the Omicron variant of SARS-CoV-2 into the community by the continued use of strict border controls through to January 2022. This allowed time for vaccination rates to increase and the roll out of third doses of the vaccine (boosters) to begin. It also meant more data on the characteristics of Omicron became available prior to the first cases of community transmission. Here we present a mathematical model of an Omicron epidemic, incorporating the effects of the booster roll out and waning of vaccine-induced immunity, and based on estimates of vaccine effectiveness and disease severity from international data. The model considers differing levels of immunity against infection, severe illness and death, and ignores waning of infection-induced immunity. This model was used to provide an assessment of the potential impact of an Omicron wave in the New Zealand population, which helped inform government preparedness and response. At the time the modelling was carried out, the date of introduction of Omicron into the New Zealand community was unknown. We therefore simulated outbreaks with different start dates, as well as investigating different levels of booster uptake. We found that an outbreak starting on 1 February or 1 March led to a lower health burden than an outbreak starting on 1 January because of increased booster coverage, particularly in older age groups. We also found that outbreaks starting later in the year led to worse health outcomes than an outbreak starting on 1 March. This is because waning immunity in older groups started to outweigh the increased protection from higher booster coverage in younger groups. For an outbreak starting on 1 February and with high booster uptake, the number of occupied hospital beds in the model peaked between 800 and 3,300 depending on assumed transmission rates. We conclude that combining an accelerated booster programme with public health measures to flatten the curve are key to avoid overwhelming the healthcare system.
Collapse
Affiliation(s)
- Giorgia Vattiato
- School of Mathematics and Statistics, University of Canterbury, Christchurch, New Zealand
- Department of Physics, University of Auckland, Auckland, New Zealand
| | - Oliver Maclaren
- Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | | | | | - Shaun C. Hendy
- Department of Physics, University of Auckland, Auckland, New Zealand
| | - Michael J. Plank
- School of Mathematics and Statistics, University of Canterbury, Christchurch, New Zealand
| |
Collapse
|
187
|
Chen C, Nadeau S, Topolsky I, Beerenwinkel N, Stadler T. Advancing genomic epidemiology by addressing the bioinformatics bottleneck: Challenges, design principles, and a Swiss example. Epidemics 2022; 39:100576. [PMID: 35605437 PMCID: PMC9107180 DOI: 10.1016/j.epidem.2022.100576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 11/28/2022] Open
Abstract
The SARS-CoV-2 pandemic led to a huge increase in global pathogen genome sequencing efforts, and the resulting data are becoming increasingly important to detect variants of concern, monitor outbreaks, and quantify transmission dynamics. However, this rapid up-scaling in data generation brought with it many IT infrastructure challenges. In this paper, we report about developing an improved system for genomic epidemiology. We (i) highlight key challenges that were exacerbated by the pandemic situation, (ii) provide data infrastructure design principles to address them, and (iii) give an implementation example developed by the Swiss SARS-CoV-2 Sequencing Consortium (S3C) in response to the COVID-19 pandemic. Finally, we discuss remaining challenges to data infrastructure for genomic epidemiology. Improving these infrastructures will help better detect, monitor, and respond to future public health threats.
Collapse
Affiliation(s)
- Chaoran Chen
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, CH 4058, Switzerland; Swiss Institute of Bioinformatics, Lausanne, CH 1015, Switzerland
| | - Sarah Nadeau
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, CH 4058, Switzerland; Swiss Institute of Bioinformatics, Lausanne, CH 1015, Switzerland
| | - Ivan Topolsky
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, CH 4058, Switzerland; Swiss Institute of Bioinformatics, Lausanne, CH 1015, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, CH 4058, Switzerland; Swiss Institute of Bioinformatics, Lausanne, CH 1015, Switzerland
| | - Tanja Stadler
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, CH 4058, Switzerland; Swiss Institute of Bioinformatics, Lausanne, CH 1015, Switzerland.
| |
Collapse
|
188
|
Sokhansanj BA, Rosen GL. Mapping Data to Deep Understanding: Making the Most of the Deluge of SARS-CoV-2 Genome Sequences. mSystems 2022; 7:e0003522. [PMID: 35311562 PMCID: PMC9040592 DOI: 10.1128/msystems.00035-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2022] [Indexed: 12/22/2022] Open
Abstract
Next-generation sequencing has been essential to the global response to the COVID-19 pandemic. As of January 2022, nearly 7 million severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sequences are available to researchers in public databases. Sequence databases are an abundant resource from which to extract biologically relevant and clinically actionable information. As the pandemic has gone on, SARS-CoV-2 has rapidly evolved, involving complex genomic changes that challenge current approaches to classifying SARS-CoV-2 variants. Deep sequence learning could be a potentially powerful way to build complex sequence-to-phenotype models. Unfortunately, while they can be predictive, deep learning typically produces "black box" models that cannot directly provide biological and clinical insight. Researchers should therefore consider implementing emerging methods for visualizing and interpreting deep sequence models. Finally, researchers should address important data limitations, including (i) global sequencing disparities, (ii) insufficient sequence metadata, and (iii) screening artifacts due to poor sequence quality control.
Collapse
Affiliation(s)
- Bahrad A. Sokhansanj
- Drexel University, Ecological and Evolutionary Signal-Processing and Informatics Laboratory, Department of Electrical & Computer Engineering, College of Engineering, Philadelphia, Pennsylvania, USA
| | - Gail L. Rosen
- Drexel University, Ecological and Evolutionary Signal-Processing and Informatics Laboratory, Department of Electrical & Computer Engineering, College of Engineering, Philadelphia, Pennsylvania, USA
| |
Collapse
|
189
|
Anderegg N, Panczak R, Egger M, Low N, Riou J. Survival among people hospitalized with COVID-19 in Switzerland: a nationwide population-based analysis. BMC Med 2022; 20:164. [PMID: 35468785 PMCID: PMC9038218 DOI: 10.1186/s12916-022-02364-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Increasing age, male sex, and pre-existing comorbidities are associated with lower survival from SARS-CoV-2 infection. The interplay between different comorbidities, age, and sex is not fully understood, and it remains unclear if survival decreases linearly with higher ICU occupancy or if there is a threshold beyond which survival falls. METHOD This national population-based study included 22,648 people who tested positive for SARS-CoV-2 infection and were hospitalized in Switzerland between February 24, 2020, and March 01, 2021. Bayesian survival models were used to estimate survival after positive SARS-CoV-2 test among people hospitalized with COVID-19 by epidemic wave, age, sex, comorbidities, and ICU occupancy. Two-way interactions between age, sex, and comorbidities were included to assess the differential risk of death across strata. ICU occupancy was modeled using restricted cubic splines to allow for a non-linear association with survival. RESULTS Of 22,648 people hospitalized with COVID-19, 4785 (21.1%) died. The survival was lower during the first epidemic wave than in the second (predicted survival at 40 days after positive test 76.1 versus 80.5%). During the second epidemic wave, occupancy among all available ICU beds in Switzerland varied between 51.7 and 78.8%. The estimated survival was stable at approximately 81.5% when ICU occupancy was below 70%, but worse when ICU occupancy exceeded this threshold (survival at 80% ICU occupancy: 78.2%; 95% credible interval [CrI] 76.1 to 80.1%). Periods with higher ICU occupancy (>70 vs 70%) were associated with an estimated number of 137 (95% CrI 27 to 242) excess deaths. Comorbid conditions reduced survival more in younger people than in older people. Among comorbid conditions, hypertension and obesity were not associated with poorer survival. Hypertension appeared to decrease survival in combination with cardiovascular disease. CONCLUSIONS Survival after hospitalization with COVID-19 has improved over time, consistent with improved management of severe COVID-19. The decreased survival above 70% national ICU occupancy supports the need to introduce measures for prevention and control of SARS-CoV-2 transmission in the population well before ICUs are full.
Collapse
Affiliation(s)
- Nanina Anderegg
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland. .,Federal Office of Public Health, Bern, Switzerland.
| | - Radoslaw Panczak
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Matthias Egger
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Centre for Infectious Disease Epidemiology and Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Julien Riou
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Federal Office of Public Health, Bern, Switzerland
| |
Collapse
|
190
|
Williams AH, Zhan CG. Generalized Methodology for the Quick Prediction of Variant SARS-CoV-2 Spike Protein Binding Affinities with Human Angiotensin-Converting Enzyme II. J Phys Chem B 2022; 126:2353-2360. [PMID: 35315274 PMCID: PMC8982491 DOI: 10.1021/acs.jpcb.1c10718] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/24/2022] [Indexed: 12/25/2022]
Abstract
Variants of the SARS-CoV-2 virus continue to remain a threat 2 years from the beginning of the pandemic. As more variants arise, and the B.1.1.529 (Omicron) variant threatens to create another wave of infections, a method is needed to predict the binding affinity of the spike protein quickly and accurately with human angiotensin-converting enzyme II (ACE2). We present an accurate and convenient energy minimization/molecular mechanics Poisson-Boltzmann surface area methodology previously used with engineered ACE2 therapeutics to predict the binding affinity of the Omicron variant. Without any additional data from the variants discovered after the publication of our first model, the methodology can accurately predict the binding of the spike/ACE2 variant complexes. From this methodology, we predicted that the Omicron variant spike has a Kd of ∼22.69 nM (which is very close to the experimental Kd of 20.63 nM published during the review process of the current report) and that spike protein of the new "Stealth" Omicron variant (BA.2) will display a Kd of ∼12.9 nM with the wild-type ACE2 protein. This methodology can be used with as-yet discovered variants, allowing for quick determinations regarding the variant's infectivity versus either the wild-type virus or its variants.
Collapse
Affiliation(s)
- Alexander H. Williams
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| |
Collapse
|
191
|
Jiao S, Chen Z, Zhang L, Zhou X, Shi L. ATGPred-FL: sequence-based prediction of autophagy proteins with feature representation learning. Amino Acids 2022; 54:799-809. [PMID: 35286461 DOI: 10.1007/s00726-022-03145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/28/2022] [Indexed: 11/26/2022]
Abstract
Autophagy plays an important role in biological evolution and is regulated by many autophagy proteins. Accurate identification of autophagy proteins is crucially important to reveal their biological functions. Due to the expense and labor cost of experimental methods, it is urgent to develop automated, accurate and reliable sequence-based computational tools to enable the identification of novel autophagy proteins among numerous proteins and peptides. For this purpose, a new predictor named ATGPred-FL was proposed for the efficient identification of autophagy proteins. We investigated various sequence-based feature descriptors and adopted the feature learning method to generate corresponding, more informative probability features. Then, a two-step feature selection strategy based on accuracy was utilized to remove irrelevant and redundant features, leading to the most discriminative 14-dimensional feature set. The final predictor was built using a support vector machine classifier, which performed favorably on both the training and testing sets with accuracy values of 94.40% and 90.50%, respectively. ATGPred-FL is the first ATG machine learning predictor based on protein primary sequences. We envision that ATGPred-FL will be an effective and useful tool for autophagy protein identification, and it is available for free at http://lab.malab.cn/~acy/ATGPred-FL , the source code and datasets are accessible at https://github.com/jiaoshihu/ATGPred .
Collapse
Affiliation(s)
- Shihu Jiao
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Zheng Chen
- School of Applied Chemistry and Biological Technology, Shenzhen Polytechnic, 7098 Liuxian Street, Shenzhen, 518055, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, No.4 Block 2 North Jianshe Road, Chengdu, 61005, China
| | - Lichao Zhang
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, Shenzhen, 518172, China
| | - Xun Zhou
- Beidahuang Industry Group General Hospital, Harbin, 150001, China.
| | - Lei Shi
- Department of Spine Surgery, Changzheng Hospital, Naval Medical University, No 415, Fengyang Road, Huangpu District, Shanghai, 210000, China.
| |
Collapse
|
192
|
Caduff L, Dreifuss D, Schindler T, Devaux AJ, Ganesanandamoorthy P, Kull A, Stachler E, Fernandez-Cassi X, Beerenwinkel N, Kohn T, Ort C, Julian TR. Inferring transmission fitness advantage of SARS-CoV-2 variants of concern from wastewater samples using digital PCR, Switzerland, December 2020 through March 2021. Euro Surveill 2022; 27:2100806. [PMID: 35272748 PMCID: PMC8915404 DOI: 10.2807/1560-7917.es.2022.27.10.2100806] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/21/2022] [Indexed: 04/19/2023] Open
Abstract
BackgroundThroughout the COVID-19 pandemic, SARS-CoV-2 genetic variants of concern (VOCs) have repeatedly and independently arisen. VOCs are characterised by increased transmissibility, increased virulence or reduced neutralisation by antibodies obtained from prior infection or vaccination. Tracking the introduction and transmission of VOCs relies on sequencing, typically whole genome sequencing of clinical samples. Wastewater surveillance is increasingly used to track the introduction and spread of SARS-CoV-2 variants through sequencing approaches.AimHere, we adapt and apply a rapid, high-throughput method for detection and quantification of the relative frequency of two deletions characteristic of the Alpha, Beta, and Gamma VOCs in wastewater.MethodsWe developed drop-off RT-dPCR assays and an associated statistical approach implemented in the R package WWdPCR to analyse temporal dynamics of SARS-CoV-2 signature mutations (spike Δ69-70 and ORF1a Δ3675-3677) in wastewater and quantify transmission fitness advantage of the Alpha VOC.ResultsBased on analysis of Zurich wastewater samples, the estimated transmission fitness advantage of SARS-CoV-2 Alpha based on the spike Δ69-70 was 0.34 (95% confidence interval (CI): 0.30-0.39) and based on ORF1a Δ3675-3677 was 0.53 (95% CI: 0.49-0.57), aligning with the transmission fitness advantage of Alpha estimated by clinical sample sequencing in the surrounding canton of 0.49 (95% CI: 0.38-0.61).ConclusionDigital PCR assays targeting signature mutations in wastewater offer near real-time monitoring of SARS-CoV-2 VOCs and potentially earlier detection and inference on transmission fitness advantage than clinical sequencing.
Collapse
Affiliation(s)
- Lea Caduff
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - David Dreifuss
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Tobias Schindler
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Alexander J Devaux
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | | | - Anina Kull
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Elyse Stachler
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Xavier Fernandez-Cassi
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Tamar Kohn
- Laboratory of Environmental Chemistry, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Christoph Ort
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Timothy R Julian
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
193
|
Chen J, Zhang Y, Shen B. Bioinformatics for the Origin and Evolution of Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1368:53-71. [DOI: 10.1007/978-981-16-8969-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
194
|
Sant’Anna FH, Muterle Varela AP, Prichula J, Comerlato J, Comerlato CB, Roglio VS, Mendes Pereira GF, Moreno F, Seixas A, Wendland EM. Emergence of the novel SARS-CoV-2 lineage VUI-NP13L and massive spread of P.2 in South Brazil. Emerg Microbes Infect 2021; 10:1431-1440. [PMID: 34184973 PMCID: PMC8284128 DOI: 10.1080/22221751.2021.1949948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 12/16/2022]
Abstract
In this study, we analyzed 340 whole genomes of SARS-CoV-2, which were sampled between April and November 2020 in 33 cities of Rio Grande do Sul, South Brazil. We demonstrated the circulation of two novel emergent lineages, VUI-NP13L and VUI-NP13L-like, and five major lineages that had already been assigned (B.1.1.33, B.1.1.28, P.2, B.1.91, B.1.195). P.2 and VUI-NP13L demonstrated a massive spread in October 2020. Constant and consistent genomic surveillance is crucial to identify newly emerging SARS-CoV-2 lineages in Brazil and to guide decision making in the Brazilian Public Healthcare System.
Collapse
Affiliation(s)
| | - Ana Paula Muterle Varela
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Janira Prichula
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | | | | | | | | | - Flávia Moreno
- Department of Chronic Conditions and Sexually Transmitted Infections, Ministry of Health, Brasília, Brazil
| | - Adriana Seixas
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Eliana Márcia Wendland
- Hospital Moinhos de Vento, PROADI – SUS, Porto Alegre, Brazil
- Department of Community Health, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| |
Collapse
|