301
|
Brisotto G, Muraro E, Montico M, Corso C, Evangelista C, Casarotto M, Caffau C, Vettori R, Cozzi MR, Zanussi S, Turetta M, Ronchese F, Steffan A. IgG antibodies against SARS-CoV-2 decay but persist 4 months after vaccination in a cohort of healthcare workers. Clin Chim Acta 2021; 523:476-482. [PMID: 34755649 PMCID: PMC8555109 DOI: 10.1016/j.cca.2021.10.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Monitoring the immune response against SARS-CoV-2 is pivotal in the evaluation of long-term vaccine efficacy. Immunoglobulin G (IgG) antibodies represent an advisable tool to reach this goal, especially for the still poorly defined antibody trend induced by the new class of mRNA vaccines against SARS-CoV-2. MATERIALS AND METHODS Anti-Spike RBD IgG antibodies were monitored in a cohort of healthcare workers at CRO Aviano, National Cancer Institute, through MAGLUMI® chemiluminescence assay, at 1 and 4 months after full-schedule of BNT162b2 or mRNA-1273 vaccination. RESULTS At 1 month after vaccination, 99.9% of 767 healthcare workers showed a reactive antibody response, which was inversely correlated with age, and positively associated with a previous history of COVID-19, and mRNA-1273 vaccination. Serological response was maintained in 99.6% of the 516 subjects monitored also at follow-up. An antibody decay from 559.8 AU/mL (IQR 359.7-845.7) to 92.7 AU/mL (IQR 65.1-148.6; p < 0.001) was observed, independently from age and sex. CONCLUSION Our data supported the ability of SARS-CoV-2 mRNA vaccines to induce at least a 4 months-lasting IgG response, even outside the rules of clinical trials. The antibody decay observed at follow-up suggested to deepen the immune response characterization to identify subjects with low anti-SARS-CoV-2 immunity possibly requiring a vaccination boost.
Collapse
Affiliation(s)
- Giulia Brisotto
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Elena Muraro
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy.
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Chiara Corso
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Chiara Evangelista
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Mariateresa Casarotto
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Cristina Caffau
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Roberto Vettori
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Maria Rita Cozzi
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Stefania Zanussi
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Matteo Turetta
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| | - Federico Ronchese
- Clinical Unit of Occupational Medicine, Department of Medical Sciences, University of Trieste, 34100 Trieste, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, PN, Italy
| |
Collapse
|
302
|
Qiu H, Yuan XY, Cabral T, Manguiat K, Robinson A, Wood H, Grant C, McQueen P, Westmacott G, Beniac DR, Lin L, Carpenter M, Kobasa D, Gräfenhan T. Development and characterization of SARS-CoV-2 variant-neutralizing monoclonal antibodies. Antiviral Res 2021; 196:105206. [PMID: 34762975 PMCID: PMC8572761 DOI: 10.1016/j.antiviral.2021.105206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 11/27/2022]
Abstract
Vaccination and administration of monoclonal antibody cocktails are effective tools to control the progression of infectious diseases and to terminate pandemics such as COVID-19. However, the emergence of SARS-CoV-2 mutants with enhanced transmissibility and altered antigenicity requires broad-spectrum therapies. Here we developed a panel of SARS-CoV-2 specific mouse monoclonal antibodies (mAbs), and characterized them based on ELISA, Western immunoblot, isotyping, and virus neutralization. Six neutralizing mAbs that exhibited high-affinity binding to SARS-CoV-2 spike protein were identified, and their amino acid sequences were determined by mass spectrometry. Functional assays confirmed that three mAbs, F461G11, F461G15, and F461G16 neutralized four variants of concern (VOC): B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma) and B.1.617.2 (delta) These mAbs are promising candidates for COVID-19 therapy, and understanding their interactions with virus spike protein should support further vaccine and antibody development.
Collapse
Affiliation(s)
- Hongyu Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada.
| | - Xin-Yong Yuan
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Teresa Cabral
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Kathy Manguiat
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Alyssia Robinson
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Heidi Wood
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Chris Grant
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Peter McQueen
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Garrett Westmacott
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Daniel R Beniac
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Lisa Lin
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Michael Carpenter
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Darwyn Kobasa
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Tom Gräfenhan
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada.
| |
Collapse
|
303
|
Tian D, Sun Y, Zhou J, Ye Q. The Global Epidemic of the SARS-CoV-2 Delta Variant, Key Spike Mutations and Immune Escape. Front Immunol 2021; 12:751778. [PMID: 34917076 PMCID: PMC8669155 DOI: 10.3389/fimmu.2021.751778] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022] Open
Abstract
During the COVID-19 pandemic, SARS-CoV-2 variants have emerged and spread worldwide. The Delta (B.1.617.2) variant was first reported in India in October 2020 and was classified as a "variant of concern (VOC)" by the WHO on 11 May, 2021. Compared to the wild-type strain, several studies have shown that the Delta variant is more transmissible and has higher viral loads in infected samples. COVID-19 patients infected with the Delta variant have a higher risk of hospitalization, intensive care unit (ICU) admission, and mortality. The Delta variant is becoming the dominant strain in many countries around the world. This review summarizes and analyses the biological characteristics of key amino acid mutations, the epidemic characteristics, and the immune escape of the Delta variant. We hope to provide scientific reference for the monitoring and prevention measures of the SARS-CoV-2 Delta variant and the development strategy of a second-generation vaccine.
Collapse
Affiliation(s)
| | | | | | - Qing Ye
- National Clinical Research Center for Child Health, National Children’s Regional Medical Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
304
|
Evolution of the SARS-CoV-2 genome and emergence of variants of concern. Arch Virol 2021; 167:293-305. [PMID: 34846601 PMCID: PMC8629736 DOI: 10.1007/s00705-021-05295-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/22/2021] [Indexed: 01/08/2023]
Abstract
The high transmission and mortality rates associated with SARS-CoV-2 have led to tragic consequences worldwide. Large-scale whole-genome sequencing of the SARS-CoV-2 genome since its identification in late 2019 has identified many sequence changes and the emergence of novel strains, each described by co-segregation of a particular set of sequence variations. Variants designated G, alpha (B.1.1.7), beta (B.1.351), gamma (P.1), and delta (B.1.617.2) are important lineages that emerged sequentially and are considered variants of concern. A notable feature of the last four, each of which ultimately evolved from clade G, is the large number (≥ 20) of co-segregating sequence variations associated with them. Several variations are in the spike gene, and some variations are shared among or between strains. Meanwhile, observation of recurrent infections with the same or different SARS-CoV-2 lineages has raised concerns about the duration of the immune responses induced by the initial infection or the vaccine that was administered. While the alpha strain is sensitive to immune responses induced by earlier strains, the beta, gamma, and delta strains can escape antibody neutralization. Apart from random replication errors, intra-host RNA editing, chronic infections, and recombination are processes that may promote the accumulation of sequence changes in the SARS-CoV-2 genome. The known contribution of recombination to coronavirus evolution and recent data pertaining to SARS-CoV-2 suggest that recombination may be particularly important. Continued surveillance of the SARS-CoV-2 genome is imperative.
Collapse
|
305
|
Immune response against SARS-CoV-2 variants: the role of neutralization assays. NPJ Vaccines 2021; 6:142. [PMID: 34845231 PMCID: PMC8630184 DOI: 10.1038/s41541-021-00404-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 11/03/2021] [Indexed: 12/17/2022] Open
Abstract
Since the emergence of the novel coronavirus SARS-CoV-2 in late 2019, the COVID-19 pandemic has hindered social life and global economic activity. As of July 2021, SARS-CoV-2 has caused over four million deaths. The rapid spread and high mortality of the disease demanded the international scientific community to develop effective vaccines in a matter of months. However, unease about vaccine efficacy has arisen with the spread of the SARS-CoV-2 variants of concern (VOCs). Time- and cost-efficient in vitro neutralization assays are widely used to measure neutralizing antibody responses against VOCs. However, the extent to which in vitro neutralization reflects protection from infection remains unclear. Here, we describe common neutralization assays based on infectious and pseudotyped viruses and evaluate their role in testing neutralizing responses against new SARS-CoV-2 variants. Additionally, we briefly review the recent findings on the immune response elicited by available vaccines against major SARS-CoV-2 variants, including Alpha, Beta, Gamma, and Delta.
Collapse
|
306
|
Khandker SS, Godman B, Jawad MI, Meghla BA, Tisha TA, Khondoker MU, Haq MA, Charan J, Talukder AA, Azmuda N, Sharmin S, Jamiruddin MR, Haque M, Adnan N. A Systematic Review on COVID-19 Vaccine Strategies, Their Effectiveness, and Issues. Vaccines (Basel) 2021; 9:1387. [PMID: 34960133 PMCID: PMC8708628 DOI: 10.3390/vaccines9121387] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
COVID-19 vaccines are indispensable, with the number of cases and mortality still rising, and currently no medicines are routinely available for reducing morbidity and mortality, apart from dexamethasone, although others are being trialed and launched. To date, only a limited number of vaccines have been given emergency use authorization by the US Food and Drug Administration and the European Medicines Agency. There is a need to systematically review the existing vaccine candidates and investigate their safety, efficacy, immunogenicity, unwanted events, and limitations. The review was undertaken by searching online databases, i.e., Google Scholar, PubMed, and ScienceDirect, with finally 59 studies selected. Our findings showed several types of vaccine candidates with different strategies against SARS-CoV-2, including inactivated, mRNA-based, recombinant, and nanoparticle-based vaccines, are being developed and launched. We have compared these vaccines in terms of their efficacy, side effects, and seroconversion based on data reported in the literature. We found mRNA vaccines appeared to have better efficacy, and inactivated ones had fewer side effects and similar seroconversion in all types of vaccines. Overall, global variant surveillance and systematic tweaking of vaccines, coupled with the evaluation and administering vaccines with the same or different technology in successive doses along with homologous and heterologous prime-booster strategy, have become essential to impede the pandemic. Their effectiveness appreciably outweighs any concerns with any adverse events.
Collapse
Affiliation(s)
- Shahad Saif Khandker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
| | - Brian Godman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G1 1XQ, UK;
- Division of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Md. Irfan Jawad
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Bushra Ayat Meghla
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Taslima Akter Tisha
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Mohib Ullah Khondoker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Community Medicine, Gonoshasthaya Samaj Vittik Medical College, Savar 1344, Bangladesh
| | - Md. Ahsanul Haq
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
| | - Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur 342005, India;
| | - Ali Azam Talukder
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Nafisa Azmuda
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Shahana Sharmin
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Mohd. Raeed Jamiruddin
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sugai Besi, Kuala Lumpur 57000, Malaysia
| | - Nihad Adnan
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| |
Collapse
|
307
|
Maeda K, Amano M, Uemura Y, Tsuchiya K, Matsushima T, Noda K, Shimizu Y, Fujiwara A, Takamatsu Y, Ichikawa Y, Nishimura H, Kinoshita M, Matsumoto S, Gatanaga H, Yoshimura K, Oka SI, Mikami A, Sugiura W, Sato T, Yoshida T, Shimada S, Mitsuya H. Correlates of neutralizing/SARS-CoV-2-S1-binding antibody response with adverse effects and immune kinetics in BNT162b2-vaccinated individuals. Sci Rep 2021; 11:22848. [PMID: 34819514 PMCID: PMC8613264 DOI: 10.1038/s41598-021-01930-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023] Open
Abstract
While mRNA vaccines against SARS-CoV-2 are exceedingly effective in preventing symptomatic infection, their immune response features remain to be clarified. In the present prospective study, 225 healthy individuals in Japan, who received two BNT162b2 doses, were enrolled. Correlates of BNT162b2-elicited SARS-CoV-2-neutralizing activity (50% neutralization titer: NT50; assessed using infectious virions) with various determinants were examined and the potency of sera against variants of concerns was determined. Significant rise in NT50s was seen in sera on day 28 post-1st dose. A moderate inverse correlation was seen between NT50s and ages, but no correlation seen between NT50s and adverse effects. NT50s and SARS-CoV-2-S1-binding-IgG levels on day 28 post-1st dose and pain scores following the 2nd dose were greater in women than in men. The average half-life of NT50s was ~ 68 days, and 23.6% (49 out of 208 individuals) failed to show detectable neutralizing activity on day 150. While sera from elite-responders (NT50s > 1,500: the top 4% among the participants) potently to moderately blocked all variants of concerns examined, some sera with low NT50s failed to block the B.1.351-beta strain. Since BNT162b2-elicited immunity against SARS-CoV-2 is short, an additional vaccine or other protective measures are needed.
Collapse
Affiliation(s)
- Kenji Maeda
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan.
| | - Masayuki Amano
- Department of Clinical Sciences, Kumamoto University Hospital, Kumamoto, Japan
| | | | | | | | | | | | - Asuka Fujiwara
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | - Yuki Takamatsu
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | - Hiroaki Mitsuya
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan.
- Department of Clinical Sciences, Kumamoto University Hospital, Kumamoto, Japan.
- Experimental Retrovirology Section, HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
308
|
Herd immunity to SARS-COV-2 in the population of the Southern regions of the Far East of Russia. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2021-6.5.25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The aim: to study the structure and dynamics of population immunity to SARSCoV-2 of the population of the Southern Regions of the Far East (SRFE): Khabarovsk, Primorsky Krai and Amur Region during the COVID-19 epidemic in 2020.Materials and methods. The work was carried out according to the program for assessing population immunity to SARS -CoV-2 of the population of the Russian Federation according to the methodology developed by the Rospotrebnadzor with the participation of the St. Petersburg Pasteur Institute. The study was approved by the ethical committee of the St. Petersburg Pasteur Institute. The selection of participants was carried out by a questionnaire method using cloud technologies. The volunteers were randomized by age by stratification into 7 age groups: 1–17, 18–29, 30–39, 40–49, 50–59, 60–69, 70+ years old. Territorial randomization consisted in limiting the engaging of volunteers – no more than 30 people from one enterprise. After the initial cross-sectional study, a 3-stage seromonitoring was carried out, in which the same volunteers participated. Antibodies to the SARS-CoV-2 nucleocapsid were determined in peripheral blood serum by the enzyme immunoassay using an appropriate set of reagents produced by the State Scientific Center for Medical and Biological Sciences of the Rospotrebnadzor (Obolensk). Statistical analysis was performed using the Excel package. The confidence interval for the proportion was calculated using the A. Wald, J. Wolfowitz method with A. Agresti, B.A. Coull’s correction. The statistical significance of the differences was calculated online using a specialized calculator. The statistical significance of the differences was assessed with a probability of p˂ 0.05, unless otherwise indicated.Results. In a comparative analysis, the highest morbidity was observed in the Khabarovsk Territory, the lowest – in the Primorsky Territory. The level of seroprevalence among the population of the region was 19.6 % (95 % CI: 18.2–21.1) in the Khabarovsk Territory, 19.6 % (95 % CI: 18.1–21.2) in the Primorsky Territory19,6 % and 45,5 % (95 % CI: 43.7–47.3) in the Amur region. The highest seroprevalence was noted among 1–17 years old children, mainly due to the subgroup of 14–17-years-olds. The smallest proportion of seropositive was found among 40–49-year-olds in the Khabarovsk Territory (14.7 %, 95 % CI: 11.2–18.6), 18–28-yearolds in the Primorsky Territory (13.3 %, 95 % CI: 10.0–17.1) and 30–39-year-olds in the Amur Region (36.3 %, 95% CI: 31.7–41.6). No statistically significant dependence of seroprevalence on territorial and occupational factors has been established, with the exception of an increase in the proportion of seropositive medical workers in Primorsky Territory. In the process of 3-stage seromonitoring, a regular increase in the proportion of seropositive people was revealed in all SRFE. The resulting tendency is correctly described by a second-order polynomial. A relationship was revealed between the number of convalescents and persons in contact with them, which made it possible to calculate the base reproductive number (R0) in the range from 1.4 (Primorsky Territory) to 2.4 (Amur Region). Analysis of seroprevalent volunteers showed that the number of asymptomatic individuals varied from 94.1 % (95 % CI: 92.8–95.3) to 98.3 % (95 % CI: 98.8–99.2). This indicates that most of the volunteers had COVID-19 asymptomatically.Conclusions. A comparative study showed the prevalence of seroprevalence in the Amur Region compared with the Khabarovsk and Primorsky Territories. The relationship between the number of convalescents and persons in contact with them was noted. The value of the base R0 is calculated. It has been shown that more than 90 % of seropositive individuals in the COVID-10 SRFE were asymptomatic.
Collapse
|
309
|
Intapiboon P, Seepathomnarong P, Ongarj J, Surasombatpattana S, Uppanisakorn S, Mahasirimongkol S, Sawaengdee W, Phumiamorn S, Sapsutthipas S, Sangsupawanich P, Chusri S, Pinpathomrat N. Immunogenicity and Safety of an Intradermal BNT162b2 mRNA Vaccine Booster after Two Doses of Inactivated SARS-CoV-2 Vaccine in Healthy Population. Vaccines (Basel) 2021; 9:1375. [PMID: 34960122 PMCID: PMC8703694 DOI: 10.3390/vaccines9121375] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 01/01/2023] Open
Abstract
Effective vaccine coverage is urgently needed to tackle the COVID-19 pandemic. Inactivated vaccines have been introduced in many countries for emergency usage, but have only provided limited protection. Heterologous vaccination is a promising strategy to maximise vaccine immunogenicity. Here, we conducted a phase I, randomised control trial to observe the safety and immunogenicity after an intradermal boost, using a fractional dosage (1:5) of BNT162b2 mRNA vaccine in healthy participants in Songkhla, Thailand. In total, 91 volunteers who had been administered with two doses of inactivated SARS-CoV-2 (CoronaVac) were recruited into the study, and then randomised (1:1:1) to received different regimens of the third dose. An intramuscular booster with a full dose of BNT162b2 was included as a conventional control, and a half dose group was included as reciprocal comparator. Both, immediate and delayed adverse events following immunisation (AEFI) were monitored. Humoral and cellular immune responses were examined to observe the booster effects. The intradermal booster provided significantly fewer systemic side effects, from 70% down to 19.4% (p < 0.001); however, they were comparable to local reactions with the conventional intramuscular booster. In the intradermal group after receiving only one fifth of the conventional dosage, serum Anti-RBD IgG was halved compared to the full dose of an intramuscular injection. However, the neutralising function against the Delta strain remained intact. T cell responses were also less effective in the intradermal group compared to the intramuscular booster. Together, the intradermal booster, using a fractional dose of BNT162b2, can reduce systemic reactions and provides a good level and function of antibody responses compared to the conventional booster. This favourable intradermal boosting strategy provides a suitable alternative for vaccines and effective vaccine management to increase the coverage during the vaccine shortage.
Collapse
Affiliation(s)
- Porntip Intapiboon
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.I.); (S.C.)
| | - Purilap Seepathomnarong
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.S.); (J.O.)
| | - Jomkwan Ongarj
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.S.); (J.O.)
| | | | - Supattra Uppanisakorn
- Clinical Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (S.U.); (P.S.)
| | | | - Waritta Sawaengdee
- Department of Medical Science, Ministry of Public Health, Nonthaburi 11000, Thailand; (S.M.); (W.S.)
| | - Supaporn Phumiamorn
- Institute of Biological Products, Department of Medical Sciences, Ministry of Public Health, Nonthaburi 11000, Thailand; (S.P.); (S.S.)
| | - Sompong Sapsutthipas
- Institute of Biological Products, Department of Medical Sciences, Ministry of Public Health, Nonthaburi 11000, Thailand; (S.P.); (S.S.)
| | - Pasuree Sangsupawanich
- Clinical Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (S.U.); (P.S.)
| | - Sarunyou Chusri
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.I.); (S.C.)
| | - Nawamin Pinpathomrat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.S.); (J.O.)
| |
Collapse
|
310
|
Yang W, Shaman J. COVID-19 pandemic dynamics in India, the SARS-CoV-2 Delta variant, and implications for vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.06.21.21259268. [PMID: 34845460 PMCID: PMC8629204 DOI: 10.1101/2021.06.21.21259268] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
BACKGROUND The COVID-19 Delta pandemic wave in India surged and declined within 3 months; cases then remained low despite the continued spread of Delta elsewhere. Here we aim to estimate key epidemiological characteristics of the Delta variant based on data from India and examine the underpinnings of its dynamics. METHODS We utilize multiple datasets and model-inference methods to reconstruct COVID-19 pandemic dynamics in India during March 2020 - June 2021. We further use model estimates to retrospectively predict cases and deaths during July - mid-Oct 2021, under various vaccination and vaccine effectiveness (VE) settings to estimate the impact of vaccination and VE for non-Delta-infection recoverees. FINDINGS We estimate that Delta escaped immunity in 34.6% (95% CI: 0 - 64.2%) of individuals with prior wildtype infection and was 57.0% (95% CI: 37.9 - 75.6%) more infectious than wildtype SARS-CoV-2. Models assuming higher VE among those with prior non-Delta infection, particularly after the 1 st dose, generated more accurate predictions than those assuming no such increases (best-performing VE setting: 90/95% vs. 30/67% baseline for the 1 st /2 nd dose). Counterfactual modeling indicates that high vaccination coverage for 1 st vaccine-dose in India (∼50% by mid-Oct 2021) combined with the boosting of VE among recoverees averted around 60% of infections during July - mid-Oct 2021. INTERPRETATION Non-pharmaceutical interventions, infection seasonality, and high coverage of 1-dose vaccination likely all contributed to pandemic dynamics in India during 2021. Given the shortage of COVID-19 vaccines globally and boosting of VE, for populations with high prior infection rates, prioritizing the first vaccine-dose may protect more people. RESEARCH IN CONTEXT Evidence before this study: We searched PubMed for studies published through Nov 3, 2021 on the Delta (B.1.617.2) SARS-CoV-2 variant that focused on three areas: 1) transmissibility [search terms: ("Delta variant" OR "B.1.617") AND ("transmission rate" OR "growth rate" OR "secondary attack rate" OR "transmissibility")]; 2) immune response ([search terms: ("Delta variant" OR "B.1.617") AND ("immune evas" OR "immune escape")]; and 3) vaccine effectiveness ([search terms: ("Delta variant" OR "B.1.617") AND ("vaccine effectiveness" OR "vaccine efficacy" OR "vaccination")]. Our search returned 256 papers, from which we read the abstracts and identified 54 relevant studies.Forty-two studies addressed immune evasion and/or vaccine effectiveness. Around half (n=19) of these studies measured the neutralizing ability of convalescent sera and/or vaccine sera against Delta and most reported some reduction (around 2-to 8-fold) compared to ancestral variants. The remainder (n=23) used field observations (often with a test-negative or cohort-design) and reported lower VE against infection but similar VE against hospitalization or death. Together, these laboratory and field observations consistently indicate that Delta can evade preexisting immunity. In addition, five studies reported higher B-cell and/or T-cell vaccine-induced immune response among recovered vaccinees than naïve vaccinees, suggesting potential boosting of pre-existing immunity; however, all studies were based on small samples (n = 10 to 198 individuals).Sixteen studies examined transmissibility, including 1) laboratory experiments (n=6) showing that Delta has higher affinity to the cell receptor, fuses membranes more efficiently, and/or replicates faster than other SARS-CoV-2 variants, providing biological mechanisms for its higher transmissibility; 2) field studies (n=5) showing higher rates of breakthrough infections by Delta and/or higher viral load among Delta infections than other variants; and 3) modeling/mixed studies (n=5) using genomic or case data to estimate the growth rate or reproduction number, reporting a 60-120% increase. Only one study jointly estimated the increase in transmissibility (1.3-1.7-fold, 50% CI) and immune evasion (10-50%, 50% CI); this study also reported a 27.5% (25/91) reinfection rate by Delta.Added value of this study: We utilize observed pandemic dynamics and the differential vaccination coverage for two vaccine doses in India, where the Delta variant was first identified, to estimate the epidemiological properties of Delta and examine the impact of prior non-Delta infection on immune boosting at the population level. We estimate that Delta variant can escape immunity from prior wildtype infection roughly one-third of the time and is around 60% more infectious than wildtype SARS-CoV-2. In addition, our analysis suggests the large increase in population receiving their first vaccine dose (∼50% by end of Oct 2021) combined with the boosting effect of vaccination for non-Delta infection recoverees likely mitigated epidemic intensity in India during July - Oct 2021.Implications of all the available evidence: Our analysis reconstructs the interplay and effects of non-pharmaceutical interventions, infection seasonality, Delta variant emergence, and vaccination on COVID-19 pandemic dynamics in India. Modeling findings support prioritizing the first vaccine dose in populations with high prior infection rates, given vaccine shortages.
Collapse
|
311
|
Tada T, Zhou H, Dcosta BM, Samanovic MI, Mulligan MJ, Landau NR. Partial resistance of SARS-CoV-2 Delta variants to vaccine-elicited antibodies and convalescent sera. iScience 2021; 24:103341. [PMID: 34723159 PMCID: PMC8541826 DOI: 10.1016/j.isci.2021.103341] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/13/2021] [Accepted: 10/20/2021] [Indexed: 12/03/2022] Open
Abstract
Highly transmissible SARS-CoV-2 variants identified in India and designated B.1.617, Kappa (B.1.617.1), Delta (B.1.617.2), B.1.618, and B.1.36.29 contain spike mutations L452R, T478K, E484K, E484Q, and N440K located within the spike receptor-binding domain and thus could contribute to increased transmissibility and potentially allow re-infection or cause resistance to vaccine-elicited antibody. To address these issues, we used lentiviruses pseudotyped by variant spikes to measure their neutralization by convalescent sera, vaccine-elicited and Regeneron therapeutic antibodies, and ACE2 affinity. Convalescent sera and vaccine-elicited antibodies neutralized viruses with Delta spike with 2- to 5-fold decrease in titer in different donors. Regeneron antibody cocktail neutralized virus with the Delta spike with a 2.6-fold decrease in titer. Neutralization resistance to serum antibodies and monoclonal antibodies was mediated by L452R mutation. These relatively modest decreases in antibody neutralization titer for viruses with variant spike proteins suggest that current vaccines will remain protective against the family of Delta variants. Vaccine-elicited antibodies neutralize Delta spike with 4- to 5-fold decrease in titer Delta variant is resistant to REGN10933 monoclonal antibody Neutralization resistance is mediated by L452R
Collapse
Affiliation(s)
- Takuya Tada
- Department of Microbiology, NYU Grossman School of Medicine, 430 East 29th Street, Alexandria West Building, New York, NY 10016, USA
| | - Hao Zhou
- Department of Microbiology, NYU Grossman School of Medicine, 430 East 29th Street, Alexandria West Building, New York, NY 10016, USA
| | - Belinda M Dcosta
- Department of Microbiology, NYU Grossman School of Medicine, 430 East 29th Street, Alexandria West Building, New York, NY 10016, USA
| | - Marie I Samanovic
- NYU Langone Vaccine Center and Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Mark J Mulligan
- NYU Langone Vaccine Center and Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Nathaniel R Landau
- Department of Microbiology, NYU Grossman School of Medicine, 430 East 29th Street, Alexandria West Building, New York, NY 10016, USA
| |
Collapse
|
312
|
Abstract
Since the start of the pandemic, SARS-CoV-2 has infected almost 200 million human hosts and is set to encounter and gain entry in many more in the coming months. As the coronavirus flourish, the evolutionary pressure selects those variants that can complete the infection cycle faster and reproduce in large numbers compared to others. This increase in infectivity and transmissibility coupled with the immune response from high viral load may cause moderate to severe disease. Whether this leads to enhanced virulence in the prevalent Alpha and Delta variants is still not clear. This review describes the different types of SARS-CoV-2 variants that are now prevalent, their emergence, the mutations responsible for their growth advantages, and how they affect vaccine efficacy and increase chances of reinfection. Finally, we have also summarized the efforts made to recognize and predict the mutations, which can cause immune escape and track their emergence through impactful genomic surveillance.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/chemistry
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/immunology
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Binding Sites
- COVID-19/epidemiology
- COVID-19/pathology
- COVID-19/transmission
- COVID-19/virology
- COVID-19 Vaccines
- Genome, Viral
- Humans
- Immune Evasion/genetics
- Models, Molecular
- Mutation
- Phylogeny
- Protein Binding
- Protein Interaction Domains and Motifs
- Receptors, Virus/chemistry
- Receptors, Virus/genetics
- Receptors, Virus/immunology
- SARS-CoV-2/classification
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/pathogenicity
- Serine Endopeptidases/chemistry
- Serine Endopeptidases/genetics
- Serine Endopeptidases/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Virulence
Collapse
Affiliation(s)
- Raju Mukherjee
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Rohit Satardekar
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| |
Collapse
|
313
|
Marr LC, Tang JW. A Paradigm Shift to Align Transmission Routes With Mechanisms. Clin Infect Dis 2021; 73:1747-1749. [PMID: 34415335 DOI: 10.1093/cid/ciab722] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Indexed: 11/14/2022] Open
Abstract
Current infection-control guidelines subscribe to a contact/droplet/airborne paradigm that is based on outdated understanding. Here, we propose to modify and align existing guidelines with a more accurate description of the different transmission routes. This will improve the effectiveness of control measures as more transmissible variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerge.
Collapse
Affiliation(s)
- Linsey C Marr
- Civil and Environmental Engineering, Virginia Tech, Blacksburg, Virginia, USA
| | - Julian W Tang
- Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
314
|
Sonabend R, Whittles LK, Imai N, Perez-Guzman PN, Knock ES, Rawson T, Gaythorpe KAM, Djaafara BA, Hinsley W, FitzJohn RG, Lees JA, Kanapram DT, Volz EM, Ghani AC, Ferguson NM, Baguelin M, Cori A. Non-pharmaceutical interventions, vaccination, and the SARS-CoV-2 delta variant in England: a mathematical modelling study. Lancet 2021; 398:1825-1835. [PMID: 34717829 PMCID: PMC8550916 DOI: 10.1016/s0140-6736(21)02276-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/28/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND England's COVID-19 roadmap out of lockdown policy set out the timeline and conditions for the stepwise lifting of non-pharmaceutical interventions (NPIs) as vaccination roll-out continued, with step one starting on March 8, 2021. In this study, we assess the roadmap, the impact of the delta (B.1.617.2) variant of SARS-CoV-2, and potential future epidemic trajectories. METHODS This mathematical modelling study was done to assess the UK Government's four-step process to easing lockdown restrictions in England, UK. We extended a previously described model of SARS-CoV-2 transmission to incorporate vaccination and multi-strain dynamics to explicitly capture the emergence of the delta variant. We calibrated the model to English surveillance data, including hospital admissions, hospital occupancy, seroprevalence data, and population-level PCR testing data using a Bayesian evidence synthesis framework, then modelled the potential trajectory of the epidemic for a range of different schedules for relaxing NPIs. We estimated the resulting number of daily infections and hospital admissions, and daily and cumulative deaths. Three scenarios spanning a range of optimistic to pessimistic vaccine effectiveness, waning natural immunity, and cross-protection from previous infections were investigated. We also considered three levels of mixing after the lifting of restrictions. FINDINGS The roadmap policy was successful in offsetting the increased transmission resulting from lifting NPIs starting on March 8, 2021, with increasing population immunity through vaccination. However, because of the emergence of the delta variant, with an estimated transmission advantage of 76% (95% credible interval [95% CrI] 69-83) over alpha, fully lifting NPIs on June 21, 2021, as originally planned might have led to 3900 (95% CrI 1500-5700) peak daily hospital admissions under our central parameter scenario. Delaying until July 19, 2021, reduced peak hospital admissions by three fold to 1400 (95% CrI 700-1700) per day. There was substantial uncertainty in the epidemic trajectory, with particular sensitivity to the transmissibility of delta, level of mixing, and estimates of vaccine effectiveness. INTERPRETATION Our findings show that the risk of a large wave of COVID-19 hospital admissions resulting from lifting NPIs can be substantially mitigated if the timing of NPI relaxation is carefully balanced against vaccination coverage. However, with the delta variant, it might not be possible to fully lift NPIs without a third wave of hospital admissions and deaths, even if vaccination coverage is high. Variants of concern, their transmissibility, vaccine uptake, and vaccine effectiveness must be carefully monitored as countries relax pandemic control measures. FUNDING National Institute for Health Research, UK Medical Research Council, Wellcome Trust, and UK Foreign, Commonwealth and Development Office.
Collapse
Affiliation(s)
- Raphael Sonabend
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Lilith K Whittles
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK; National Institute for Health Research Health Protection Research Unit in Modelling Methodology, Imperial College London, Public Health England, London School of Hygiene & Tropical Medicine, London, UK; Modelling and Economics Unit, National Infection Service, Public Health England, London, UK
| | - Natsuko Imai
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Pablo N Perez-Guzman
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Edward S Knock
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK; National Institute for Health Research Health Protection Research Unit in Modelling Methodology, Imperial College London, Public Health England, London School of Hygiene & Tropical Medicine, London, UK
| | - Thomas Rawson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Katy A M Gaythorpe
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Bimandra A Djaafara
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Wes Hinsley
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Richard G FitzJohn
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - John A Lees
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Divya Thekke Kanapram
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Erik M Volz
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Azra C Ghani
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Neil M Ferguson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK; National Institute for Health Research Health Protection Research Unit in Modelling Methodology, Imperial College London, Public Health England, London School of Hygiene & Tropical Medicine, London, UK.
| | - Marc Baguelin
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK; National Institute for Health Research Health Protection Research Unit in Modelling Methodology, Imperial College London, Public Health England, London School of Hygiene & Tropical Medicine, London, UK; Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Anne Cori
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK; National Institute for Health Research Health Protection Research Unit in Modelling Methodology, Imperial College London, Public Health England, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
315
|
Nguyen D, Xiao J, Simmonds P, Lamikanra A, Odon V, Ratcliff J, Townsend A, Roberts DJ, Harvala H. Effects of SARS-CoV-2 strain variation on virus neutralisation titres: therapeutic use of convalescent plasma. J Infect Dis 2021; 225:971-976. [PMID: 34751775 PMCID: PMC8689936 DOI: 10.1093/infdis/jiab563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
We compared neutralising antibody titres of convalescent samples collected before and after the emergence of novel strains of SARS-CoV-2, against the wild-type virus (WT), Alpha (B.1.1.7) and Beta (B.1.351) variants. Plasma collected in 2020 before emergence of variants showed reduced titres against the Alpha variants, and both sets of samples demonstrated significantly reduced titres against Beta. Comparison of microneutralisation titres to those obtained with pseudotype and HAT assays showed a good correlation of titres and effects of strain variation, supporting the use of these simpler assays for assessment of convalescent plasma potency against currently circulating and emerging strains of SARS-CoV-2.
Collapse
Affiliation(s)
- Dung Nguyen
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Julie Xiao
- MRC Human Immunology Unit, MRC Weatherall Institute, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, UK
| | - Peter Simmonds
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | | | - Valerie Odon
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Jeremy Ratcliff
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Alain Townsend
- MRC Human Immunology Unit, MRC Weatherall Institute, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, UK.,Chinese Academy of Medical Science, Oxford Institute, University of Oxford, Oxford, UK
| | - David J Roberts
- Clinical Services, NHS Blood and Transplant, Oxford, UK.,Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, Oxford, UK
| | - Heli Harvala
- Chinese Academy of Medical Science, Oxford Institute, University of Oxford, Oxford, UK.,Microbiology Services, NHS Blood and Transplant, Colindale, UK
| |
Collapse
|
316
|
Neerukonda SN, Vassell R, Lusvarghi S, Wang R, Echegaray F, Bentley L, Eakin AE, Erlandson KJ, Katzelnick LC, Weiss CD, Wang W. SARS-COV-2 Delta variant displays moderate resistance to neutralizing antibodies and spike protein properties of higher soluble ACE2 sensitivity, enhanced cleavage and fusogenic activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.11.05.467523. [PMID: 34790980 PMCID: PMC8597883 DOI: 10.1101/2021.11.05.467523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The SARS-CoV-2 B.1.617 lineage variants, Kappa (B.1.617.1) and Delta (B.1.617.2, AY) emerged during the second wave of infections in India, but the Delta variants have become dominant worldwide and continue to evolve. The spike proteins of B.1.617.1, B.1.617.2, and AY.1 variants have several substitutions in the receptor binding domain (RBD), including L452R+E484Q, L452R+T478K, and K417N+L452R+T478K, respectively, that could potentially reduce effectiveness of therapeutic antibodies and current vaccines. Here we compared B.1.617 variants, and their single and double RBD substitutions for resistance to neutralization by convalescent sera, mRNA vaccine-elicited sera, and therapeutic neutralizing antibodies using a pseudovirus neutralization assay. Pseudoviruses with the B.1.617.1, B.1.617.2, and AY.1 spike showed a modest 1.5 to 4.4-fold reduction in neutralization titer by convalescent sera and vaccine-elicited sera. In comparison, similar modest reductions were also observed for pseudoviruses with C.37, P.1, R.1, and B.1.526 spikes, but seven- and sixteen-fold reduction for vaccine-elicited and convalescent sera, respectively, was seen for pseudoviruses with the B.1.351 spike. Four of twenty-three therapeutic neutralizing antibodies showed either complete or partial loss of neutralization against B.1.617.2 pseudoviruses due to the L452R substitution, whereas six of twenty-three therapeutic neutralizing antibodies showed either complete or partial loss of neutralization against B.1.617.1 pseudoviruses due to either the E484Q or L452R substitution. Against AY.1 pseudoviruses, the L452R and K417N substitutions accounted for the loss of neutralization by four antibodies and one antibody, respectively, whereas one antibody lost potency that could not be fully accounted for by a single RBD substitution. The modest resistance of B.1.617 variants to vaccine-elicited sera suggest that current mRNA-based vaccines will likely remain effective in protecting against B.1.617 variants, but the therapeutic antibodies need to be carefully selected based on their resistance profiles. Finally, the spike proteins of B.1.617 variants are more efficiently cleaved due to the P681R substitution, and the spike of Delta variants exhibited greater sensitivity to soluble ACE2 neutralization, as well as fusogenic activity, which may contribute to enhanced spread of Delta variants.
Collapse
|
317
|
Lou F, Li M, Pang Z, Jiang L, Guan L, Tian L, Hu J, Fan J, Fan H. Understanding the Secret of SARS-CoV-2 Variants of Concern/Interest and Immune Escape. Front Immunol 2021; 12:744242. [PMID: 34804024 PMCID: PMC8602852 DOI: 10.3389/fimmu.2021.744242] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/29/2021] [Indexed: 11/29/2022] Open
Abstract
The global pandemic of the coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), places a heavy burden on global public health. Four SARS-CoV-2 variants of concern including B.1.1.7, B.1.351, B.1.617.2, and P.1, and two variants of interest including C.37 and B.1.621 have been reported to have potential immune escape, and one or more mutations endow them with worrisome epidemiologic, immunologic, or pathogenic characteristics. This review introduces the latest research progress on SARS-CoV-2 variants of interest and concern, key mutation sites, and their effects on virus infectivity, mortality, and immune escape. Moreover, we compared the effects of various clinical SARS-CoV-2 vaccines and convalescent sera on epidemic variants, and evaluated the neutralizing capability of several antibodies on epidemic variants. In the end, SARS-CoV-2 evolution strategies in different transmission stages, the impact of different vaccination strategies on SARS-CoV-2 immune escape, antibody therapy strategies and COVID-19 epidemic control prospects are discussed. This review will provide a systematic and comprehensive understanding of the secret of SARS-CoV-2 variants of interest/concern and immune escape.
Collapse
Affiliation(s)
- Fuxing Lou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lin Jiang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lin Guan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jiaming Hu
- Tandon School of Engineering, New York University, New York, NY, United States
| | - Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
318
|
Zhang Z, Pandey R, Li J, Gu J, White D, Stacey HD, Ang JC, Steinberg C, Capretta A, Filipe CDM, Mossman K, Balion C, Miller MS, Salena BJ, Yamamura D, Soleymani L, Brennan JD, Li Y. High‐Affinity Dimeric Aptamers Enable the Rapid Electrochemical Detection of Wild‐Type and B.1.1.7 SARS‐CoV‐2 in Unprocessed Saliva. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202110819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Zijie Zhang
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
| | - Richa Pandey
- Department of Engineering Physics McMaster University Canada
| | - Jiuxing Li
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
| | - Dawn White
- Biointerfaces Institute McMaster University Canada
| | - Hannah D. Stacey
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- McMaster Immunology Research Centre McMaster University Canada
| | - Jann C. Ang
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- McMaster Immunology Research Centre McMaster University Canada
| | | | - Alfredo Capretta
- Biointerfaces Institute McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
| | | | - Karen Mossman
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- Department of Medicine McMaster University Canada
| | - Cynthia Balion
- Department of Pathology and Molecular Medicine McMaster University Canada
| | - Matthew S. Miller
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- McMaster Immunology Research Centre McMaster University Canada
| | | | - Deborah Yamamura
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- Department of Pathology and Molecular Medicine McMaster University Canada
| | - Leyla Soleymani
- Department of Engineering Physics McMaster University Canada
- School of Biomedical Engineering McMaster University 1280 Main Street West Hamilton Ontario L8S 4K1 Canada
| | | | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Biointerfaces Institute McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- School of Biomedical Engineering McMaster University 1280 Main Street West Hamilton Ontario L8S 4K1 Canada
| |
Collapse
|
319
|
Zhang Z, Pandey R, Li J, Gu J, White D, Stacey HD, Ang JC, Steinberg C, Capretta A, Filipe CDM, Mossman K, Balion C, Miller MS, Salena BJ, Yamamura D, Soleymani L, Brennan JD, Li Y. High-Affinity Dimeric Aptamers Enable the Rapid Electrochemical Detection of Wild-Type and B.1.1.7 SARS-CoV-2 in Unprocessed Saliva. Angew Chem Int Ed Engl 2021; 60:24266-24274. [PMID: 34464491 PMCID: PMC8596624 DOI: 10.1002/anie.202110819] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Indexed: 01/05/2023]
Abstract
We report a simple and rapid saliva-based SARS-CoV-2 antigen test that utilizes a newly developed dimeric DNA aptamer, denoted as DSA1N5, that specifically recognizes the spike proteins of the wildtype virus and its Alpha and Delta variants with dissociation constants of 120, 290 and 480 pM, respectively, and binds pseudotyped lentiviruses expressing the wildtype and alpha trimeric spike proteins with affinity constants of 2.1 pM and 2.3 pM, respectively. To develop a highly sensitive test, DSA1N5 was immobilized onto gold electrodes to produce an electrochemical impedance sensor, which was capable of detecting 1000 viral particles per mL in 1:1 diluted saliva in under 10 min without any further sample processing. Evaluation of 36 positive and 37 negative patient saliva samples produced a clinical sensitivity of 80.5 % and specificity of 100 % and the sensor could detect the wildtype virus as well as the Alpha and Delta variants in the patient samples, which is the first reported rapid test that can detect any emerging variant of SARS-CoV-2.
Collapse
Affiliation(s)
- Zijie Zhang
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
| | - Richa Pandey
- Department of Engineering PhysicsMcMaster UniversityCanada
| | - Jiuxing Li
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
| | - Dawn White
- Biointerfaces InstituteMcMaster UniversityCanada
| | - Hannah D. Stacey
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- McMaster Immunology Research CentreMcMaster UniversityCanada
| | - Jann C. Ang
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- McMaster Immunology Research CentreMcMaster UniversityCanada
| | | | - Alfredo Capretta
- Biointerfaces InstituteMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
| | | | - Karen Mossman
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- Department of MedicineMcMaster UniversityCanada
| | - Cynthia Balion
- Department of Pathology and Molecular MedicineMcMaster UniversityCanada
| | - Matthew S. Miller
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- McMaster Immunology Research CentreMcMaster UniversityCanada
| | | | - Deborah Yamamura
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- Department of Pathology and Molecular MedicineMcMaster UniversityCanada
| | - Leyla Soleymani
- Department of Engineering PhysicsMcMaster UniversityCanada
- School of Biomedical EngineeringMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | | | - Yingfu Li
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Biointerfaces InstituteMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- School of Biomedical EngineeringMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| |
Collapse
|
320
|
Chau NVV, Ngoc NM, Nguyet LA, Quang VM, Ny NTH, Khoa DB, Phong NT, Toan LM, Hong NTT, Tuyen NTK, Phat VV, Nhu LNT, Truc NHT, That BTT, Thao HP, Thao TNP, Vuong VT, Tam TTT, Tai NT, Bao HT, Nhung HTK, Minh NTN, Tien NTM, Huy NC, Choisy M, Man DNH, Ty DTB, Anh NT, Uyen LTT, Tu TNH, Yen LM, Dung NT, Hung LM, Truong NT, Thanh TT, Thwaites G, Tan LV. An observational study of breakthrough SARS-CoV-2 Delta variant infections among vaccinated healthcare workers in Vietnam. EClinicalMedicine 2021; 41:101143. [PMID: 34608454 PMCID: PMC8481205 DOI: 10.1016/j.eclinm.2021.101143] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Data on breakthrough SARS-CoV-2 Delta variant infections in vaccinated individuals are limited. METHODS We studied breakthrough infections among Oxford-AstraZeneca vaccinated healthcare workers in an infectious diseases hospital in Vietnam. We collected demographic and clinical data alongside serial PCR testing, measurement of SARS-CoV-2 antibodies, and viral whole-genome sequencing. FINDINGS Between 11th-25th June 2021 (7-8 weeks after the second dose), 69 staff tested positive for SARS-CoV-2. 62 participated in the study. Most were asymptomatic or mildly symptomatic and all recovered. Twenty-two complete-genome sequences were obtained; all were Delta variant and were phylogenetically distinct from contemporary viruses obtained from the community or from hospital patients admitted prior to the outbreak. Viral loads inferred from Ct values were 251 times higher than in cases infected with the original strain in March/April 2020. Median time from diagnosis to negative PCR was 21 days (range 8-33). Neutralizing antibodies (expressed as percentage of inhibition) measured after the second vaccine dose, or at diagnosis, were lower in cases than in uninfected, fully vaccinated controls (median (IQR): 69.4 (50.7-89.1) vs. 91.3 (79.6-94.9), p=0.005 and 59.4 (32.5-73.1) vs. 91.1 (77.3-94.2), p=0.002). There was no correlation between vaccine-induced neutralizing antibody levels and peak viral loads or the development of symptoms. INTERPRETATION Breakthrough Delta variant infections following Oxford-AstraZeneca vaccination may cause asymptomatic or mild disease, but are associated with high viral loads, prolonged PCR positivity and low levels of vaccine-induced neutralizing antibodies. Epidemiological and sequence data suggested ongoing transmission had occurred between fully vaccinated individuals. FUNDING Wellcome and NIH/NIAID.
Collapse
Affiliation(s)
| | - Nghiem My Ngoc
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | - Lam Anh Nguyet
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Vo Minh Quang
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | | | - Dao Bach Khoa
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | | | - Le Mau Toan
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | | | | | - Voong Vinh Phat
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | | | | | | | | | - Vo Trong Vuong
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | | | - Ngo Tan Tai
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | - Ho The Bao
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | | | | | | | - Nguy Cam Huy
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | - Marc Choisy
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Nguyen To Anh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | | | - Lam Minh Yen
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | - Le Manh Hung
- Hospital for Tropical Diseaes, Ho Chi Minh City, Vietnam
| | | | - Tran Tan Thanh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Guy Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Le Van Tan
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| |
Collapse
|
321
|
Arora P, Kempf A, Nehlmeier I, Graichen L, Sidarovich A, Winkler MS, Schulz S, Jäck HM, Stankov MV, Behrens GMN, Pöhlmann S, Hoffmann M. Delta variant (B.1.617.2) sublineages do not show increased neutralization resistance. Cell Mol Immunol 2021; 18:2557-2559. [PMID: 34635807 PMCID: PMC8503871 DOI: 10.1038/s41423-021-00772-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 02/08/2023] Open
Affiliation(s)
- Prerna Arora
- grid.418215.b0000 0000 8502 7018Infection Biology Unit, German Primate Center, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Amy Kempf
- grid.418215.b0000 0000 8502 7018Infection Biology Unit, German Primate Center, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Inga Nehlmeier
- grid.418215.b0000 0000 8502 7018Infection Biology Unit, German Primate Center, Göttingen, Germany
| | - Luise Graichen
- grid.418215.b0000 0000 8502 7018Infection Biology Unit, German Primate Center, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Anzhalika Sidarovich
- grid.418215.b0000 0000 8502 7018Infection Biology Unit, German Primate Center, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Martin S. Winkler
- grid.7450.60000 0001 2364 4210Department of Anesthesiology, University of Göttingen Medical Center, Georg-August University Göttingen, Göttingen, Germany
| | - Sebastian Schulz
- grid.5330.50000 0001 2107 3311Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- grid.5330.50000 0001 2107 3311Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Metodi V. Stankov
- grid.10423.340000 0000 9529 9877Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Georg M. N. Behrens
- grid.10423.340000 0000 9529 9877Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Stefan Pöhlmann
- grid.418215.b0000 0000 8502 7018Infection Biology Unit, German Primate Center, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Markus Hoffmann
- grid.418215.b0000 0000 8502 7018Infection Biology Unit, German Primate Center, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
322
|
Mlcochova P, Kemp SA, Dhar MS, Papa G, Meng B, Ferreira IATM, Datir R, Collier DA, Albecka A, Singh S, Pandey R, Brown J, Zhou J, Goonawardane N, Mishra S, Whittaker C, Mellan T, Marwal R, Datta M, Sengupta S, Ponnusamy K, Radhakrishnan VS, Abdullahi A, Charles O, Chattopadhyay P, Devi P, Caputo D, Peacock T, Wattal C, Goel N, Satwik A, Vaishya R, Agarwal M, Mavousian A, Lee JH, Bassi J, Silacci-Fegni C, Saliba C, Pinto D, Irie T, Yoshida I, Hamilton WL, Sato K, Bhatt S, Flaxman S, James LC, Corti D, Piccoli L, Barclay WS, Rakshit P, Agrawal A, Gupta RK. SARS-CoV-2 B.1.617.2 Delta variant replication and immune evasion. Nature 2021; 599:114-119. [PMID: 34488225 DOI: 10.1101/2021.05.08.443253] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/23/2021] [Indexed: 05/23/2023]
Abstract
The B.1.617.2 (Delta) variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in the state of Maharashtra in late 2020 and spread throughout India, outcompeting pre-existing lineages including B.1.617.1 (Kappa) and B.1.1.7 (Alpha)1. In vitro, B.1.617.2 is sixfold less sensitive to serum neutralizing antibodies from recovered individuals, and eightfold less sensitive to vaccine-elicited antibodies, compared with wild-type Wuhan-1 bearing D614G. Serum neutralizing titres against B.1.617.2 were lower in ChAdOx1 vaccinees than in BNT162b2 vaccinees. B.1.617.2 spike pseudotyped viruses exhibited compromised sensitivity to monoclonal antibodies to the receptor-binding domain and the amino-terminal domain. B.1.617.2 demonstrated higher replication efficiency than B.1.1.7 in both airway organoid and human airway epithelial systems, associated with B.1.617.2 spike being in a predominantly cleaved state compared with B.1.1.7 spike. The B.1.617.2 spike protein was able to mediate highly efficient syncytium formation that was less sensitive to inhibition by neutralizing antibody, compared with that of wild-type spike. We also observed that B.1.617.2 had higher replication and spike-mediated entry than B.1.617.1, potentially explaining the B.1.617.2 dominance. In an analysis of more than 130 SARS-CoV-2-infected health care workers across three centres in India during a period of mixed lineage circulation, we observed reduced ChAdOx1 vaccine effectiveness against B.1.617.2 relative to non-B.1.617.2, with the caveat of possible residual confounding. Compromised vaccine efficacy against the highly fit and immune-evasive B.1.617.2 Delta variant warrants continued infection control measures in the post-vaccination era.
Collapse
Affiliation(s)
- Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- University College London, London, UK
| | | | - Guido Papa
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Bo Meng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rawlings Datir
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dami A Collier
- Department of Medicine, University of Cambridge, Cambridge, UK
- University College London, London, UK
| | - Anna Albecka
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Sujeet Singh
- National Centre for Disease Control, Delhi, India
| | - Rajesh Pandey
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Jonathan Brown
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Jie Zhou
- Department of Infectious Diseases, Imperial College London, London, UK
| | | | - Swapnil Mishra
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Charles Whittaker
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Thomas Mellan
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Robin Marwal
- National Centre for Disease Control, Delhi, India
| | - Meena Datta
- National Centre for Disease Control, Delhi, India
| | | | | | | | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Priti Devi
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | | | - Tom Peacock
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | | | | | | | | | | | | | - Joo Hyeon Lee
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jessica Bassi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Christian Saliba
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Dora Pinto
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Takashi Irie
- Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Isao Yoshida
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | | | - Kei Sato
- Division of Systems Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Saitama, Japan
| | - Samir Bhatt
- National Centre for Disease Control, Delhi, India
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Seth Flaxman
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Leo C James
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Wendy S Barclay
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | | | - Anurag Agrawal
- CSIR Institute of Genomics and Integrative Biology, Delhi, India.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Africa Health Research Institute, Durban, South Africa.
| |
Collapse
|
323
|
Dupont L, Snell LB, Graham C, Seow J, Merrick B, Lechmere T, Maguire TJA, Hallett SR, Pickering S, Charalampous T, Alcolea-Medina A, Huettner I, Jimenez-Guardeño JM, Acors S, Almeida N, Cox D, Dickenson RE, Galao RP, Kouphou N, Lista MJ, Ortega-Prieto AM, Wilson H, Winstone H, Fairhead C, Su JZ, Nebbia G, Batra R, Neil S, Shankar-Hari M, Edgeworth JD, Malim MH, Doores KJ. Neutralizing antibody activity in convalescent sera from infection in humans with SARS-CoV-2 and variants of concern. Nat Microbiol 2021; 6:1433-1442. [PMID: 34654917 PMCID: PMC8556155 DOI: 10.1038/s41564-021-00974-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/03/2021] [Indexed: 12/17/2022]
Abstract
COVID-19 vaccine design and vaccination rollout need to take into account a detailed understanding of antibody durability and cross-neutralizing potential against SARS-CoV-2 and emerging variants of concern (VOCs). Analyses of convalescent sera provide unique insights into antibody longevity and cross-neutralizing activity induced by variant spike proteins, which are putative vaccine candidates. Using sera from 38 individuals infected in wave 1, we show that cross-neutralizing activity can be detected up to 305 days pos onset of symptoms, although sera were less potent against B.1.1.7 (Alpha) and B1.351 (Beta). Over time, despite a reduction in overall neutralization activity, differences in sera neutralization potency against SARS-CoV-2 and the Alpha and Beta variants decreased, which suggests that continued antibody maturation improves tolerance to spike mutations. We also compared the cross-neutralizing activity of wave 1 sera with sera from individuals infected with the Alpha, the Beta or the B.1.617.2 (Delta) variants up to 79 days post onset of symptoms. While these sera neutralize the infecting VOC and parental virus to similar levels, cross-neutralization of different SARS-CoV-2 VOC lineages is reduced. These findings will inform the optimization of vaccines to protect against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Liane Dupont
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Luke B Snell
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Carl Graham
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jeffrey Seow
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Blair Merrick
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Thomas Lechmere
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Thomas J A Maguire
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Sadie R Hallett
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Suzanne Pickering
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Themoula Charalampous
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Adela Alcolea-Medina
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Isabella Huettner
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jose M Jimenez-Guardeño
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Sam Acors
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Nathalia Almeida
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Daniel Cox
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Ruth E Dickenson
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Rui Pedro Galao
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Neophytos Kouphou
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Marie Jose Lista
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Ana Maria Ortega-Prieto
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Harry Wilson
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Helena Winstone
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Cassandra Fairhead
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jia Zhe Su
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gaia Nebbia
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Rahul Batra
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Stuart Neil
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Manu Shankar-Hari
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jonathan D Edgeworth
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Michael H Malim
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Katie J Doores
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK.
| |
Collapse
|
324
|
Daniels RS, Harvey R, Ermetal B, Xiang Z, Galiano M, Adams L, McCauley JW. A Sanger sequencing protocol for SARS-CoV-2 S-gene. Influenza Other Respir Viruses 2021; 15:707-710. [PMID: 34346163 PMCID: PMC8447197 DOI: 10.1111/irv.12892] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/14/2021] [Accepted: 07/18/2021] [Indexed: 11/29/2022] Open
Abstract
We describe a Sanger sequencing protocol for SARS-CoV-2 S-gene the Spike (S)-glycoprotein product of which, composed of receptor-binding (S1) and membrane fusion (S2) segments, is the target of vaccines used to combat COVID-19. The protocol can be used in laboratories with basic Sanger sequencing capabilities and allows rapid "at source" screening for SARS-CoV-2 variants, notably those of concern. The protocol has been applied for surveillance, with clinical specimens collected in either nucleic acid preservation lysis-mix or virus transport medium, and research involving cultured viruses, and can yield data of public health importance in a timely manner.
Collapse
Affiliation(s)
- Rodney S. Daniels
- Worldwide Influenza Centre (WIC: a WHO Collaborating Centre for Reference and Research on Influenza)The Francis Crick InstituteLondonUK
| | - Ruth Harvey
- Worldwide Influenza Centre (WIC: a WHO Collaborating Centre for Reference and Research on Influenza)The Francis Crick InstituteLondonUK
| | - Burcu Ermetal
- Worldwide Influenza Centre (WIC: a WHO Collaborating Centre for Reference and Research on Influenza)The Francis Crick InstituteLondonUK
| | - Zheng Xiang
- Worldwide Influenza Centre (WIC: a WHO Collaborating Centre for Reference and Research on Influenza)The Francis Crick InstituteLondonUK
| | - Monica Galiano
- Worldwide Influenza Centre (WIC: a WHO Collaborating Centre for Reference and Research on Influenza)The Francis Crick InstituteLondonUK
| | - Lorin Adams
- Worldwide Influenza Centre (WIC: a WHO Collaborating Centre for Reference and Research on Influenza)The Francis Crick InstituteLondonUK
| | - John W. McCauley
- Worldwide Influenza Centre (WIC: a WHO Collaborating Centre for Reference and Research on Influenza)The Francis Crick InstituteLondonUK
| |
Collapse
|
325
|
Khan A, Khan T, Ali S, Aftab S, Wang Y, Qiankun W, Khan M, Suleman M, Ali S, Heng W, Ali SS, Wei DQ, Mohammad A. SARS-CoV-2 new variants: Characteristic features and impact on the efficacy of different vaccines. Biomed Pharmacother 2021; 143:112176. [PMID: 34562770 PMCID: PMC8433040 DOI: 10.1016/j.biopha.2021.112176] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 01/10/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its new variants reported in different countries have posed a serious threat to human health and social fabrics worldwide. In addition, these new variants hindered the efforts of vaccines and other therapeutic developments. In this review article, we explained the emergence of new variants of SARS-CoV-2, their transmission risk, mortality rate, and, more importantly, the impact of each new variant on the efficacy of the developed vaccines reported in different literature and findings. The literature reported that with the emergence of new variants, the efficacy of different vaccines is declined, hospitalization and the risk of reinfection is increased. The reports concluded that the emergence of a variant that entirely evades the immune response triggered by the vaccine is improbable. The emergence of new variants and reports of re-infections are creating a more distressing situation and therefore demands further investigation to formulate an effective therapeutic strategy.
Collapse
Affiliation(s)
- Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Taimoor Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shughla Ali
- Department of Zoology, Swat College of Science and Technology (SCST), Swat, Khyber Pakhtunkhwa, Pakistan
| | - Summiya Aftab
- Department of Zoology, Government Girls Degree College, Thana, Khyber Pakhtunkhwa, Pakistan
| | - Yanjing Wang
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Wang Qiankun
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Mazhar Khan
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei 230027, Anhui, PR China
| | - Muhammad Suleman
- Center for Biotechnology and Microbiology, University of Swat, Kanju Campus, Swat, Pakistan
| | - Shahid Ali
- Center for Biotechnology and Microbiology, University of Swat, Kanju Campus, Swat, Pakistan
| | - Wang Heng
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Syed Shujait Ali
- Center for Biotechnology and Microbiology, University of Swat, Kanju Campus, Swat, Pakistan
| | - Dong-Qing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Peng Cheng Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nashan District, Shenzhen, Guangdong 518055, PR China; State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait
| |
Collapse
|
326
|
Abstract
Serological assays for measuring severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies have crucial applications in the control and surveillance of the current COVID-19 pandemic. A large number of such assays have been developed and are now commercially available. However, there are limited studies evaluating the performance of these tests. We evaluated the performances of the following six commercially available serological assays for detecting SARS-CoV-2 antibodies: (i) Genscript cPass surrogate virus neutralization test (Genscript cPass), (ii) Diasorin-SARS-CoV-2 S1/S2 IgG detection (Diasorin-S1/S2 IgG), (iii) Alinity SARS-CoV-2 IgG II (Alinity IgG II), (iv) Diasorin-SARS-CoV-2 TrimericS IgG (Diasorin-TrimericS IgG), (v) Roche Elecsys anti-SARS-CoV-2-cobas (Roche Elecsys), and (vi) AESKU enzyme linked immunosorbent assay (AESKULISA). The results of these tests were compared against the gold standard plaque reduction neutralization test (PRNT). Roche Elecsys had the highest sensitivity, and the Genscript cPass had the highest specificity. Diasorin-TrimericS IgG had the best overall performance with the highest agreement with the PRNT results. Parallel testing of Genscript cPass with Diasorin-TrimericS IgG and Diasorin-S1/S2 IgG had the optimum performance. Based on the receiver operating characteristic (ROC) curve, lowering the cutoff from 30% to 20% in the Genscript cPass significantly increased the sensitivity and the overall agreement with the PRNT results. Commercially available serological assays are good alternatives to the standard PRNT. However, further studies on larger sample numbers are required for optimization of the assay cutoff values and for evaluation of cost effectiveness. IMPORTANCE Commercial serological assays for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are now widely available. This study adds new knowledge regarding the optimization of these assays for evaluating postvaccination antibodies status. It highlights the positive and negative aspects of each assay in terms of sensitivity, specificity, and positive and negative predictive values, compared to the gold standard neutralization test. When using serological assays to assess postvaccine immune status, a balance of all parameters needs to be considered and not simply the high specificity. This balance is particularly relevant in the current situation where countries are aiming to mass vaccinate their populations and bring this pandemic under control. Assays with good sensitivity will have a lower percentage of false negatives and thus provide confidence for vaccination. Understanding the strengths and limitations of commercially available serological assays is important, not only for better application of these tests but also to understand the immune response and the duration of protection postvaccination.
Collapse
|
327
|
Alleva DG, Delpero AR, Scully MM, Murikipudi S, Ragupathy R, Greaves EK, Sathiyaseelan T, Haworth JR, Shah NJ, Rao V, Nagre S, Lancaster TM, Webb SS, Jasa AI, Ronca SE, Green FM, Elyard HA, Yee J, Klein J, Karnes L, Sollie F, Zion TC. Development of an IgG-Fc fusion COVID-19 subunit vaccine, AKS-452. Vaccine 2021; 39:6601-6613. [PMID: 34642088 PMCID: PMC8491978 DOI: 10.1016/j.vaccine.2021.09.077] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
AKS-452 is a biologically-engineered vaccine comprising an Fc fusion protein of the SARS-CoV-2 viral spike protein receptor binding domain antigen (Ag) and human IgG1 Fc (SP/RBD-Fc) in clinical development for the induction and augmentation of neutralizing IgG titers against SARS-CoV-2 viral infection to address the COVID-19 pandemic. The Fc moiety is designed to enhance immunogenicity by increasing uptake via Fc-receptors (FcγR) on Ag-presenting cells (APCs) and prolonging exposure due to neonatal Fc receptor (FcRn) recycling. AKS-452 induced approximately 20-fold greater neutralizing IgG titers in mice relative to those induced by SP/RBD without the Fc moiety and induced comparable long-term neutralizing titers with a single dose vs. two doses. To further enhance immunogenicity, AKS-452 was evaluated in formulations containing a panel of adjuvants in which the water-in-oil adjuvant, Montanide™ ISA 720, enhanced neutralizing IgG titers by approximately 7-fold after one and two doses in mice, including the neutralization of live SARS-CoV-2 virus infection of VERO-E6 cells. Furthermore, ISA 720-adjuvanted AKS-452 was immunogenic in rabbits and non-human primates (NHPs) and protected from infection and clinical symptoms with live SARS-CoV-2 virus in NHPs (USA-WA1/2020 viral strain) and the K18 human ACE2-trangenic (K18-huACE2-Tg) mouse (South African B.1.351 viral variant). These preclinical studies support the initiation of Phase I clinical studies with adjuvanted AKS-452 with the expectation that this room-temperature stable, Fc-fusion subunit vaccine can be rapidly and inexpensively manufactured to provide billions of doses per year especially in regions where the cold-chain is difficult to maintain.
Collapse
Affiliation(s)
- David G Alleva
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Andrea R Delpero
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Melanie M Scully
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Sylaja Murikipudi
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Ramya Ragupathy
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Emma K Greaves
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | | | - Jeffrey R Haworth
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Nishit J Shah
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Vidhya Rao
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Shashikant Nagre
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Thomas M Lancaster
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Sarah S Webb
- Biomere Biomedical Research Models, 57 Union St., Worcester, MA 01608, United States
| | - Allison I Jasa
- Biomere Biomedical Research Models, 57 Union St., Worcester, MA 01608, United States
| | - Shannon E Ronca
- Feigin ABSL-3 Facility, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Freedom M Green
- Feigin ABSL-3 Facility, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Hanne Andersen Elyard
- BIOQUAL, Inc., 9600 Medical Center Drive, Suite 101, Rockville, MD 20850-3336, United States
| | - JoAnn Yee
- Primate Assay Laboratory, CA National Primate Research Center, University of California, Davis, CA 95616, United States
| | - Jeffrey Klein
- Sinclair Research Center, 562 State Road DD, Auxvasse, MO 65231, United States
| | - Larry Karnes
- Sinclair Research Center, 562 State Road DD, Auxvasse, MO 65231, United States
| | - Frans Sollie
- Pharmaceutical Research Associates Group B.V., Amerikaweg 18, 9407 TK Assen, Netherlands
| | - Todd C Zion
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States.
| |
Collapse
|
328
|
Bernasconi A, Mari L, Casagrandi R, Ceri S. Data-driven analysis of amino acid change dynamics timely reveals SARS-CoV-2 variant emergence. Sci Rep 2021; 11:21068. [PMID: 34702903 PMCID: PMC8548498 DOI: 10.1038/s41598-021-00496-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023] Open
Abstract
Since its emergence in late 2019, the diffusion of SARS-CoV-2 is associated with the evolution of its viral genome. The co-occurrence of specific amino acid changes, collectively named ‘virus variant’, requires scrutiny (as variants may hugely impact the agent’s transmission, pathogenesis, or antigenicity); variant evolution is studied using phylogenetics. Yet, never has this problem been tackled by digging into data with ad hoc analysis techniques. Here we show that the emergence of variants can in fact be traced through data-driven methods, further capitalizing on the value of large collections of SARS-CoV-2 sequences. For all countries with sufficient data, we compute weekly counts of amino acid changes, unveil time-varying clusters of changes with similar—rapidly growing—dynamics, and then follow their evolution. Our method succeeds in timely associating clusters to variants of interest/concern, provided their change composition is well characterized. This allows us to detect variants’ emergence, rise, peak, and eventual decline under competitive pressure of another variant. Our early warning system, exclusively relying on deposited sequences, shows the power of big data in this context, and concurs to calling for the wide spreading of public SARS-CoV-2 genome sequencing for improved surveillance and control of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Anna Bernasconi
- Departement of Electronics, Information, and Bioengineering, Politecnico di Milano, 20133, Milan, Italy.
| | - Lorenzo Mari
- Departement of Electronics, Information, and Bioengineering, Politecnico di Milano, 20133, Milan, Italy
| | - Renato Casagrandi
- Departement of Electronics, Information, and Bioengineering, Politecnico di Milano, 20133, Milan, Italy
| | - Stefano Ceri
- Departement of Electronics, Information, and Bioengineering, Politecnico di Milano, 20133, Milan, Italy
| |
Collapse
|
329
|
Zhu W, Feng J, Li C, Wang H, Zhong Y, Zhou L, Zhang X, Zhang T. COVID-19 Risk Assessment for the Tokyo Olympic Games. Front Public Health 2021; 9:730611. [PMID: 34760863 PMCID: PMC8572808 DOI: 10.3389/fpubh.2021.730611] [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: 06/25/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023] Open
Abstract
Introduction: As of June 7, 2021, the outbreak of Coronavirus Disease 2019 (COVID-19) has spread to more than 200 countries. The global number of reported cases is more than 172.9 million, with more than 3.7 million deaths, and the number of infected individuals is still growing rapidly. Consequently, events and activities around the world were canceled or postponed, and the preparation for sporting events were greatly challenged. Under such circumstances, about 11,000 athletes from ~206 countries are arriving in Tokyo for the 32nd Summer Olympic Games. Therefore, it is urgently necessary to assess the occurrence and spread risk of COVID-19 for the Games. Objectives: To explore effective prevention and control measures for COVID-19 in large international events through simulations of different interventions according to risk assessment. Methods: We used a random model to calculate the number of initial infected patients and used Poisson distribution to determine the number of initial infected patients based on the number of countries involved. Furthermore, to simulate the COVID-19 transmission, the susceptible-exposed-symptomatic-asymptomatic-recovered-hospitalized (SEIARH) model was established based on the susceptible-exposed-infectious-recovered (SEIR) mathematical model of epidemic diseases. According to risk assessment indicators produced by different scenarios of the simulated interventions, the risk of COVID-19 transmission in Tokyo Olympic Games was assessed. Results: The current COVID-19 prevention measures proposed by the Japan Olympic Committee need to be enhanced. And large-scale vaccination will effectively control the spread of COVID-19. When the protective efficacy of vaccines is 78.1% or 89.8%, and if the vaccination rate of athletes reaches 80%, an epidemic prevention barrier can be established.
Collapse
Affiliation(s)
- Wenhui Zhu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jie Feng
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Cheng Li
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huimin Wang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yang Zhong
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lijun Zhou
- Sichuan Center for Disease Control and Prevention, Chengdu, China
| | - Xingyu Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Tao Zhang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
330
|
Vilches TN, Sah P, Abdollahi E, Moghadas SM, Galvani AP. Importance of non-pharmaceutical interventions in the COVID-19 vaccination era: A case study of the Seychelles. J Glob Health 2021; 11:03104. [PMID: 34671458 PMCID: PMC8501397 DOI: 10.7189/jogh.11.03104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Thomas N Vilches
- Center for Infectious Disease Modeling and Analysis (CIDMA), Yale School of Public Health, New Haven, Connecticut, USA.,Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada
| | - Pratha Sah
- Center for Infectious Disease Modeling and Analysis (CIDMA), Yale School of Public Health, New Haven, Connecticut, USA
| | - Elaheh Abdollahi
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada
| | - Seyed M Moghadas
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada
| | - Alison P Galvani
- Center for Infectious Disease Modeling and Analysis (CIDMA), Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
331
|
Ramesh S, Govindarajulu M, Parise RS, Neel L, Shankar T, Patel S, Lowery P, Smith F, Dhanasekaran M, Moore T. Emerging SARS-CoV-2 Variants: A Review of Its Mutations, Its Implications and Vaccine Efficacy. Vaccines (Basel) 2021; 9:1195. [PMID: 34696303 PMCID: PMC8537675 DOI: 10.3390/vaccines9101195] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/26/2021] [Accepted: 10/08/2021] [Indexed: 12/21/2022] Open
Abstract
The widespread increase in multiple severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants is causing a significant health concern in the United States and worldwide. These variants exhibit increased transmissibility, cause more severe disease, exhibit evasive immune properties, impair neutralization by antibodies from vaccinated individuals or convalescence sera, and reinfection. The Centers for Disease Control and Prevention (CDC) has classified SARS-CoV-2 variants into variants of interest, variants of concern, and variants of high consequence. Currently, four variants of concern (B.1.1.7, B.1.351, P.1, and B.1.617.2) and several variants of interests (B.1.526, B.1.525, and P.2) are characterized and are essential for close monitoring. In this review, we discuss the different SARS-CoV-2 variants, emphasizing variants of concern circulating the world and highlight the various mutations and how these mutations affect the characteristics of the virus. In addition, we discuss the most common vaccines and the various studies concerning the efficacy of these vaccines against different variants of concern.
Collapse
Affiliation(s)
- Sindhu Ramesh
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Rachel S. Parise
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Logan Neel
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Tharanath Shankar
- Department of Internal Medicine, Ramaiah Medical College and Hospital, Bengaluru 560054, Karnataka, India;
| | - Shriya Patel
- Department of Neuroscience, Middlebury College, Middlebury, VT 05753, USA;
| | - Payton Lowery
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Forrest Smith
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Timothy Moore
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| |
Collapse
|
332
|
Israelow B, Mao T, Klein J, Song E, Menasche B, Omer SB, Iwasaki A. Adaptive immune determinants of viral clearance and protection in mouse models of SARS-CoV-2. Sci Immunol 2021; 6:eabl4509. [PMID: 34623900 PMCID: PMC9047536 DOI: 10.1126/sciimmunol.abl4509] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/24/2021] [Indexed: 01/16/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused more than 160 million infections and more than 3 million deaths worldwide. Although effective vaccines are currently being deployed, the adaptive immune determinants that promote viral clearance and confer protection remain poorly defined. Using mouse models of SARS-CoV-2, we demonstrate that both humoral and cellular adaptive immunity contribute to viral clearance in the setting of primary infection. Furthermore, we find that either convalescent mice or mice that receive mRNA vaccination are protected from both homologous infection and infection with a variant of concern, B.1.351. In addition, we find that this protection is largely mediated by antibody response and not cellular immunity. These results highlight the in vivo protective capacity of antibodies generated to both vaccine and natural infection.
Collapse
Affiliation(s)
- Benjamin Israelow
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jonathan Klein
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Eric Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Bridget Menasche
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Saad B. Omer
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Yale Institute for Global Health, Yale University, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
333
|
Kucharski AJ, Hodcroft EB, Kraemer MUG. Sharing, synthesis and sustainability of data analysis for epidemic preparedness in Europe. LANCET REGIONAL HEALTH-EUROPE 2021; 9:100215. [PMID: 34642674 PMCID: PMC8495248 DOI: 10.1016/j.lanepe.2021.100215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Adam J Kucharski
- Centre for Epidemic Preparedness and Response, London School of Hygiene & Tropical Medicine, UK.,Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, UK
| | - Emma B Hodcroft
- Institute of Social and Preventive Medicine, University of Bern, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | |
Collapse
|
334
|
Robust Neutralizing Antibody Responses 6 Months Post Vaccination with BNT162b2: A Prospective Study in 308 Healthy Individuals. Life (Basel) 2021; 11:life11101077. [PMID: 34685448 PMCID: PMC8537997 DOI: 10.3390/life11101077] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 11/23/2022] Open
Abstract
Elucidating long-term immunity following COVID-19 vaccination is essential for decision-making regarding booster shots. The aim of this study was to investigate the kinetics of neutralizing antibodies (Nabs) against SARS-CoV-2 up to six months after the second vaccination dose with the BNT162b2 mRNA vaccine. Nabs levels were measured on days 1 (before the first vaccine shot), 8, 22 (before the second shot), 36, 50, and 3 and 6 months after the second vaccination (NCT04743388). Three hundred and eight healthy individuals without malignant disease were included in this study. At six months, 2.59% of the participants had a Nabs value less than 30%, while 11.9% had Nabs values of less than 50%. Importantly, 58% of the subjects had Nabs values of more than 75%. Nabs were initially eliminated at a relatively slow rate, but after three months their elimination was 5.7 times higher. Older age was inversely associated with Nabs levels at all examined timepoints. Interestingly, a population modeling analysis estimated that half of the subjects will have Nabs values less than 73.8% and 64.6% at 9 and 12 months, respectively, post vaccination completion. In conclusion, we found a persistent but declining anti-SARS-CoV-2 humoral immunity at six months following full vaccination with BNT162b2 in healthy individuals, which was more pronounced among older persons. These data may inform the public health policies regarding the prioritization of booster vaccine shots.
Collapse
|
335
|
Arora P, Sidarovich A, Krüger N, Kempf A, Nehlmeier I, Graichen L, Moldenhauer AS, Winkler MS, Schulz S, Jäck HM, Stankov MV, Behrens GMN, Pöhlmann S, Hoffmann M. B.1.617.2 enters and fuses lung cells with increased efficiency and evades antibodies induced by infection and vaccination. Cell Rep 2021; 37:109825. [PMID: 34614392 PMCID: PMC8487035 DOI: 10.1016/j.celrep.2021.109825] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/20/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022] Open
Abstract
The Delta variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), B.1.617.2, emerged in India and has spread to over 80 countries. B.1.617.2 replaced B.1.1.7 as the dominant virus in the United Kingdom, resulting in a steep increase in new infections, and a similar development is expected for other countries. Effective countermeasures require information on susceptibility of B.1.617.2 to control by antibodies elicited by vaccines and used for coronavirus disease 2019 (COVID-19) therapy. We show, using pseudotyping, that B.1.617.2 evades control by antibodies induced upon infection and BNT162b2 vaccination, although to a lesser extent as compared to B.1.351. We find that B.1.617.2 is resistant against bamlanivimab, a monoclonal antibody with emergency use authorization for COVID-19 therapy. Finally, we show increased Calu-3 lung cell entry and enhanced cell-to-cell fusion of B.1.617.2, which may contribute to augmented transmissibility and pathogenicity of this variant. These results identify B.1.617.2 as an immune evasion variant with increased capacity to enter and fuse lung cells.
Collapse
Affiliation(s)
- Prerna Arora
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Anzhalika Sidarovich
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Nadine Krüger
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Luise Graichen
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | | | - Martin S Winkler
- Department of Anesthesiology, University of Göttingen Medical Center, Göttingen, Georg-August University of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Sebastian Schulz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany
| | - Metodi V Stankov
- Department for Rheumatology and Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Georg M N Behrens
- Department for Rheumatology and Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany.
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany.
| |
Collapse
|
336
|
Restrepo-Henao A, Yomayusa N, Vega R, Morón L, Vaca C, Oñate J. La variante Delta del SARS-CoV-2: características e implicaciones para la salud pública en Colombia. REVISTA DE LA FACULTAD DE MEDICINA 2021. [DOI: 10.15446/revfacmed.v70n1.97460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
La variante Delta del SARS-CoV-2 se ha convertido en uno de los mayores desafíos en salud pública a nivel mundial, ya que, luego de su identificación en la India en diciembre de 2020, se ha extendido de manera rápida, afectando principalmente a los países con bajas tasas de vacunación, y aquellos que han flexibilizado las medidas de salud pública establecidas para controlar la pandemia por COVID-19. La variante Delta tiene una mayor capacidad de replicación y se asocia con cargas virales hasta 1.260 veces más altas en comparación con las de infecciones causadas por la cepa original, lo cual puede estar asociado a mayores probabilidades de hospitalización, ingreso a UCI, necesidad de oxigenoterapia, neumonía, o incluso muerte. Las personas con vacunación completa cuentan con una protección casi similar contra las variantes Delta y Alfa. Dado el impacto de Delta en los países afectados en los que es la variante dominante, es necesario que todos los países desarrollen planes de acción sistemáticos enfocados en implementar estrictas medidas de salud pública y sociales en el contexto de la pandemia por COVID-19 y aumentar la cobertura de vacunación.. Teniendo en cuenta lo anterior, el objetivo de esta reflexión es describir las principales características de la variante Delta, su impacto en la dinámica de la pandemia en algunos de los países en que ha sido detectada, la efectividad de las vacunas contra esta variante, y sus implicaciones para la salud pública en Colombia.
Collapse
|
337
|
Amatya B, Pandey P, Dawadi S, Manandhar S. COVID-19 in fully vaccinated Everest trekkers in Nepal. J Travel Med 2021; 28:6316239. [PMID: 34230969 PMCID: PMC8344616 DOI: 10.1093/jtm/taab098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 11/12/2022]
Abstract
COVID-19 in a fully mRNA-1273 vaccinated aspiring Everest summiteer and an Everest trekker who had two doses of ChAdOx1nCoV-19 are presented. Delta B.1.617.2 variant was sequenced in one of the cases. Travellers should be wary of starting up travel to under-vaccinated regions, particularly if they are at high risk for severe disease.
Collapse
Affiliation(s)
- Bhawana Amatya
- CIWEC Hospital and Travel Medicine Center, Lainchaur, Kathmandu, Nepal
| | - Prativa Pandey
- CIWEC Hospital and Travel Medicine Center, Lainchaur, Kathmandu, Nepal
| | - Suvash Dawadi
- CIWEC Hospital and Travel Medicine Center, Lainchaur, Kathmandu, Nepal
| | - Shanta Manandhar
- CIWEC Hospital and Travel Medicine Center, Lainchaur, Kathmandu, Nepal
| |
Collapse
|
338
|
Kamat S, Kumari M, Jayabaskaran C. Nano-engineered tools in the diagnosis, therapeutics, prevention, and mitigation of SARS-CoV-2. J Control Release 2021; 338:813-836. [PMID: 34478750 PMCID: PMC8406542 DOI: 10.1016/j.jconrel.2021.08.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/13/2021] [Accepted: 08/28/2021] [Indexed: 01/07/2023]
Abstract
The recent outbreak of SARS-CoV-2 has forever altered mankind resulting in the COVID-19 pandemic. This respiratory virus further manifests into vital organ damage, resulting in severe post COVID-19 complications. Nanotechnology has been moonlighting in the scientific community to combat several severe diseases. This review highlights the triune of the nano-toolbox in the areas of diagnostics, therapeutics, prevention, and mitigation of SARS-CoV-2. Nanogold test kits have already been on the frontline of rapid detection. Breath tests, magnetic nanoparticle-based nucleic acid detectors, and the use of Raman Spectroscopy present myriads of possibilities in developing point of care biosensors, which will ensure sensitive, affordable, and accessiblemass surveillance. Most of the therapeutics are trying to focus on blocking the viral entry into the cell and fighting with cytokine storm, using nano-enabled drug delivery platforms. Nanobodies and mRNA nanotechnology with lipid nanoparticles (LNPs) as vaccines against S and N protein have regained importance. All the vaccines coming with promising phase 3 clinical trials have used nano-delivery systems for delivery of vaccine-cargo, which are currently administered widely in many countries. The use of chemically diverse metal, carbon and polymeric nanoparticles, nanocages and nanobubbles demonstrate opportunities to develop anti-viral nanomedicine. In order to prevent and mitigate the viral spread, high-performance charged nanofiber filters, spray coating of nanomaterials on surfaces, novel materials for PPE kits and facemasks have been developed that accomplish over 90% capture of airborne SARS-CoV-2. Nano polymer-based disinfectants are being tested to make smart-transport for human activities. Despite the promises of this toolbox, challenges in terms of reproducibility, specificity, efficacy and emergence of new SARS-CoV-2 variants are yet to overcome.
Collapse
Affiliation(s)
- Siya Kamat
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Madhuree Kumari
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India.
| | - C Jayabaskaran
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| |
Collapse
|
339
|
Bar-On YM, Goldberg Y, Mandel M, Bodenheimer O, Freedman L, Kalkstein N, Mizrahi B, Alroy-Preis S, Ash N, Milo R, Huppert A. Protection of BNT162b2 Vaccine Booster against Covid-19 in Israel. N Engl J Med 2021. [PMID: 34525275 DOI: 10.1056/nejmoa2114255/suppl_file/nejmoa2114255_disclosures.pdf] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
BACKGROUND On July 30, 2021, the administration of a third (booster) dose of the BNT162b2 messenger RNA vaccine (Pfizer-BioNTech) was approved in Israel for persons who were 60 years of age or older and who had received a second dose of vaccine at least 5 months earlier. Data are needed regarding the effect of the booster dose on the rate of confirmed coronavirus 2019 disease (Covid-19) and the rate of severe illness. METHODS We extracted data for the period from July 30 through August 31, 2021, from the Israeli Ministry of Health database regarding 1,137,804 persons who were 60 years of age or older and had been fully vaccinated (i.e., had received two doses of BNT162b2) at least 5 months earlier. In the primary analysis, we compared the rate of confirmed Covid-19 and the rate of severe illness between those who had received a booster injection at least 12 days earlier (booster group) and those who had not received a booster injection (nonbooster group). In a secondary analysis, we evaluated the rate of infection 4 to 6 days after the booster dose as compared with the rate at least 12 days after the booster. In all the analyses, we used Poisson regression after adjusting for possible confounding factors. RESULTS At least 12 days after the booster dose, the rate of confirmed infection was lower in the booster group than in the nonbooster group by a factor of 11.3 (95% confidence interval [CI], 10.4 to 12.3); the rate of severe illness was lower by a factor of 19.5 (95% CI, 12.9 to 29.5). In a secondary analysis, the rate of confirmed infection at least 12 days after vaccination was lower than the rate after 4 to 6 days by a factor of 5.4 (95% CI, 4.8 to 6.1). CONCLUSIONS In this study involving participants who were 60 years of age or older and had received two doses of the BNT162b2 vaccine at least 5 months earlier, we found that the rates of confirmed Covid-19 and severe illness were substantially lower among those who received a booster (third) dose of the BNT162b2 vaccine.
Collapse
Affiliation(s)
- Yinon M Bar-On
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Yair Goldberg
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Micha Mandel
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Omri Bodenheimer
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Laurence Freedman
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Nir Kalkstein
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Barak Mizrahi
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Sharon Alroy-Preis
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Nachman Ash
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Ron Milo
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Amit Huppert
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| |
Collapse
|
340
|
Bar-On YM, Goldberg Y, Mandel M, Bodenheimer O, Freedman L, Kalkstein N, Mizrahi B, Alroy-Preis S, Ash N, Milo R, Huppert A. Protection of BNT162b2 Vaccine Booster against Covid-19 in Israel. N Engl J Med 2021; 385:1393-1400. [PMID: 34525275 PMCID: PMC8461568 DOI: 10.1056/nejmoa2114255] [Citation(s) in RCA: 812] [Impact Index Per Article: 203.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND On July 30, 2021, the administration of a third (booster) dose of the BNT162b2 messenger RNA vaccine (Pfizer-BioNTech) was approved in Israel for persons who were 60 years of age or older and who had received a second dose of vaccine at least 5 months earlier. Data are needed regarding the effect of the booster dose on the rate of confirmed coronavirus 2019 disease (Covid-19) and the rate of severe illness. METHODS We extracted data for the period from July 30 through August 31, 2021, from the Israeli Ministry of Health database regarding 1,137,804 persons who were 60 years of age or older and had been fully vaccinated (i.e., had received two doses of BNT162b2) at least 5 months earlier. In the primary analysis, we compared the rate of confirmed Covid-19 and the rate of severe illness between those who had received a booster injection at least 12 days earlier (booster group) and those who had not received a booster injection (nonbooster group). In a secondary analysis, we evaluated the rate of infection 4 to 6 days after the booster dose as compared with the rate at least 12 days after the booster. In all the analyses, we used Poisson regression after adjusting for possible confounding factors. RESULTS At least 12 days after the booster dose, the rate of confirmed infection was lower in the booster group than in the nonbooster group by a factor of 11.3 (95% confidence interval [CI], 10.4 to 12.3); the rate of severe illness was lower by a factor of 19.5 (95% CI, 12.9 to 29.5). In a secondary analysis, the rate of confirmed infection at least 12 days after vaccination was lower than the rate after 4 to 6 days by a factor of 5.4 (95% CI, 4.8 to 6.1). CONCLUSIONS In this study involving participants who were 60 years of age or older and had received two doses of the BNT162b2 vaccine at least 5 months earlier, we found that the rates of confirmed Covid-19 and severe illness were substantially lower among those who received a booster (third) dose of the BNT162b2 vaccine.
Collapse
Affiliation(s)
- Yinon M Bar-On
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Yair Goldberg
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Micha Mandel
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Omri Bodenheimer
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Laurence Freedman
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Nir Kalkstein
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Barak Mizrahi
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Sharon Alroy-Preis
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Nachman Ash
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Ron Milo
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Amit Huppert
- From the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), Technion-Israel Institute of Technology, Haifa (Y.G.), Hebrew University of Jerusalem (M.M.) and Israel Ministry of Health (O.B., S.A.-P., N.A.), Jerusalem, the Biostatistical and Biomathematical Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.F., A.H.), KI Research Institute, Kfar Malal (N.K., B.M.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| |
Collapse
|
341
|
Speer C, Schaier M, Nusshag C, Töllner M, Buylaert M, Kälble F, Reichel P, Grenz J, Süsal C, Zeier M, Schnitzler P, Morath C, Klein K, Benning L. Longitudinal Humoral Responses after COVID-19 Vaccination in Peritoneal and Hemodialysis Patients over Twelve Weeks. Vaccines (Basel) 2021; 9:1130. [PMID: 34696238 PMCID: PMC8539130 DOI: 10.3390/vaccines9101130] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/28/2021] [Accepted: 10/02/2021] [Indexed: 12/22/2022] Open
Abstract
It has been demonstrated that patients on hemo- or peritoneal dialysis are particularly susceptible to SARS-CoV-2 infection and impaired seroconversion compared to healthy controls. Follow-up data on vaccination response in dialysis patients is limited but is greatly needed to individualize and guide (booster) vaccination strategies. In this prospective, multicenter study we measured anti-spike S1 and neutralizing antibodies in 124 hemodialysis patients, 41 peritoneal dialysis patients, and 20 age- and sex-matched healthy controls over 12 weeks after homologous BNT162b2 vaccination. Compared to healthy controls, both hemodialysis and peritoneal dialysis patients had lower anti-S1 IgG antibodies (median (IQR) 7.0 (2.8-24.3) and 21.8 (5.8-103.9) versus 134.9 (23.8-283.6), respectively; p < 0.001 and p < 0.05) and a reduced SARS-CoV-2 spike protein-ACE2 binding inhibition caused by vaccine-induced antibodies (median (IQR) 56% (40-81) and 77% (52-89) versus 96% (90-98), respectively; p < 0.001 and p < 0.01) three weeks after the second vaccination. Twelve weeks after the second vaccination, the spike protein-ACE2 binding inhibition significantly decreased to a median (IQR) of 45% (31-60) in hemodialysis patients and 55% (36-78) in peritoneal dialysis patients, respectively (p < 0.001 and p < 0.05). Peritoneal dialysis patients mounted higher antibody levels compared with hemodialysis patients at all time points during the 12-week follow-up. Individual booster vaccinations in high-risk individuals without seroconversion or rapidly waning neutralizing antibody levels are required and further data on the neutralization of emerging variants of concern in these patients are urgently needed.
Collapse
Affiliation(s)
- Claudius Speer
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
- Molecular Medicine Partnership Unit Heidelberg, EMBL, 69120 Heidelberg, Germany
| | - Matthias Schaier
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Christian Nusshag
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Maximilian Töllner
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Mirabel Buylaert
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Florian Kälble
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Paula Reichel
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Julia Grenz
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Caner Süsal
- Institute of Immunology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Martin Zeier
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Christian Morath
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Katrin Klein
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| | - Louise Benning
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (C.N.); (M.T.); (M.B.); (F.K.); (P.R.); (J.G.); (M.Z.); (C.M.); (K.K.)
| |
Collapse
|
342
|
Caucci S, Corvaro B, Tiano SML, Valenza A, Longo R, Marinelli K, Ferreri ML, Spiridigliozzi P, Salvoni G, Bagnarelli P, Menzo S. Weak Cross-Lineage Neutralization by Anti SARS-CoV-2 Spike Antibodies after Natural Infection or Vaccination Is Rescued by Repeated Immunological Stimulation. Vaccines (Basel) 2021; 9:1124. [PMID: 34696232 PMCID: PMC8537215 DOI: 10.3390/vaccines9101124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 12/03/2022] Open
Abstract
After over one year of evolution, through billions of infections in humans, SARS-CoV-2 has evolved into a score of slightly divergent lineages. A few different amino acids in the spike proteins of these lineages can hamper both natural immunity against reinfection, and vaccine efficacy. In this study, the in vitro neutralizing potency of sera from convalescent COVID-19 patients and vaccinated subjects was analyzed against six different SARS-CoV-2 lineages, including the latest B.1.617.2 (or Delta variant), in order to assess the cross-neutralization by anti-spike antibodies. After both single dose vaccination, or natural infection, the neutralizing activity was low and fully effective only against the original lineage, while a double dose or a single dose of vaccine, even one year after natural infection, boosted the cross-neutralizing activity against different lineages. Neither binding, nor the neutralizing activity of sera after vaccination, could predict vaccine failure, underlining the need for additional immunological markers. This study points at the importance of the anamnestic response and repeated vaccine stimulations to elicit a reasonable cross-lineage neutralizing antibody response.
Collapse
Affiliation(s)
- Sara Caucci
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.C.); (B.C.); (S.M.L.T.); (P.B.)
| | - Benedetta Corvaro
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.C.); (B.C.); (S.M.L.T.); (P.B.)
| | - Sofia Maria Luigia Tiano
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.C.); (B.C.); (S.M.L.T.); (P.B.)
| | - Anna Valenza
- Virology Laboratory, Azienda Ospedaliera Ospedali Riuniti di Ancona, 60126 Ancona, Italy; (A.V.); (K.M.); (M.L.F.); (P.S.); (G.S.)
| | - Roberta Longo
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.C.); (B.C.); (S.M.L.T.); (P.B.)
| | - Katia Marinelli
- Virology Laboratory, Azienda Ospedaliera Ospedali Riuniti di Ancona, 60126 Ancona, Italy; (A.V.); (K.M.); (M.L.F.); (P.S.); (G.S.)
| | - Monica Lucia Ferreri
- Virology Laboratory, Azienda Ospedaliera Ospedali Riuniti di Ancona, 60126 Ancona, Italy; (A.V.); (K.M.); (M.L.F.); (P.S.); (G.S.)
| | - Patrik Spiridigliozzi
- Virology Laboratory, Azienda Ospedaliera Ospedali Riuniti di Ancona, 60126 Ancona, Italy; (A.V.); (K.M.); (M.L.F.); (P.S.); (G.S.)
| | - Giovanna Salvoni
- Virology Laboratory, Azienda Ospedaliera Ospedali Riuniti di Ancona, 60126 Ancona, Italy; (A.V.); (K.M.); (M.L.F.); (P.S.); (G.S.)
| | - Patrizia Bagnarelli
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.C.); (B.C.); (S.M.L.T.); (P.B.)
- Virology Laboratory, Azienda Ospedaliera Ospedali Riuniti di Ancona, 60126 Ancona, Italy; (A.V.); (K.M.); (M.L.F.); (P.S.); (G.S.)
| | - Stefano Menzo
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.C.); (B.C.); (S.M.L.T.); (P.B.)
- Virology Laboratory, Azienda Ospedaliera Ospedali Riuniti di Ancona, 60126 Ancona, Italy; (A.V.); (K.M.); (M.L.F.); (P.S.); (G.S.)
| |
Collapse
|
343
|
Quick C, Dey R, Lin X. Rejoinder: Regression Models for Understanding COVID-19 Epidemic Dynamics With Incomplete Data. J Am Stat Assoc 2021. [DOI: 10.1080/01621459.2021.2001340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Corbin Quick
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Rounak Dey
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Statistics, Faculty of Arts and Sciences, Harvard University, Cambridge, MA
| |
Collapse
|
344
|
Sharma P, Mishra S, Basu S, Kumar R, Tanwar N. Breakthrough Infection With Severe Acute Respiratory Syndrome Coronavirus 2 Among Healthcare Workers in Delhi: A Single-Institution Study. Cureus 2021; 13:e19070. [PMID: 34824945 PMCID: PMC8610433 DOI: 10.7759/cureus.19070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 11/10/2022] Open
Abstract
Introduction This study aimed to determine the breakthrough infection rate of coronavirus disease 2019 (COVID-19) (severe acute respiratory syndrome coronavirus 2 {SARS-CoV-2}) infection in healthcare workers (HCWs) vaccinated with either BBV152 or AZD1222 (ChAdOx1-S) vaccine. Methods A cross-sectional analysis was conducted at a medical college and hospital complex in Delhi, India, through telephonic interviews among HCWs who had received at least one dose of a COVID-19 vaccine during January-March 2021. Breakthrough infections were operationally defined as the occurrence of COVID-19 infection ≥14 days after administration of two doses of either COVID-19 vaccine. Data were entered in Epidata 3.1 (Odense, Denmark: EpiData Association) (single entered) and analyzed with IBM SPSS version 25 (Armonk, NY: IBM Corp.). A p-value < 0.05 was considered statistically significant. Results We enrolled 325 HCWs with a mean (SD) age of 29.1 (9.9) years including 211 (64.9%) males and 114 (35.1%) females. A total of 37 (13.3%, 95% CI 9.8, 17.7) COVID-19 breakthrough infections were observed in the HCWs. Additionally, 20 (6.1%) non-breakthrough infections were reported in the HCWs who were vaccinated with at least a single dose of a COVID-19 vaccine, or both doses, but prior to 14 days since the administration of the second dose. Most breakthrough infections were mild without needing supplemental oxygen for recovery. Conclusion Nearly one in seven HCWs experienced a COVID-19 breakthrough infection in the present study. A history of SARS-CoV-2 natural infection followed by at least one dose of COVID-19 vaccination was associated with significant protection against breakthrough infections.
Collapse
Affiliation(s)
- Pragya Sharma
- Community Medicine, Maulana Azad Medical College, Delhi, IND
| | - Suruchi Mishra
- Community Medicine, Maulana Azad Medical College, Delhi, IND
| | - Saurav Basu
- Community Medicine, Maulana Azad Medical College, Delhi, IND
| | - Rajesh Kumar
- Community Medicine, Maulana Azad Medical College, Delhi, IND
| | - Neha Tanwar
- Community Medicine, Maulana Azad Medical College, Delhi, IND
| |
Collapse
|
345
|
Motta FC, McGoff KA, Deckard A, Wolfe CR, Bonsignori M, Moody MA, Cavanaugh K, Denny TN, Harer J, Haase SB. Assessment of Simulated Surveillance Testing and Quarantine in a SARS-CoV-2-Vaccinated Population of Students on a University Campus. JAMA HEALTH FORUM 2021; 2:e213035. [PMID: 35977169 PMCID: PMC8727034 DOI: 10.1001/jamahealthforum.2021.3035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
Importance The importance of surveillance testing and quarantine on university campuses to limit SARS-CoV-2 transmission needs to be reevaluated in the context of a complex and rapidly changing environment that includes vaccines, variants, and waning immunity. Also, recent US Centers for Disease Control and Prevention guidelines suggest that vaccinated students do not need to participate in surveillance testing. Objective To evaluate the use of surveillance testing and quarantine in a fully vaccinated student population for whom vaccine effectiveness may be affected by the type of vaccination, presence of variants, and loss of vaccine-induced or natural immunity over time. Design Setting and Participants In this simulation study, an agent-based Susceptible, Exposed, Infected, Recovered model was developed with some parameters estimated using data from the 2020 to 2021 academic year at Duke University (Durham, North Carolina) that described a simulated population of 5000 undergraduate students residing on campus in residential dormitories. This study assumed that 100% of residential undergraduates are vaccinated. Under varying levels of vaccine effectiveness (90%, 75%, and 50%), the reductions in the numbers of positive cases under various mitigation strategies that involved surveillance testing and quarantine were estimated. Main Outcomes and Measures The percentage of students infected with SARS-CoV-2 each day for the course of the semester (100 days) and the total number of isolated or quarantined students were estimated. Results A total of 5000 undergraduates were simulated in the study. In simulations with 90% vaccine effectiveness, weekly surveillance testing was associated with only marginally reduced viral transmission. At 50% to 75% effectiveness, surveillance testing was estimated to reduce the number of infections by as much as 93.6%. A 10-day quarantine protocol for exposures was associated with only modest reduction in infections until vaccine effectiveness dropped to 50%. Increased testing of reported contacts was estimated to be at least as effective as quarantine at limiting infections. Conclusions and Relevance In this simulated modeling study of infection dynamics on a college campus where 100% of the student body is vaccinated, weekly surveillance testing was associated with a substantial reduction of campus infections with even a modest loss of vaccine effectiveness. Model simulations also suggested that an increased testing cadence can be as effective as a 10-day quarantine period at limiting infections. Together, these findings provide a potential foundation for universities to design appropriate mitigation protocols for the 2021 to 2022 academic year.
Collapse
Affiliation(s)
- Francis C. Motta
- Department of Mathematical Sciences, Florida Atlantic University, Boca Raton
| | - Kevin A. McGoff
- Department of Mathematics and Statistics, University of North Carolina, Charlotte
| | - Anastasia Deckard
- Office of Information Technology, Duke University, Durham, North Carolina
| | - Cameron R. Wolfe
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Mattia Bonsignori
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
- Department of Medicine, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina
| | - M. Anthony Moody
- Department of Pediatrics and Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina
| | - Kyle Cavanaugh
- Office of Vice President of Administration, Department of Family Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Thomas N. Denny
- Department of Medicine, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina
| | - John Harer
- Department of Mathematics, Duke University, Durham, North Carolina
| | - Steven B. Haase
- Departments of Biology and Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
346
|
Mahajan NN, Pophalkar M, Patil S, Yewale B, Chaaithanya IK, Mahale SD, Gajbhiye RK. Pregnancy Outcomes and Maternal Complications During the Second Wave of Coronavirus Disease 2019 (COVID-19) in India. Obstet Gynecol 2021; 138:660-662. [PMID: 34233345 PMCID: PMC8454281 DOI: 10.1097/aog.0000000000004529] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022]
Abstract
Rates of severe coronavirus disease 2019 (COVID-19), intensive care unit admission, and maternal mortality increased among pregnant and postpartum women admitted for COVID-19 in the second wave compared with the first wave in India.
Collapse
Affiliation(s)
- Niraj N. Mahajan
- Department of Obstetrics and Gynecology, Topiwala National Medical College & BYL Nair Charitable Hospital, and the ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Madhura Pophalkar
- Department of Obstetrics and Gynecology, Topiwala National Medical College & BYL Nair Charitable Hospital, and the ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Sarika Patil
- Department of Obstetrics and Gynecology, Topiwala National Medical College & BYL Nair Charitable Hospital, and the ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Bhagyashree Yewale
- Department of Obstetrics and Gynecology, Topiwala National Medical College & BYL Nair Charitable Hospital, and the ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Itta Krishna Chaaithanya
- Department of Obstetrics and Gynecology, Topiwala National Medical College & BYL Nair Charitable Hospital, and the ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Smita D. Mahale
- Department of Obstetrics and Gynecology, Topiwala National Medical College & BYL Nair Charitable Hospital, and the ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Rahul K. Gajbhiye
- Department of Obstetrics and Gynecology, Topiwala National Medical College & BYL Nair Charitable Hospital, and the ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
347
|
Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination for Paediatric Patients With Inflammatory Bowel Diseases. J Pediatr Gastroenterol Nutr 2021; 73:433-436. [PMID: 34347673 DOI: 10.1097/mpg.0000000000003260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this communication, the members of the Porto group (the European Society for Paediatric Gastroenterology, Hepatology and Nutrition [ESPGHAN], inflammatory bowel diseases [IBD] working group) provide the current available evidence regarding vaccination of children and young adolescents with IBD against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our intent is to help provide meaningful answers to the concerns that parents and adolescents may have.
Collapse
|
348
|
Chauhan N, Soni S, Jain U. Optimizing testing regimes for the detection of COVID-19 in children and older adults. Expert Rev Mol Diagn 2021; 21:999-1016. [PMID: 34324823 PMCID: PMC8425447 DOI: 10.1080/14737159.2021.1962708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/28/2021] [Indexed: 02/08/2023]
Abstract
Introduction: Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection is a major pandemic and continuously emerging due to unclear prognosis and unavailability of reliable detection tools. Older adults are more susceptible to COVID-19 than children showing mature Angiotensin-Converting Enzyme 2 (ACE2), low concentration of immune targets, and comorbid conditions. Several detection platforms have been commercialized to date and more are in pipeline, however, the rate of false-positive results and rapid mutation of SARS-CoV-2 is increasing. Additionally, physiological, and geographical variations of affected individuals are also calling for diagnostic methods optimization.Areas Covered: Extensive information related to the optimization and usefulness of SARS-CoV-2 diagnostic methods based on sensitivity and specificity as definitive and feasible investigative tools is discussed. Moreover, an option of combining laboratory diagnostic methods to improve diagnostic strategies is also proposed and discussed in the comparative section of optimization studies.Expert Opinion: The review article explains the importance of optimization strategies for SARS-CoV-2 detection in children and older adults. There are advancements in COVID-19 detection including CRISPR-based, electrochemical, and optical-based sensing systems. However, the lack of sufficient studies on a comparative evaluation of standardized SARS-CoV-2 diagnostic methods among children and older adults, limit the authentication of commercialized kits.
Collapse
Affiliation(s)
- Nidhi Chauhan
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Shringika Soni
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Utkarsh Jain
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| |
Collapse
|
349
|
Tregoning JS, Flight KE, Higham SL, Wang Z, Pierce BF. Progress of the COVID-19 vaccine effort: viruses, vaccines and variants versus efficacy, effectiveness and escape. Nat Rev Immunol 2021; 21:626-636. [PMID: 34373623 PMCID: PMC8351583 DOI: 10.1038/s41577-021-00592-1] [Citation(s) in RCA: 736] [Impact Index Per Article: 184.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 02/07/2023]
Abstract
Where 2020 saw the development and testing of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) at an unprecedented pace, the first half of 2021 has seen vaccine rollout in many countries. In this Progress article, we provide a snapshot of ongoing vaccine efficacy studies, as well as real-world data on vaccine effectiveness and the impact of virus variants of concern. Where they have been deployed in a high proportion of the adult population, the currently approved vaccines have been extremely effective in preventing COVID-19, particularly severe disease. Nonetheless, there are still significant challenges in ensuring equitable vaccine access around the globe and lessons that can be learned for controlling this pandemic and for the next pandemic.
Collapse
Affiliation(s)
- John S Tregoning
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK.
| | - Katie E Flight
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| | - Sophie L Higham
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| | - Ziyin Wang
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| | - Benjamin F Pierce
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, UK
| |
Collapse
|
350
|
Böttcher L, Nagler J. Decisive conditions for strategic vaccination against SARS-CoV-2. CHAOS (WOODBURY, N.Y.) 2021; 31:101105. [PMID: 34717322 DOI: 10.1063/5.0066992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
While vaccines against severe acute respiratory syndrome coronavirus (SARS-CoV-2) are being administered, in many countries it may still take months until their supply can meet demand. The majority of available vaccines elicit strong immune responses when administered as prime-boost regimens. Since the immunological response to the first ("prime") dose may provide already a substantial reduction in infectiousness and protection against severe disease, it may be more effective-under certain immunological and epidemiological conditions-to vaccinate as many people as possible with only one dose instead of administering a person a second ("booster") dose. Such a vaccination campaign may help to more effectively slow down the spread of SARS-CoV-2 and reduce hospitalizations and fatalities. The conditions that make prime-first vaccination favorable over prime-boost campaigns, however, are not well understood. By combining epidemiological modeling, random-sampling techniques, and decision tree learning, we find that prime-first vaccination is robustly favored over prime-boost vaccination campaigns even for low single-dose efficacies. For epidemiological parameters that describe the spread of coronavirus disease 2019 (COVID-19), recent data on new variants included, we show that the difference between prime-boost and single-shot waning rates is the only discriminative threshold, falling in the narrow range of 0.01-0.02 day-1 below which prime-first vaccination should be considered.
Collapse
Affiliation(s)
- Lucas Böttcher
- Computational Social Science, Centre for Human and Machine Intelligence, Frankfurt School of Finance & Management, 60322 Frankfurt am Main, Germany
| | - Jan Nagler
- Deep Dynamics Group, Centre for Human and Machine Intelligence, Frankfurt School of Finance & Management, 60322 Frankfurt am Main, Germany
| |
Collapse
|