51
|
Fülöp GA, Gógl Á, Lakatos B, Sydó N, Csulak E, Mester B, Tóth S, Petrov Á, Nagy VK, Merkely G, Merkely B. Swimming competitions in the era of COVID-19: Lessons from successfully hosting the International Swimming League. Physiol Int 2022; 109:511-523. [DOI: 10.1556/2060.2022.00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 08/08/2022] [Accepted: 09/16/2022] [Indexed: 12/13/2022]
Abstract
AbstractBackgroundOrganization of mass sport events in the COVID-19 era is utterly complicated. Containments measures, required to avoid a virus outbreak, force athletes to compete under circumstances they never experienced before, most likely having a deleterious effect on their performance.PurposeWe aimed to design a so-called athlete-friendly bubble system for the International Swimming League 2020 event, which is strict enough to avoid a COVID-19 outbreak, but still provides a supportive environment for the athletes.MethodsTo avoid the feeling of imprisonment, athletes were permitted to spend a certain amount of time in the parks surrounding the hotels. Such alleviations were possible to apply with strict adherence to the hygienic and social distancing protocols and regular COVID-19 testing. Evaluation of every COVID-19 positive case was key, and if prolonged PCR positivity or false positive PCR result was identified, the unnecessary quarantine was planned to be lifted. Return to play protocol (RTP) was planned, in case of a COVID-19 infection of an athlete inside the bubble. To test, if the athlete-friendly system provided a supportive environment, we evaluated athlete performance.Results11,480 PCR tests were performed for 1,421 individuals. 63 COVID-19 positive cases were detected, of which 5 turned out to be clinically insignificant, either because of prolonged PCR positivity or because of a false positive result. 93.1% of the positive cases were detected in the local crew, while no athlete got infected inside the bubble, as the two infected athletes were tested positive upon arrival. RTP was provided for two athletes. 85% of the athletes showed improvement during the bubble and 8 world records were broken.ConclusionThe applied protocol proved to be effective, as no athlete got infected inside the bubble, moreover, the athlete-friendly system supported the athletes to improve their performance.
Collapse
Affiliation(s)
- Gábor A. Fülöp
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Álmos Gógl
- Szent Rókus Hospital of Semmelweis University, Budapest, Hungary
| | - Bálint Lakatos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Nóra Sydó
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Emese Csulak
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Bianka Mester
- Division of Hospital Hygiene, Semmelweis University, Budapest, Hungary
| | | | | | | | - Gergo Merkely
- Semmelweis University, Budapest, Hungary
- Orthopedic Department, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
52
|
SARIMA model-based forecasting required number of COVID-19 vaccines globally and empirical analysis of peoples’ view towards the vaccines. ALEXANDRIA ENGINEERING JOURNAL 2022; 61:12091-12110. [PMCID: PMC9174436 DOI: 10.1016/j.aej.2022.05.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/02/2022] [Accepted: 05/30/2022] [Indexed: 06/19/2023]
Abstract
Recent studies regarding COVID-19 show a growing tendency to talk about the COVID-19 Pandemic on online channels. With the recent release of the Pfizer vaccine of COVID-19, people keep posting many rumors regarding the safety concerns of the Vaccine, especially among older people. Due to the rapid spread of the COVID-19 virus and the worldwide Pandemic developed, the rush to develop the COVID-19 Vaccine has become an alarming priority in health care services worldwide. In this research work, we have systematically evaluated people’s views towards the COVID-19 Vaccine, and shreds of evidence are supported empirically. The study mainly focuses on the empirical evidence and intensive discussions on what is currently known about the mechanism of action, efficacy, and toxicity of the most promising vaccines (Moderna), (Pfizer/BioNtech), (Astrazenac/Oxford), and (Sputnik V) against COVID-19. Our study’s primary objective is to provide an analysis of the questionnaire regarding people’s opinions, preferences, and acceptance of the COVID-19 vaccines. We have created an online questionnaire using a google form to collect data from various countries supposed to employ COVID-19 vaccines. The questionnaires were distributed to people in many Arab and foreign countries such as Egypt, Saudi Arabia, India, England, China, and Japan. A total of 516 responses were returned and analyzed using statistical, and Seasonal Autoregressive Integrated Moving Average (SARIMA) approaches. The SARIMA model is used to predict the total number of vaccines in the next few days. To attain the most accurate forecast and prediction, the SARIMA model parameters are investigated with a grid search method. Finally, the combination of the parameters (1,0,1)×(1,0,0,1) is considered to be the best SARIMA model because it has the lowest AIC values of −4100.11 and the best Correlation coefficients of 0.984.
Collapse
|
53
|
Deng L, Li P, Zhang X, Jiang Q, Turner D, Zhou C, Gao Y, Qian F, Zhang C, Lu H, Zou H, Vermund SH, Qian HZ. Risk of SARS-CoV-2 reinfection: a systematic review and meta-analysis. Sci Rep 2022; 12:20763. [PMID: 36456577 PMCID: PMC9714387 DOI: 10.1038/s41598-022-24220-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 11/11/2022] [Indexed: 12/02/2022] Open
Abstract
This meta-analysis aims to synthesize global evidence on the risk of reinfection among people previously infected with SARS-CoV-2. We systematically searched PubMed, Scopus, Embase and Web of Science as of April 5, 2021. We conducted: (1) meta-analysis of cohort studies containing data sufficient for calculating the incidence rate of SARS-CoV-2 reinfection; (2) systematic review of case reports with confirmed SARS-CoV-2 reinfection cases. The reinfection incidence was pooled by zero-inflated beta distribution. The hazard ratio (HR) between reinfection incidence among previously infected individuals and new infection incidence among infection-naïve individuals was calculated using random-effects models. Of 906 records retrieved and reviewed, 11 studies and 11 case reports were included in the meta-analysis and the systematic review, respectively. The pooled SARS-CoV-2 reinfection incidence rate was 0.70 (standard deviation [SD] 0.33) per 10,000 person-days. The incidence of reinfection was lower than the incidence of new infection (HR = 0.12, 95% confidence interval 0.09-0.17). Our meta-analysis of studies conducted prior to the emergency of the more transmissible Omicron variant showed that people with a prior SARS-CoV-2 infection could be re-infected, and they have a lower risk of infection than those without prior infection. Continuing reviews are needed as the reinfection risk may change due to the rapid evolution of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Luojia Deng
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Peiqi Li
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuezhixing Zhang
- Yale School of Public Health, Yale University, 300 George Street, New Haven, CT, USA
| | - Qianxue Jiang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | | | - Chao Zhou
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yanxiao Gao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Frank Qian
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ci Zhang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Hui Lu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Huachun Zou
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Sten H Vermund
- Yale School of Public Health, Yale University, 300 George Street, New Haven, CT, USA
| | - Han-Zhu Qian
- Yale School of Public Health, Yale University, 300 George Street, New Haven, CT, USA.
- GSK plc, Rockville, MD, USA.
| |
Collapse
|
54
|
Field CJ, Heinly TA, Patel DR, Sim DG, Luley E, Gupta SL, Vanderford TH, Wrammert J, Sutton TC. Immune durability and protection against SARS-CoV-2 re-infection in Syrian hamsters. Emerg Microbes Infect 2022; 11:1103-1114. [PMID: 35333692 PMCID: PMC9037228 DOI: 10.1080/22221751.2022.2058419] [Citation(s) in RCA: 9] [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: 12/13/2021] [Accepted: 03/22/2022] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has caused a pandemic. As immunity to endemic human coronaviruses (i.e. NL63 or OC43) wanes leading to re-infection, it was unknown if SARS-CoV-2 immunity would also decline permitting repeat infections. Recent case reports confirm previously infected individuals can become re-infected; however, re-infection may be due to heterogeneity in the initial infection or the host immune response, or may be the result of infection with a variant strain that escapes pre-existing immunity. To control these variables, we utilized the Syrian hamster model to evaluate the duration of immunity and susceptibility to re-infection with SARS-CoV-2. Hamsters were given a primary mock or SARS-CoV-2 infection (culture media or 105 TCID50 USA/WA1/2020 isolate, respectively). Mock and SARS-CoV-2 infected hamsters were then given a secondary SARS-CoV-2 infection at 1, 2, 4, or 6 months post-primary infection (n = 14/time point/group). After the primary SARS-CoV-2 infection, hamsters developed anti-spike protein IgG, IgA, and neutralizing antibodies, and these antibodies were maintained for at least 6 months. Upon secondary SARS-CoV-2 challenge, previously SARS-CoV-2 infected animals were protected from weight loss, while all previously mock-infected animals became infected and lost weight. Importantly, despite having high titres of antibodies, one SARS-CoV-2 infected animal re-challenged at 4 months had a breakthrough infection with replicating virus in the upper and lower respiratory tract. These studies demonstrate immunity to SARS-CoV-2 is maintained for 6 months; however, protection may be incomplete and, even in the presence of high antibody titres, previously infected hosts may become re-infected.
Collapse
Affiliation(s)
- C. J. Field
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA, USA
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), University Park, PA, USA
| | - T. A. Heinly
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA, USA
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), University Park, PA, USA
| | - D. R. Patel
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA, USA
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - D. G. Sim
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - E. Luley
- Animal Diagnostic Lab, The Pennsylvania State University, University Park, PA, USA
| | - S. L. Gupta
- Department of Pediatrics, Division of Infectious Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - T. H. Vanderford
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - J. Wrammert
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), University Park, PA, USA
- Department of Pediatrics, Division of Infectious Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - T. C. Sutton
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA, USA
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), University Park, PA, USA
| |
Collapse
|
55
|
Abdolmaleki G, Taheri MA, Paridehpour S, Mohammadi NM, Tabatabaei YA, Mousavi T, Amin M. A comparison between SARS-CoV-1 and SARS-CoV2: an update on current COVID-19 vaccines. Daru 2022; 30:379-406. [PMID: 36050585 PMCID: PMC9436716 DOI: 10.1007/s40199-022-00446-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/05/2022] [Indexed: 10/31/2022] Open
Abstract
Since the outbreak of the novel coronavirus disease 2019 (COVID-19) in Wuhan, China, many health care systems have been heavily engaged in treating and preventing the disease, and the year 2020 may be called as "historic COVID-19 vaccine breakthrough". Due to the COVID-19 pandemic, many companies have initiated investigations on developing an efficient and safe vaccine against the virus. From Moderna and Pfizer in the United States to PastocoVac in Pasteur Institute of Iran and the University of Oxford in the United Kingdom, different candidates have been introduced to the market. COVID-19 vaccine research has been facilitated based on genome and structural information, bioinformatics predictions, epitope mapping, and data obtained from the previous developments of severe acute respiratory syndrome coronavirus (SARS-CoV or SARS-CoV-1) and middle east respiratory syndrome coronavirus (MERS-CoV) vaccine candidates. SARS-CoV genome sequence is highly homologous to the one in COVID-19 and both viruses use the same receptor, angiotensin-converting enzyme 2 (ACE2). Moreover, the immune system responds to these viruses, partially in the same way. Considering the on-going COVID-19 pandemic and previous attempts to manufacture SARS-CoV vaccines, this paper is going to discuss clinical cases as well as vaccine challenges, including those related to infrastructures, transportation, possible adverse reactions, utilized delivery systems (e.g., nanotechnology and electroporation) and probable vaccine-induced mutations.
Collapse
Affiliation(s)
- Gelareh Abdolmaleki
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Mina Azam Taheri
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Sarina Paridehpour
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Neshaut Mashreghi Mohammadi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Ahmadi Tabatabaei
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Taraneh Mousavi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran.
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
56
|
Luo WR, Wu XM, Wang W, Yu JL, Chen QQ, Zhou X, Huang X, Pan HF, Liu ZR, Gao Y, He J. Novel coronavirus mutations: Vaccine development and challenges. Microb Pathog 2022; 173:105828. [PMID: 36243381 PMCID: PMC9561474 DOI: 10.1016/j.micpath.2022.105828] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The ongoing global pandemic of novel coronavirus pneumonia (COVID-19) caused by the SARS-CoV-2 has a significant impact on global health and economy system. In this context, there have been some landmark advances in vaccine development. Over 100 new coronavirus vaccine candidates have been approved for clinical trials, with ten WHO-approved vaccines including four inactivated virus vaccines, two mRNA vaccines, three recombinant viral vectored vaccines and one protein subunit vaccine on the "Emergency Use Listing". Although the SARS-CoV-2 has an internal proofreading mechanism, there have been a number of mutations emerged in the pandemic affecting its transmissibility, pathogenicity and immunogenicity. Of these, mutations in the spike (S) protein and the resultant mutant variants have posed new challenges for vaccine development and application. In this review article, we present an overview of vaccine development, the prevalence of new coronavirus variants and their impact on protective efficacy of existing vaccines and possible immunization strategies coping with the viral mutation and diversity.
Collapse
Affiliation(s)
- Wan-Rong Luo
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Xiao-Min Wu
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Wei Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), 17 Lujiang Road, Hefei, Anhui, China
| | - Jun-Ling Yu
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Qing-Qing Chen
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Xue Zhou
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Xin'er Huang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Zhi-Rong Liu
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China; Department of Microbiology Laboratory, Public Health Research Institute of Anhui Province, 12560, Fanhua Avenue, Hefei, Anhui, China.
| | - Yong Gao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), 17 Lujiang Road, Hefei, Anhui, China.
| | - Jun He
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China; Department of Microbiology Laboratory, Public Health Research Institute of Anhui Province, 12560, Fanhua Avenue, Hefei, Anhui, China; Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
57
|
Fatouh MM, Kandil NM, EL Garhy NM. Correlation between chest CT scores and clinical impact in patients re-infected with COVID-19 during the two attacks: an observational study. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2022. [PMCID: PMC8900098 DOI: 10.1186/s43055-022-00739-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Worldwide, millions of people got COVID-19 infection since the start of the pandemic with a large number of deaths. Re-infection with SARS-CoV-2 is possible, because it can mutate into new strains as it is an RNA virus. The main objective of our study is to correlate between CT severity score of the patients re-infected with COVID-19 during the first and second attack and its clinical impact.
Results We performed a retrospective cohort study. It was carried out on fifty symptomatic patients (11 females and 39 males). Their ages ranged from 38 to 71 years. We included only patients who were re-infected after more than 6 months of the first infection and showed clinical symptoms with SARS-CoV-2 PCR-positive test. We found that CT severity score was decreased in the second infection in 47 (94%) of our patients associated with decreased respiratory distress as well as oxygen requirements, while the CT severity score was increased in two patients and only one patient showed no change in CT score severity between two infections. Conclusion The reduction in CT severity score in the majority of re-infected patients suggested the role of the immunity developed from first infection in protection against severe lung affection in case of repeated infection even after 6 months despite poor immunity against re-infection.
Collapse
|
58
|
Turkkan A, Saglik I, Turan C, Sahin A, Akalin H, Ener B, Kara A, Celebi S, Sahin E, Hacimustafaoglu M. Nine-month course of SARS-CoV-2 antibodies in individuals with COVID-19 infection. Ir J Med Sci 2022; 191:2803-2811. [PMID: 35048229 PMCID: PMC8769943 DOI: 10.1007/s11845-021-02716-x] [Citation(s) in RCA: 4] [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: 04/01/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The continual course of the pandemic points to the importance of studies on the rate and durability of protective immunity after infection or vaccination. AIMS In this study, we aimed to monitor anti-nucleocapsid (N) and anti-spike (S) antibodies against SARS-CoV-2 nearly 9 months duration after infection. METHODS Anti-nucleocapsid (N) (at 11-15-20-29-38 weeks) and anti-spike antibodies (at 11 and 38 weeks) against SARS-CoV-2 were monitored during 38 weeks after the initial symptoms of COVID-19. RESULTS Of 37 cases between 18 and 57 years old, 54% were women. The findings showed that anti-N antibodies decreased significantly after the 15th week (between 15 and 20 weeks, p = 0.016; 20-29 weeks, p = 0.0009; and 29-38 weeks, p = 0.049). At the 38th week, mean antibody levels decreased 35% compared to the 11th week, and 8% of the cases turned negative results. Anti-N antibody average level was 56.48 on the 11th week (the cut-off index threshold ≥ 1). It was estimated statistically that it would decrease to an average of 20.48 in weeks 53-62. In females, average antibody levels of all measurements were lower than males (p > 0.05). Anti-S antibody levels 14% increased at 38th week compared to 11th week (quantitative positivity threshold ≥ 0.8 U/ml), and no cases were negative at 38th week. CONCLUSIONS Patients had ≥ 90% positivity after at least 9 months of symptoms, both anti-N and anti-S antibodies. In all samples, both anti-N and anti-S antibody levels were lower in females. The findings suggest that the quantitative values of anti-S antibodies remained high for at least 9 months and could provide protection.
Collapse
Affiliation(s)
- Alpaslan Turkkan
- Department of Public Health, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Imran Saglik
- Department of Medical Microbiology, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Cansu Turan
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Ahmet Sahin
- Biochemistry and Clinical Biochemistry, Guven Tip Laboratuarı, Bursa, Turkey
| | - Halis Akalin
- Department of Infectious Diseases and Clinical Microbiology, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Beyza Ener
- Department of Medical Microbiology, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Ates Kara
- Department of Pediatric Infectious Diseases, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Solmaz Celebi
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Emre Sahin
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Mustafa Hacimustafaoglu
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey.
| |
Collapse
|
59
|
Primary SARS-CoV-2 Infections, Re-infections and Vaccine Effectiveness during the Omicron Transmission Period in Healthcare Workers of Trieste and Gorizia (Northeast Italy), 1 December 2021-31 May 2022. Viruses 2022; 14:v14122688. [PMID: 36560692 PMCID: PMC9784665 DOI: 10.3390/v14122688] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Objective: To evaluate the incidence of primary and recurrent COVID-19 infections in healthcare workers (HCWs) routinely screened for SARS-CoV-2 by nasopharyngeal swabs during the Omicron wave. Design: Dynamic Cohort study of HCWs (N = 7723) of the University Health Agency Giuliano Isontina (ASUGI), covering health services of the provinces of Trieste and Gorizia (Northeast Italy). Cox proportional hazard model was employed to estimate the risk of primary as well as recurrent SARS-CoV-2 infection from 1 December 2021 through 31 May 2022, adjusting for a number of confounding factors. Results: By 1 December 2021, 46.8% HCWs of ASUGI had received the booster, 37.2% were immunized only with two doses of COVID-19 vaccines, 6.0% only with one dose and 10.0% were unvaccinated. During 1 March 2020-31 May 2022, 3571 primary against 406 SARS-CoV-2 recurrent infections were counted among HCWs of ASUGI, 59.7% (=2130/3571) versus 95.1% (=386/406) of which occurring from 1 December 2021 through 31 May 2022, respectively. All HCWs infected by SARS-CoV-2 during 1 December 2021 through 31 May 2022 presented mild flu-like disease. Compared to staff working in administrative services, the risk of primary as well as recurrent SARS-CoV-2 infection increased in HCWs with patient-facing clinical tasks (especially nurses and other categories of HCWs) and in all clinical wards but COVID-19 units and community health services. Regardless of the number of swab tests performed during the study period, primary infections were less likely in HCWs immunized with one dose of COVID-19 vaccine. By contrast, the risk of SARS-CoV-2 re-infection was significantly lower in HCWs immunized with three doses (aHR = 0.58; 95%CI: 0.41; 0.80). During the study period, vaccine effectiveness (VE = 1-aHR) of the booster dose declined to 42% against re-infections, vanishing against primary SARS-CoV-2 infections. Conclusions: Though generally mild, SARS-CoV-2 infections and re-infections surged during the Omicron transmission period. Compared to unvaccinated colleagues, the risk of primary SARS-CoV-2 infection was significantly lower in HCWs immunized just with one dose of COVID-19 vaccines. By Italian law, HCWs immunized only with one dose were either suspended or re-assigned to job tasks not entailing patient facing contact; hence, while sharing the same biological risk of unvaccinated colleagues, they arguably had a higher level of protection against COVID-19 infection. By contrast, SARS-CoV-2 re-infections were less likely in HCWs vaccinated with three doses, suggesting that hybrid humoral immunity by vaccination combined with natural infection provided a higher level of protection than vaccination only. In this stage of the pandemic, where SARS-CoV-2 is more infectious yet much less pathogenic, health protection measures in healthcare premises at higher biological risk seem the rational approach to control the transmission of the virus.
Collapse
|
60
|
Hill HJ, Uppal T, Hau D, Pandit SG, Arias-Umana J, Foster AJ, Gorzalski A, Pflughoeft KJ, Burnham-Marusich AR, Reed DE, Gates-Hollingsworth MA, Gumbleton L, Verma SC, AuCoin DP. Comparison of a Prototype SARS-CoV-2 Lateral Flow IMMUNOASSAY with the BinaxNOW TM COVID-19 Antigen CARD. Viruses 2022; 14:v14122609. [PMID: 36560613 PMCID: PMC9786212 DOI: 10.3390/v14122609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for the COVID-19 pandemic. From the onset of the pandemic, rapid antigen tests have quickly proved themselves to be an accurate and accessible diagnostic platform. The initial (and still most commonly used antigen tests) for COVID-19 diagnosis were constructed using monoclonal antibodies (mAbs) specific to severe acute respiratory syndrome coronavirus (SARS-CoV) nucleocapsid protein (NP). These mAbs are able to bind SARS-CoV-2 NP due to high homology between the two viruses. However, since first being identified in 2019, SARS-CoV-2 has continuously mutated, and a multitude of variants have appeared. These mutations have an elevated risk of leading to possible diagnostic escape when using tests produced with SARS-CoV-derived mAbs. Here, we established a library of 18 mAbs specific to SARS-CoV-2 NP and used two of these mAbs (1CV7 and 1CV14) to generate a prototype antigen-detection lateral flow immunoassay (LFI). A side-by-side analysis of the 1CV7/1CV14 LFI and the commercially available BinaxNOWTM COVID-19 Antigen CARD was performed. Results indicated the 1CV7/1CV14 LFI outperformed the BinaxNOWTM test in the detection of BA.2, BA.2.12.1, and BA.5 Omicron sub-variants when testing remnant RT-PCR positive patient nasopharyngeal swabs diluted in viral transport media.
Collapse
Affiliation(s)
- Haydon J. Hill
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Timsy Uppal
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Derrick Hau
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Sujata G. Pandit
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Jose Arias-Umana
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Abigail J. Foster
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | | | | | | | - Dana E. Reed
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | | | | | - Subhash C. Verma
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - David P. AuCoin
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
- DxDiscovery Incorporated, Reno, NV 89557, USA
- Correspondence:
| |
Collapse
|
61
|
Piazza MF, Amicizia D, Marchini F, Astengo M, Grammatico F, Battaglini A, Sticchi C, Paganino C, Lavieri R, Andreoli GB, Orsi A, Icardi G, Ansaldi F. Who Is at Higher Risk of SARS-CoV-2 Reinfection? Results from a Northern Region of Italy. Vaccines (Basel) 2022; 10:vaccines10111885. [PMID: 36366393 PMCID: PMC9692964 DOI: 10.3390/vaccines10111885] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
The SARS-CoV-2 pandemic continues to spread worldwide, generating a high impact on healthcare systems. The aim of the study was to examine the epidemiological burden of SARS-CoV-2 reinfections and to identify potential related risk factors. A retrospective observational study was conducted in Liguria Region, combining data from National Vaccines Registry and Regional Chronic Condition Data Warehouse. In the study period (September 2021 to May 2022), 335,117 cases of SARS-CoV-2 infection were recorded in Liguria, of which 15,715 were reinfected once. During the Omicron phase (which predominated from 3 January 2022), the risk of reinfection was 4.89 times higher (p < 0.001) than during the Delta phase. Unvaccinated and vaccinated individuals with at least one dose for more than 120 days were at increased risk of reinfection compared with vaccinated individuals with at least one dose for ≤120 days, respectively (odds ratio (OR) of 1.26, p < 0.001; OR of 1.18, p < 0.001). Healthcare workers were more than twice as likely to be reinfected than non-healthcare workers (OR of 2.38, p < 0.001). Lower ORs were seen among people aged 60 to 79 years. Two doses or more of vaccination were found to be protective against the risk of reinfection rather than a single dose (mRNA vaccines: OR of 0.06, p < 0.0001, and OR of 0.1, p < 0.0001; vector vaccines: OR of 0.05, p < 0.0001). Patients with chronic renal failure, cardiovascular disease, bronchopneumopathy, neuropathy and autoimmune diseases were at increased risk of reinfection (OR of 1.38, p = 0.0003; OR of 1.09, p < 0.0296; OR of 1.14, p = 0.0056; OR of 1.78, p < 0.0001; OR of 1.18, p = 0.0205). Estimating the epidemiological burden of SARS-CoV-2 reinfections and the role played by risk factors in reinfections is relevant for identifying risk-based preventive strategies in a pandemic context characterized by a high circulation of the virus and a high rate of pathogen mutations.
Collapse
Affiliation(s)
- Maria Francesca Piazza
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
- Correspondence: ; Tel.: +39-010-548-4680
| | - Daniela Amicizia
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
- Department of Health Sciences (DiSSal), University of Genoa, 16132 Genoa, Italy
| | - Francesca Marchini
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
- Department of Health Sciences (DiSSal), University of Genoa, 16132 Genoa, Italy
| | - Matteo Astengo
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
| | - Federico Grammatico
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
- Department of Health Sciences (DiSSal), University of Genoa, 16132 Genoa, Italy
| | - Alberto Battaglini
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
- Department of Health Sciences (DiSSal), University of Genoa, 16132 Genoa, Italy
| | - Camilla Sticchi
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
| | - Chiara Paganino
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
| | - Rosa Lavieri
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
| | | | - Andrea Orsi
- Department of Health Sciences (DiSSal), University of Genoa, 16132 Genoa, Italy
- Hygiene Unit, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy
| | - Giancarlo Icardi
- Department of Health Sciences (DiSSal), University of Genoa, 16132 Genoa, Italy
- Hygiene Unit, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy
| | - Filippo Ansaldi
- Regional Health Agency of Liguria (ALiSa), 16121 Genoa, Italy
- Department of Health Sciences (DiSSal), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
62
|
Romeo I, Ambrosio FA, Costa G, Corona A, Alkhatib M, Salpini R, Lemme S, Vergni D, Svicher V, Santoro MM, Tramontano E, Ceccherini-Silberstein F, Artese A, Alcaro S. Targeting SARS-CoV-2 nsp13 Helicase and Assessment of Druggability Pockets: Identification of Two Potent Inhibitors by a Multi-Site In Silico Drug Repurposing Approach. Molecules 2022; 27:7522. [PMID: 36364347 PMCID: PMC9654784 DOI: 10.3390/molecules27217522] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 06/14/2024] Open
Abstract
The SARS-CoV-2 non-structural protein 13 (nsp13) helicase is an essential enzyme for viral replication and has been identified as an attractive target for the development of new antiviral drugs. In detail, the helicase catalyzes the unwinding of double-stranded DNA or RNA in a 5' to 3' direction and acts in concert with the replication-transcription complex (nsp7/nsp8/nsp12). In this work, bioinformatics and computational tools allowed us to perform a detailed conservation analysis of the SARS-CoV-2 helicase genome and to further predict the druggable enzyme's binding pockets. Thus, a structure-based virtual screening was used to identify valuable compounds that are capable of recognizing multiple nsp13 pockets. Starting from a database of around 4000 drugs already approved by the Food and Drug Administration (FDA), we chose 14 shared compounds capable of recognizing three out of four sites. Finally, by means of visual inspection analysis and based on their commercial availability, five promising compounds were submitted to in vitro assays. Among them, PF-03715455 was able to block both the unwinding and NTPase activities of nsp13 in a micromolar range.
Collapse
Affiliation(s)
- Isabella Romeo
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Francesca Alessandra Ambrosio
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, 09124 Cagliari, Italy
| | - Mohammad Alkhatib
- Dipartimento di Medicina Sperimentale, Università Tor Vergata di Roma, Via Montpellier, 1, 00133 Roma, Italy
| | - Romina Salpini
- Dipartimento di Medicina Sperimentale, Università Tor Vergata di Roma, Via Montpellier, 1, 00133 Roma, Italy
| | - Saverio Lemme
- Dipartimento di Medicina Sperimentale, Università Tor Vergata di Roma, Via Montpellier, 1, 00133 Roma, Italy
| | - Davide Vergni
- Istituto per le Applicazioni del Calcolo “Mauro Picone”-CNR, 00185 Rome, Italy
| | - Valentina Svicher
- Dipartimento di Medicina Sperimentale, Università Tor Vergata di Roma, Via Montpellier, 1, 00133 Roma, Italy
| | - Maria Mercedes Santoro
- Dipartimento di Medicina Sperimentale, Università Tor Vergata di Roma, Via Montpellier, 1, 00133 Roma, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, 09124 Cagliari, Italy
| | | | - Anna Artese
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| |
Collapse
|
63
|
Bowe B, Xie Y, Al-Aly Z. Acute and postacute sequelae associated with SARS-CoV-2 reinfection. Nat Med 2022; 28:2398-2405. [PMID: 36357676 PMCID: PMC9671810 DOI: 10.1038/s41591-022-02051-3] [Citation(s) in RCA: 322] [Impact Index Per Article: 107.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/23/2022] [Indexed: 11/12/2022]
Abstract
First infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is associated with increased risk of acute and postacute death and sequelae in various organ systems. Whether reinfection adds to risks incurred after first infection is unclear. Here we used the US Department of Veterans Affairs' national healthcare database to build a cohort of individuals with one SARS-CoV-2 infection (n = 443,588), reinfection (two or more infections, n = 40,947) and a noninfected control (n = 5,334,729). We used inverse probability-weighted survival models to estimate risks and 6-month burdens of death, hospitalization and incident sequelae. Compared to no reinfection, reinfection contributed additional risks of death (hazard ratio (HR) = 2.17, 95% confidence intervals (CI) 1.93-2.45), hospitalization (HR = 3.32, 95% CI 3.13-3.51) and sequelae including pulmonary, cardiovascular, hematological, diabetes, gastrointestinal, kidney, mental health, musculoskeletal and neurological disorders. The risks were evident regardless of vaccination status. The risks were most pronounced in the acute phase but persisted in the postacute phase at 6 months. Compared to noninfected controls, cumulative risks and burdens of repeat infection increased according to the number of infections. Limitations included a cohort of mostly white males. The evidence shows that reinfection further increases risks of death, hospitalization and sequelae in multiple organ systems in the acute and postacute phase. Reducing overall burden of death and disease due to SARS-CoV-2 will require strategies for reinfection prevention.
Collapse
Affiliation(s)
- Benjamin Bowe
- Clinical Epidemiology Center, Research and Development Service, Veteran Affairs Saint Louis Health Care System, St. Louis, MO, USA
- Veterans Research and Education Foundation of St. Louis, St. Louis, MO, USA
| | - Yan Xie
- Clinical Epidemiology Center, Research and Development Service, Veteran Affairs Saint Louis Health Care System, St. Louis, MO, USA
- Veterans Research and Education Foundation of St. Louis, St. Louis, MO, USA
| | - Ziyad Al-Aly
- Clinical Epidemiology Center, Research and Development Service, Veteran Affairs Saint Louis Health Care System, St. Louis, MO, USA.
- Veterans Research and Education Foundation of St. Louis, St. Louis, MO, USA.
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Nephrology Section, Medicine Service, Veteran Affairs St. Louis Health Care System, St. Louis, MO, USA.
- Institute for Public Health, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
64
|
Yang B, Zeng X, Zhang J, Kong J, Fang X. Accurate identification of SARS-CoV-2 variant delta using graphene/CRISPR-dCas9 electrochemical biosensor. Talanta 2022; 249:123687. [PMID: 35724556 PMCID: PMC9197783 DOI: 10.1016/j.talanta.2022.123687] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 12/25/2022]
Abstract
Delta (B.1.617.2), a highly infectious variant of SARS-CoV-2, has been sweeping the world, and threatening the safety of human life seriously. It is urgent to develop a highly selective and sensitive assay to accurately identify the SARS-CoV-2 variant Delta. In this work, we constructed a graphene/CRISPR-dCas9 electrochemical biosensor to accurately identify SARS-CoV-2 variant Delta, where the signal was further amplified by embedded electrochemical probe [Ru(phen)2dppz]BF4. This detection assay could be finished within 47 min totally, with the detection limit of 1.2 pM and good reproducibility with a C·V.% of 2.48% (n = 5). And the biosensor could selectively identify Delta among SARS-CoV-2 and other variants, including Alpha, Beta, Gamma. This assay was further validated by 26 real clinical samples, showing 100% clinical sensitivity and 100% clinical specificity, which provides a new direction for identifying other SARS-CoV-2 variants in the future.
Collapse
Affiliation(s)
- Bin Yang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, PR China
| | - Xiaowei Zeng
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, PR China
| | - Jin Zhang
- Qingdao International Travel Healthcare Center, Qingdao Customs, Qingdao, 266071, PR China
| | - Jilie Kong
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, PR China,Corresponding author
| | - Xueen Fang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, PR China,Corresponding author
| |
Collapse
|
65
|
A retrospective cross-sectional observational study of SARS-CoV-2 reinfection in La Ribera Health Department, Valencia, Spain. J Med Microbiol 2022; 71. [DOI: 10.1099/jmm.0.001599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. The possibility of reinfection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a widely proven fact and may have clinical implications.
Hypothesis /Gap Statement. It is not known whether there have been cases of reinfection by SARS-CoV-2 in La Ribera Health Department.
Aim. To determine whether there have been cases of reinfection by SARS-CoV-2 in La Ribera Health Department and to identify their characteristics.
Methodology. Retrospective cross-sectional observational study of cases of reinfection by SARS-CoV-2 in the population of La Ribera Department between March 2020 and February 2021. The positive baseline cohort includes all cases positive by RT-PCR for SARS-CoV-2, with reinfection cases being those that, after resolution of the first episode according to the World Health Organization (WHO) criteria, presented a new positive RT-PCR result.
Results. Out of a total of 15 687 cases with positive RT-PCR, 40 were considered to be reinfections, which meant a cumulative incidence of 0.255 % and an incidence density of 5.05 cases per 100 000 person-days. Most of the cases occurred during the highest incidence peaks of the pandemic in the department. Seventy-five per cent of the patients in these cases were older than 40 years, 42.5 % were healthcare professionals or nursing home residents and 12.5 % had an immunosuppressive comorbidity. There were no severe, critical or death cases. In the reinfection episodes, with respect to the first episode, there was a tendency to be milder, they required fewer days of hospitalization, their RT-PCR became negative earlier, they developed a greater humoral response and the sick leave period was shorter. The median period between the RT-PCR in the first episode and the RT-PCR in the second episode was 127.5 days (range: 48–301; IQR: 89.5–256.25)
Conclusions. SARS-CoV-2 reinfection cases are rare, tend to be mild and can occur within a median period of 127.5 days.
Collapse
|
66
|
Gouvea MDPG, Moulaz IR, Gouveia TM, Lança KEM, Lacerda BSDP, Thompson BP, Polese J, de Lima MD, Ribeiro-Rodrigues R, Mill JG, Valim V. Anti-SARS-CoV-2 antibody immunoreactivity profiles during COVID-19 recurrence. Rev Soc Bras Med Trop 2022; 55:e01062022. [PMID: 36287469 PMCID: PMC9592100 DOI: 10.1590/0037-8682-0106-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/09/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND This study aimed to evaluate IgG and IgM levels in COVID-19 recurrence. METHODS The serum antibody levels and clinical data from 73 healthcare workers with SARS-CoV-2 divided into seroconverted (n=51) and non-seroconverted (n=22) groups were assessed. The presence of specific anti-nucleocapsid (anti-N) IgM and IgG for SARS-CoV-2 was evaluated. IgG antibodies to the SARS-CoV-2 spike receptor-binding domain were used to confirm non-seroconversion in all negative anti-N. RESULTS Four recurrent cases displayed mild symptoms and were non-seroconverted until the recurrence of symptoms. CONCLUSIONS Undetectable anti-nucleocapsid IgM and IgG levels may be correlated with symptomatic COVID-19 recurrence.
Collapse
Affiliation(s)
| | - Isac Ribeiro Moulaz
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | - Thayná Martins Gouveia
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | | | | | - Beatriz Paoli Thompson
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | - Jéssica Polese
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Pneumologia, Vitória, ES, Brasil
| | - Marina Deorce de Lima
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | - Rodrigo Ribeiro-Rodrigues
- Departamento de Saúde do Estado do Espírito Santo, Laboratório de Saúde Pública do Estado do Espírito Santo, Vitória, ES, Brasil., Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Centro de Doenças Infecciosas, Vitória, ES, Brasil
| | - José Geraldo Mill
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Ciências Fisiológicas, Vitória, ES, Brasil
| | - Valéria Valim
- Universidade Federal do Espírito Santo, Hospital Universitário Cassiano Antonio Moraes, Divisão de Reumatologia, Vitória, ES, Brasil
| |
Collapse
|
67
|
Ruff J, Zhang Y, Kappel M, Rathi S, Watkins K, Zhang L, Lockett C. Rapid Increase in Suspected SARS-CoV-2 Reinfections, Clark County, Nevada, USA, December 2021. Emerg Infect Dis 2022; 28:1977-1981. [PMID: 35969420 PMCID: PMC9514365 DOI: 10.3201/eid2810.221045] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Genetic differences between SARS-CoV-2 variants raise concerns about reinfection. Public health authorities monitored the incidence of suspected reinfection in Clark County, Nevada, USA, during March 2020-March 2022. Suspected reinfections, defined as a second positive PCR test collected >90 days after an initial positive test, were monitored through an electronic disease surveillance system. We calculated the proportion of all new cases per week that were suspected reinfections and rates per 1,000 previously infected persons by demographic groups. The rate of suspected reinfection remained <2.7% until December 2021, then increased to ≈11%, corresponding with local Omicron variant detection. Reinfection rates were higher among adults 18-50 years of age, women, and minority groups, especially persons identifying as American Indian/Alaska Native. Suspected reinfection became more common in Clark County after introduction of the Omicron variant, and some demographic groups are disproportionately affected. Public health surveillance could clarify the SARS-CoV-2 reinfection burden in communities.
Collapse
|
68
|
Almadhi M, Alsayyad AS, Conroy R, Atkin S, Awadhi AA, Al-Tawfiq JA, AlQahtani M. Epidemiological assessment of SARS-CoV-2 reinfection. Int J Infect Dis 2022; 123:9-16. [PMID: 35931371 PMCID: PMC9345650 DOI: 10.1016/j.ijid.2022.07.075] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES SARS-CoV-2 vaccination has been shown to reduce infection severity; however, the reinfection frequency among unvaccinated, partially vaccinated, and fully vaccinated individuals remains unclear. This study aims to elucidate the rates of and factors associated with such occurrences. METHODS This retrospective epidemiological report included 1362 COVID-19 reinfection cases in Bahrain between April 2020 and July 2021. We analyzed differences in disease severity and reinfection characteristics among various vaccination statuses: fully vaccinated, interrupted vaccination, one-dose vaccination, postreinfection vaccination, and unvaccinated. RESULTS Reinfection cases increased from zero per month in April-June 2020 to a sharp peak of 579 in May 2021. A significantly larger proportion of reinfected individuals were male (60.3%, P <0.0001). Reinfection episodes were highest among those 30-39 years of age (29.7%). The fewest reinfection episodes occurred at 3-6 months after the first infection (20.6%) and most occurred ≥9 months after the initial infection (46.4%). Most individuals were asymptomatic during both episodes (35.7%). Reinfection disease severity was mild, with vaccinated patients less likely to have symptomatic reinfection (odds ratio 0.71, P = 0.004). Only 6.6% of reinfected patients required hospitalization. One death was recorded; the patient belonged to the unvaccinated group. CONCLUSION Vaccine-induced immunity and previous infection with or without vaccination were effective in reducing reinfection disease severity.
Collapse
Affiliation(s)
- Marwa Almadhi
- National Taskforce for Combating the Coronavirus (COVID-19), Supreme health council, Manama, Bahrain; University of Manchester, Manchester, UK
| | - Adel Salman Alsayyad
- Department of Family and Community Medicine, Arabian Gulf University, Manama, Bahrain
| | - Ronan Conroy
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stephen Atkin
- Royal College of Surgeons in Ireland, Busaiteen, Bahrain
| | - Abdulla Al Awadhi
- National Taskforce for Combating the Coronavirus (COVID-19), Supreme health council, Manama, Bahrain; Bahrain Defence Force Hospital, Riffa, Bahrain
| | - Jaffar A Al-Tawfiq
- Infectious Disease Unit, Specialty Internal Medicine, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia; Division of Infectious Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA; Division of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland, USA
| | - Manaf AlQahtani
- National Taskforce for Combating the Coronavirus (COVID-19), Supreme health council, Manama, Bahrain; Royal College of Surgeons in Ireland, Busaiteen, Bahrain; Bahrain Defence Force Hospital, Riffa, Bahrain.
| |
Collapse
|
69
|
Ahmed N, Abbas K, Makhoul E, Deeb R, Raya A, Khaddour N, Redwan F. Recurrence of COVID-19 infection symptoms in short time; reinfection or reactivation? Three cases of three healthcare workers and a literature review. Ann Med Surg (Lond) 2022; 82:104619. [PMID: 36117528 PMCID: PMC9467930 DOI: 10.1016/j.amsu.2022.104619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/03/2022] [Accepted: 09/04/2022] [Indexed: 12/02/2022] Open
Abstract
Background Since the beginning of the COVID-19 pandemic, many research papers have been published focusing on some recurrence cases of symptoms after a long period of free symptoms with a negative RT-PCR retest. There is no crucial evidence until now of the possibility of recurrence, immune system reactivation, or reinfection. Methods Three cases of resident doctors who recovered from COVID-19 but represented symptoms with new positive RT-PCR were discussed. Clinical data, laboratory tests, RT-PCR results, and antibodies titers all were collected. Moreover, many cases from the literature have been reviewed and compared. Results The long-term exposure has not succeeded in forming an effective immune response, especially, since they do not have any significant history of chronic illnesses or a diagnosed immune disorder. While the antibody response occurred only in the second patient, it did not prevent new infection, but did it control the severity of the infection or its complications? Conclusion Our three patients are health workers and have been in direct contact with COVID-19 patients. The inflammatory response parameters may not be reliable in predicting the activation of the immune response and the formation of the antibodies. We still need to find answers for reactivation and reinfection issues. A direct contact with COVID-19 patients failed in forming an effective response. The antibodies controlled the severity, complications and inflammatory response. The correlation between the severity of symptoms and laboratory markers.
Collapse
Affiliation(s)
- Nadeem Ahmed
- Department of Cardiovascular Diseases, Tishreen University Hospital, Latakia, Syria
| | - Kinan Abbas
- Department of Gastrointestinal Diseases, Tishreen University Hospital, Latakia, Syria
| | - Ebrahim Makhoul
- Tishreen University Hospital, Latakia, Syria
- Corresponding author.
| | - Rudy Deeb
- Department of Neurology, Tishreen University Hospital, Latakia, Syria
| | - Ashraf Raya
- Department of Gastrointestinal Diseases, Tishreen University Hospital, Latakia, Syria
| | - Nagham Khaddour
- Department of Internal Medicine, Tishreen University Hospital, Latakia, Syria
| | - Faisal Redwan
- Head of Laboratory Medicine Department and Assistant Professor of Clinical Biochemistry, Tishreen University, Latakia, Syria
| |
Collapse
|
70
|
Daw MA, Miftah MM, El-Bouzedi A, Ahmed MO. Familial Clustering and Reinfection With 2019 Novel Coronavirus (COVID-19, SARS-CoV-2) in the Libyan Community. Disaster Med Public Health Prep 2022; 16:1710-1712. [PMID: 33678217 PMCID: PMC8134895 DOI: 10.1017/dmp.2021.68] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/25/2021] [Indexed: 11/07/2022]
Affiliation(s)
- Mohamed A. Daw
- Department of Medical Microbiology & Immunology, Faculty of Medicine, University of Tripoli, Tripoli, Libya
| | | | - Abdallah El-Bouzedi
- Department of Laboratory Medicine, Faculty of Biotechnology, University of Tripoli, Tripoli, Libya
| | - Mohamed O. Ahmed
- Department of Microbiology & Parasitology, Faculty of Veterinary Medicine, University of Tripoli, Libya
| |
Collapse
|
71
|
Lueking R, Clark AE, Narasimhan M, Mahimainathan L, Muthukumar A, Larsen CP, SoRelle JA. SARS-CoV-2 coinfections with variant genomic lineages identified by multiplex fragment analysis. Front Genet 2022; 13:942713. [PMID: 36226173 PMCID: PMC9549124 DOI: 10.3389/fgene.2022.942713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Immunocompromised patients can experience prolonged SARS-CoV-2 infections in the setting of a lack of protectivity immunity despite vaccination. As circulating SARS-CoV-2 strains become more heterogeneous, concomitant infection with multiple SARS-CoV-2 variants has become an increasing concern. Immunocompromised patient populations represent potential reservoirs for the emergence of novel SARS-CoV-2 variants through mutagenic change or coinfection followed by recombinatory events. Identification of SARS-CoV-2 coinfections is challenging using traditional next generation sequencing pipelines; however, targeted genotyping approaches can facilitate detection. Here we describe five COVID-19 cases caused by coinfection with different SARS-CoV-2 variants (Delta/Omicron BA.1 and Omicron BA.1/BA.2) as identified by multiplex fragment analysis.
Collapse
Affiliation(s)
- Richard Lueking
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Andrew E. Clark
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Madhusudhanan Narasimhan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Lenin Mahimainathan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Alagarraju Muthukumar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Christian P. Larsen
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jeffrey A. SoRelle
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
72
|
Gan L, Chen Y, Tan J, Wang X, Zhang D. Does potential antibody-dependent enhancement occur during SARS-CoV-2 infection after natural infection or vaccination? A meta-analysis. BMC Infect Dis 2022; 22:742. [PMID: 36123623 PMCID: PMC9483537 DOI: 10.1186/s12879-022-07735-2] [Citation(s) in RCA: 9] [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: 01/07/2022] [Accepted: 09/09/2022] [Indexed: 12/05/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) continues to constitute an international public health emergency. Vaccination is a prospective approach to control this pandemic. However, apprehension about the safety of vaccines is a major obstacle to vaccination. Amongst health professionals, one evident concern is the risk of antibody-dependent enhancement (ADE), which may increase the severity of COVID-19. To explore whether ADE occurs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and increase confidence in the safety of vaccination, we conducted a meta-analysis to investigate the relationship between post-immune infection and disease severity from a population perspective. Databases, including PubMed, EMBASE, Chinese National Knowledge Infrastructure, SinoMed, Scopus, Science Direct, and Cochrane Library, were searched for articles on SARS-CoV-2 reinfection published until 25 October 2021. The papers were reviewed for methodological quality, and a random effects model was used to analyse the results. Heterogeneity was assessed using the I2 statistic. Publication bias was evaluated using a funnel plot and Egger's test. Eleven studies were included in the final meta-analysis. The pooled results indicated that initial infection and vaccination were protective factors against severe COVID-19 during post-immune infection (OR = 0.55, 95%CI = 0.31-0.98). A subgroup (post-immune infection after natural infection or vaccination) analysis showed similar results. Primary SARS-CoV-2 infection and vaccination provide adequate protection against severe clinical symptoms after post-immune infection. This finding demonstrates that SARS-CoV-2 may not trigger ADE at the population level.
Collapse
Affiliation(s)
- Lin Gan
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Yan Chen
- Medical College of Shaoguan University, Shaoguan, 512000, Guangdong, China
| | - Jinlin Tan
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Xuezhi Wang
- Foshan No.4 People's Hospital, Foshan, 528000, Guangdong, China
| | - Dingmei Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
73
|
Lang J. NanoCoV19: An analytical pipeline for rapid detection of severe acute respiratory syndrome coronavirus 2. Front Genet 2022; 13:1008792. [PMID: 36186464 PMCID: PMC9520466 DOI: 10.3389/fgene.2022.1008792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Nanopore sequencing technology (NST) has become a rapid and cost-effective method for the diagnosis and epidemiological surveillance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) during the coronavirus disease 2019 (COVID-19) pandemic. Compared with short-read sequencing platforms (e.g., Illumina's), nanopore long-read sequencing platforms effectively shorten the time required to complete the detection process. However, due to the principles and data characteristics of NST, the accuracy of sequencing data has been reduced, thereby limiting monitoring and lineage analysis of SARS-CoV-2. In this study, we developed an analytical pipeline for SARS-CoV-2 rapid detection and lineage identification that integrates phylogenetic-tree and hotspot mutation analysis, which we have named NanoCoV19. This method not only can distinguish and trace the lineages contained in the alpha, beta, delta, gamma, lambda, and omicron variants of SARS-CoV-2 but is also rapid and efficient, completing overall analysis within 1 h. We hope that NanoCoV19 can be used as an auxiliary tool for rapid subtyping and lineage analysis of SARS-CoV-2 and, more importantly, that it can promote further applications of NST in public-health and -safety plans similar to those formulated to address the COVID-19 outbreak.
Collapse
Affiliation(s)
- Jidong Lang
- Department of Bioinformatics, Qitan Technology (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
74
|
Marinov GK, Mladenov M, Rangachev A, Alexiev I. SARS-CoV-2 reinfections during the first three major COVID-19 waves in Bulgaria. PLoS One 2022; 17:e0274509. [PMID: 36084070 PMCID: PMC9462809 DOI: 10.1371/journal.pone.0274509] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/30/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The COVID-19 pandemic has had a devastating impact on the world over the past two years (2020-2021). One of the key questions about its future trajectory is the protection from subsequent infections and disease conferred by a previous infection, as the SARS-CoV-2 virus belongs to the coronaviruses, a group of viruses the members of which are known for their ability to reinfect convalescent individuals. Bulgaria, with high rates of previous infections combined with low vaccination rates and an elderly population, presents a somewhat unique context to study this question. METHODS We use detailed governmental data on registered COVID-19 cases to evaluate the incidence and outcomes of COVID-19 reinfections in Bulgaria in the period between March 2020 and early December 2021. RESULTS For the period analyzed, a total of 4,106 cases of individuals infected more than once were observed, including 31 cases of three infections and one of four infections. The number of reinfections increased dramatically during the Delta variant-driven wave of the pandemic towards the end of 2021. We observe a moderate reduction of severe outcomes (hospitalization and death) in reinfections relative to primary infections, and a more substantial reduction of severe outcomes in breakthrough infections in vaccinated individuals. CONCLUSIONS In the available datasets from Bulgaria, prior infection appears to provide some protection from severe outcomes, but to a lower degree than the reduction in severity of breakthrough infections in the vaccinated compared to primary infections in the unvaccinated.
Collapse
Affiliation(s)
- Georgi K. Marinov
- Department of Genetics, Stanford University, Stanford, CA, United States of America
| | | | - Antoni Rangachev
- Institute of Mathematics and Informatics, Bulgarian Academy of Sciences, Sofia, Bulgaria
- International Center for Mathematical Sciences-Sofia, Sofia, Bulgaria
| | - Ivailo Alexiev
- National Center of Infectious and Parasitic Diseases, Sofia, Bulgaria
| |
Collapse
|
75
|
Guevara R, Prado-Vivar B, Márquez S, Muñoz EB, Carvajal M, Guadalupe JJ, Becerra-Wong M, Proaño S, Bayas-Rea R, Coloma J, Grunauer M, Trueba G, Rojas-Silva P, Barragán V, Cárdenas P. Occurrence of SARS-CoV-2 reinfections at regular intervals in Ecuador. Front Cell Infect Microbiol 2022; 12:951383. [PMID: 36164552 PMCID: PMC9507970 DOI: 10.3389/fcimb.2022.951383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
SARS-CoV-2 reinfection is defined as a new infection with a different virus variant in an individual who has already recovered from a previous episode of COVID-19. The first case of reinfection in the world was described in August 2020, since then, reinfections have increased over time and their incidence has fluctuated with specific SARS-CoV-2 variant waves. Initially, reinfections were estimated to represent less than 1% of total COVID-19 infections. With the advent of the Omicron variant, reinfections became more frequent, representing up to 10% of cases (based on data from developed countries). The frequency of reinfections in Latin America has been scarcely reported. The current study shows that in Ecuador, the frequency of reinfections has increased 10-fold following the introduction of Omicron, after 22 months of surveillance in a single center of COVID-19 diagnostics. Suspected reinfections were identified retrospectively from a database of RT-qPCR-positive patients. Cases were confirmed by sequencing viral genomes from the first and second infections using the ONT MinION platform. Monthly surveillance showed that the main incidence peaks of reinfections were reached within four to five months, coinciding with the increase of COVID-19 cases in the country, suggesting that the emergence of reinfections is related to higher exposure to the virus during outbreaks. This study performed the longest monitoring of SARS-CoV-2 reinfections, showing an occurrence at regular intervals of 4-5 months and confirming a greater propensity of Omicron to cause reinfections.
Collapse
Affiliation(s)
- Rommel Guevara
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Belén Prado-Vivar
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Sully Márquez
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Erika B. Muñoz
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Mateo Carvajal
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Juan José Guadalupe
- Laboratorio de Biotecnología Vegetal, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Mónica Becerra-Wong
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Stefanie Proaño
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Rosa Bayas-Rea
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Josefina Coloma
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, United States
| | - Michelle Grunauer
- Escuela de Medicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Gabriel Trueba
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Patricio Rojas-Silva
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Verónica Barragán
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Paúl Cárdenas
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| |
Collapse
|
76
|
Kairov U, Amanzhanova A, Karabayev D, Rakhimova S, Aitkulova A, Samatkyzy D, Kalendar R, Kozhamkulov U, Molkenov A, Gabdulkayum A, Sarbassov D, Akilzhanova A. A high scale SARS-CoV-2 profiling by its whole-genome sequencing using Oxford Nanopore Technology in Kazakhstan. Front Genet 2022; 13:906318. [PMID: 36118859 PMCID: PMC9479076 DOI: 10.3389/fgene.2022.906318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Severe acute respiratory syndrome (SARS-CoV-2) is responsible for the worldwide pandemic, COVID-19. The original viral whole-genome was sequenced by a high-throughput sequencing approach from the samples obtained from Wuhan, China. Real-time gene sequencing is the main parameter to manage viral outbreaks because it expands our understanding of virus proliferation, spread, and evolution. Whole-genome sequencing is critical for SARS-CoV-2 variant surveillance, the development of new vaccines and boosters, and the representation of epidemiological situations in the country. A significant increase in the number of COVID-19 cases confirmed in August 2021 in Kazakhstan facilitated a need to establish an effective and proficient system for further study of SARS-CoV-2 genetic variants and the development of future Kazakhstan’s genomic surveillance program. The SARS-CoV-2 whole-genome was sequenced according to SARS-CoV-2 ARTIC protocol (EXP-MRT001) by Oxford Nanopore Technologies at the National Laboratory Astana, Kazakhstan to track viral variants circulating in the country. The 500 samples kindly provided by the Republican Diagnostic Center (UMC-NU) and private laboratory KDL “Olymp” were collected from individuals in Nur-Sultan city diagnosed with COVID-19 from August 2021 to May 2022 using real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR). All samples had a cycle threshold (Ct) value below 20 with an average Ct value of 17.03. The overall average value of sequencing depth coverage for samples is 244X. 341 whole-genome sequences that passed quality control were deposited in the Global initiative on sharing all influenza data (GISAID). The BA.1.1 (n = 189), BA.1 (n = 15), BA.2 (n = 3), BA.1.15 (n = 1), BA.1.17.2 (n = 1) omicron lineages, AY.122 (n = 119), B.1.617.2 (n = 8), AY.111 (n = 2), AY.126 (n = 1), AY.4 (n = 1) delta lineages, one sample B.1.1.7 (n = 1) belongs to alpha lineage, and one sample B.1.637 (n = 1) belongs to small sublineage were detected in this study. This is the first study of SARS-CoV-2 whole-genome sequencing by the ONT approach in Kazakhstan, which can be expanded for the investigation of other emerging viral or bacterial infections on the country level.
Collapse
Affiliation(s)
- Ulykbek Kairov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
- *Correspondence: Ulykbek Kairov, ; Ainur Akilzhanova,
| | - Amina Amanzhanova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Daniyar Karabayev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Saule Rakhimova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Akbota Aitkulova
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Diana Samatkyzy
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Ruslan Kalendar
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Ulan Kozhamkulov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Askhat Molkenov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Aidana Gabdulkayum
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Dos Sarbassov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Ainur Akilzhanova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
- *Correspondence: Ulykbek Kairov, ; Ainur Akilzhanova,
| |
Collapse
|
77
|
Dowran R, Damavandi AR, Azad TM. Reinfection and reactivation of SARS-CoV-2. Future Virol 2022. [PMID: 36176508 PMCID: PMC9514089 DOI: 10.2217/fvl-2021-0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 08/31/2022] [Indexed: 11/21/2022]
Abstract
As the cases of SARS-CoV-2 infection escalates, the essence of in-depth knowledge around acquired immunity and emergence of reinfection and reactivation have to be captured. While being a rare phenomenon, reinfection occurs as the result of diminishing protection conferred by antibodies, especially IgG. Reactivation is more concerned with the role of various elements including shedding lingering viral RNA for a prolonged time and incomplete resolution of infection along with the insight of dormant viral exosomes’ role. The concept of testing positive after two consecutive negative results requires proper discrimination of reinfection from reactivation. In this review, we summarized the current evidence for possible mechanisms leading to viral reactivation or test re-positivity. We also pointed out risk factors associated with both reinfection and reactivation.
Collapse
Affiliation(s)
- Razieh Dowran
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Student Scientific Association of Virology, Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirmasoud Rayati Damavandi
- Student Scientific Association of Virology, Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
78
|
Mencacci A, Gili A, Camilloni B, Bicchieraro G, Spaccapelo R, Bietta C, Stracci F. Immediate reinfection with Omicron variant after clearance of a previous SARS-CoV-2 infection. J Infect Public Health 2022; 15:983-985. [PMID: 35963101 PMCID: PMC9330370 DOI: 10.1016/j.jiph.2022.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/17/2022] [Accepted: 07/26/2022] [Indexed: 11/23/2022] Open
Abstract
SARS-CoV-2 Omicron variant is spreading worldwide, causing unprecedented epidemic peaks due to its transmissibility and immune evasion. We searched in the archive of the Regional Microbiology Laboratory (Umbria, Italy) for immediate reinfection (i.e. infection occurring 25-60 days from primary infection) among 454,764 RT-PCR tests from 261,217 individuals. Lineage heterogeneity was assessed by S gene target failure phenomenon or whole genome sequencing. We found that BA.1 Omicron variant may cause immediate reinfection of patients just recovered from Delta infection. Immediate reinfection was not observed for any other combination of variants, including Delta over Alpha variant and BA.2 over BA.1 Omicron lineage.
Collapse
Affiliation(s)
- Antonella Mencacci
- Microbiology and Clinical Microbiology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Alessio Gili
- Public Health Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Barbara Camilloni
- Microbiology and Clinical Microbiology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giulia Bicchieraro
- Microbiology and Clinical Microbiology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberta Spaccapelo
- Microbiology and Clinical Microbiology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carla Bietta
- Epidemiology Unit, Department of Preventive Medicine, Local Health Unit 1, Umbria region, Italy
| | - Fabrizio Stracci
- Public Health Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
79
|
Gorzalski AJ, Boyles C, Sepcic V, Verma S, Sevinsky J, Libuit K, Van Hooser S, Pandori MW. Rapid repeat infection of SARS-CoV-2 by two highly distinct delta-lineage viruses. Diagn Microbiol Infect Dis 2022; 104:115747. [PMID: 35843112 PMCID: PMC9217632 DOI: 10.1016/j.diagmicrobio.2022.115747] [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: 11/09/2021] [Revised: 06/01/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
An instance of sequential infection of an individual with, firstly, the Delta variant and secondly a Delta-sub-lineage has been identified. The individual was found positive for the AY.26 lineage 22 days after being found positive for the Delta [B.1.617.2] variant. The viruses associated with the cases showed dramatic genomic difference, including 31 changes that resulted in deletions or amino acid substitutions. Seven of these differences were observed in the Spike protein. The patient in question was between 30 and 35 years old and had no underlying health conditions. Though singular, this case illustrates the possibility that infection with the Delta variant may not itself be fully protective against a population of SARS-CoV-2 variants that are becoming increasingly diverse.
Collapse
Affiliation(s)
| | | | - Victoria Sepcic
- Nevada Department of Health and Human Services, Department of Public and Behavioral Health, Carson City, NV, USA
| | - Subhash Verma
- Department of Microbiology and Immunology, University of Nevada, School of Medicine, Reno, NV, USA
| | | | | | | | - Mark W Pandori
- Nevada State Public Health Laboratory, Reno, NV, USA; Department of Pathology and Laboratory Medicine, University of Nevada, School of Medicine, Reno, NV, USA.
| |
Collapse
|
80
|
Attwood SW, Hill SC, Aanensen DM, Connor TR, Pybus OG. Phylogenetic and phylodynamic approaches to understanding and combating the early SARS-CoV-2 pandemic. Nat Rev Genet 2022; 23:547-562. [PMID: 35459859 PMCID: PMC9028907 DOI: 10.1038/s41576-022-00483-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2022] [Indexed: 01/05/2023]
Abstract
Determining the transmissibility, prevalence and patterns of movement of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections is central to our understanding of the impact of the pandemic and to the design of effective control strategies. Phylogenies (evolutionary trees) have provided key insights into the international spread of SARS-CoV-2 and enabled investigation of individual outbreaks and transmission chains in specific settings. Phylodynamic approaches combine evolutionary, demographic and epidemiological concepts and have helped track virus genetic changes, identify emerging variants and inform public health strategy. Here, we review and synthesize studies that illustrate how phylogenetic and phylodynamic techniques were applied during the first year of the pandemic, and summarize their contributions to our understanding of SARS-CoV-2 transmission and control.
Collapse
Affiliation(s)
- Stephen W Attwood
- Department of Zoology, University of Oxford, Oxford, UK.
- Pathogen Genomics Unit, Public Health Wales NHS Trust, Cardiff, UK.
| | - Sarah C Hill
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, UK
| | - David M Aanensen
- Centre for Genomic Pathogen Surveillance, Wellcome Genome Campus, Hinxton, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thomas R Connor
- Pathogen Genomics Unit, Public Health Wales NHS Trust, Cardiff, UK
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford, UK.
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, UK.
| |
Collapse
|
81
|
SARS-CoV-2 Reinfection Rate and Outcomes in Saudi Arabia: A National Retrospective Study. Int J Infect Dis 2022; 122:758-766. [PMID: 35840098 PMCID: PMC9364818 DOI: 10.1016/j.ijid.2022.07.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/11/2022] [Accepted: 07/07/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The characterization of reinfection with SARS-CoV-2 has been a subject of concern and controversy, especially with the surge of infections with highly transmissible variants worldwide. METHODS This retrospective national study used comorbidities, vaccination status, SARS-CoV-2 variants of concern, and demographics data to profile participants who were reinfected with SARS-CoV-2, defined as having two reverse transcriptase-polymerase chain reaction-positive SARS-CoV-2 tests within at least 90 days apart. A multivariate logistic regression model assessed the risk factors associated with reinfection . Two control groups were selected: nonreinfected participants reporting a positive test (control group one) and those reporting a negative test (control group two). RESULTS Between March 2020 and December 2021, 4454 reinfected participants were identified in Saudi Arabia (0.8%, 95% confidence interval [CI] 0.7-0.8). The majority (67.3%) were unvaccinated (95% CI 65.9-68.7) and 0.8% (95% CI 0.6-1.1) had severe or fatal SARS-CoV-2 disease. COVID-19 vaccines were 100% effective against mortality in reinfected individuals who received at least one dose, whereas it conferred 61% (odds ratio [OR] 0.4, 95% CI 0.1-1.0) additional protection against severe disease after the first dose and 100% after the second dose. In the risk factor analysis, reinfection was highly associated with comorbidities, such as HIV (OR 2.5, 95% CI 1.3-5.2; P = 0.009), obesity (OR 2.3, 95% CI 1.3-3.9; P = 0.003), pregnancy (OR 3.2, 95% CI 1.4-7.4; P = 0.005), and working in health care facilities (OR 6.1, 95% CI 3.1-12.9; P <0.0001). The delta variant (B.1.617.2) was the most frequent variant of concern among the reinfected cohort. CONCLUSION This in-depth study of the reinfection profile identified risk factors and highlighted the associated SARS-CoV-2 variants. Results showed that naturally acquired immunity to SARS-CoV-2 through multiple reinfections together with vaccine-induced immunity provided substantial protection against severe SARS-CoV-2 disease and mortality.
Collapse
|
82
|
Cattaneo A, Vitali A, Mazzoleni M, Previdi F. An agent-based model to assess large-scale COVID-19 vaccination campaigns for the Italian territory: The case study of Lombardy region. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 224:107029. [PMID: 35908330 PMCID: PMC9287580 DOI: 10.1016/j.cmpb.2022.107029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 05/04/2023]
Abstract
BACKGROUND In Italy, the administration of COVID-19 vaccines began in late 2020. In the early stages, the number of available doses was limited. To maximize the effectiveness of the vaccine campaign, the national health agency assigned priority access to at-risk individuals, such as health care workers and the elderly. Current vaccination campaign strategies do not take full advantage of the latest mathematical models, which capture many subtle nuances, allowing different territorial situations to be analyzed aiming to make context-specific decisions. OBJECTIVES The main objective is the definition of an agent-based model using open data and scientific literature to assess and optimize the impact of vaccine campaigns for an Italian region. Specifically, the aim is twofold: (i) estimate the reduction in the number of infections and deaths attributable to vaccines, and (ii) assess the performances of alternative vaccine allocation strategies. METHODS The COVID-19 Agent-based simulator Covasim has been employed to build an agent-based model by considering the Lombardy region as case study. The model has been tailored by leveraging open data and knowledge from the scientific literature. Dynamic mobility restrictions and the presence of Variant of Concern have been explicitly represented. Free parameters have been calibrated using the grid search methodology. RESULTS The model mimics the COVID-19 wave that hit Lombardy from September 2020 to April 2021. It suggests that 168,492 cumulative infections 2,990 cumulative deaths have been avoided due to the vaccination campaign in Lombardy from January 1 to April 30, 2021. Without vaccines, the number of deaths would have been 66% greater in the 80-89 age group and 114% greater for those over 90. The best vaccine allocation strategy depends on the goal. To minimize infections, the best policy is related to dose availability. If at least 1/3 of the population can be covered in 4 months, targeting at-risk individuals and the elderly first is recommended; otherwise, the youngest people should be vaccinated first. To minimize overall deaths, priority is best given to at-risk groups and the elderly in all scenarios. CONCLUSIONS This work proposes a methodological approach that leverages open data and scientific literature to build a model of COVID-19 capable of assessing and optimizing the impact of vaccine campaigns. This methodology can help national institutions to design regional mathematical models that can support pandemic-related decision-making processes.
Collapse
Affiliation(s)
- Andrea Cattaneo
- Department of Management, Information and Production Engineering, University of Bergamo, via Salvecchio 19 - Bergamo, Italy.
| | - Andrea Vitali
- Department of Management, Information and Production Engineering, University of Bergamo, via Salvecchio 19 - Bergamo, Italy.
| | - Mirko Mazzoleni
- Department of Management, Information and Production Engineering, University of Bergamo, via Salvecchio 19 - Bergamo, Italy.
| | - Fabio Previdi
- Department of Management, Information and Production Engineering, University of Bergamo, via Salvecchio 19 - Bergamo, Italy.
| |
Collapse
|
83
|
Sen R, Sengupta D, Mukherjee A. Mechanical dependency of the SARS-CoV-2 virus and the renin-angiotensin-aldosterone (RAAS) axis: a possible new threat. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62235-62247. [PMID: 34859345 PMCID: PMC8638800 DOI: 10.1007/s11356-021-16356-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/01/2021] [Indexed: 04/12/2023]
Abstract
Pathogens in our environment can act as agents capable of inflicting severe human diseases. Among them, the SARS-CoV-2 virus has recently plagued the globe and paralyzed the functioning of ordinary human life. The virus enters the cell through the angiotensin-converting enzyme-2 (ACE-2) receptor, an integral part of the renin-angiotensin system (RAAS). Reports on hypertension and its relation to the modulation of the RAAS are generating interest in the scientific community. This short review focuses on the SARS-CoV-2 infection's direct and indirect effects on our body through modulation of the RAAS axis. A patient having severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection, which causes COVID-19 relates to hypertension as a pre-existing disease or develops it in a post-COVID scenario. Several studies on how SARS-CoV-2 modulates the RAAS axis indicate that it alters our body's physiological balance. This review seeks to establish a hypothesis on the mechanical dependency of SARS-CoV-2 and RAAS modulation in the human body. This study intends to impart ideas on drug development and designing by targeting the modulation of the RAAS axis to inactivate the pathogenicity of the SARS-CoV-2 virus. A systematic hypothesis can severely attenuate the pathogenicity of the dreadful viruses of the future.
Collapse
Affiliation(s)
- Rohit Sen
- Department of Zoology, Charuchandra College, University of Calcutta, 22, Lake Road, Kolkata, 700029 India
| | | | - Avinaba Mukherjee
- Department of Zoology, Charuchandra College, University of Calcutta, 22, Lake Road, Kolkata, 700029 India
| |
Collapse
|
84
|
Bean DJ, Monroe J, Turcinovic J, Moreau Y, Connor JH, Sagar M. Severe Acute Respiratory Syndrome Coronavirus 2 Reinfection Associates With Unstable Housing and Occurs in the Presence of Antibodies. Clin Infect Dis 2022; 75:e208-e215. [PMID: 34755830 PMCID: PMC8689949 DOI: 10.1093/cid/ciab940] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The factors associated with severe acute respiratory coronavirus 2 (SARS-CoV-2) reinfection remain poorly defined. METHODS We identified patients with SARS-CoV-2 infection and at least 1 repeat reverse transcription polymerase chain reaction result a minimum of 90 days after the initial positive test and before 21 January 2021. Those with a repeat positive test were deemed to have reinfection (n = 75), and those with only negative tests were classified as convalescents (n = 1594). Demographics, coronavirus disease 2019 (COVID-19) severity, and treatment histories were obtained from the Boston Medical Center electronic medical record. Humoral responses were analyzed using SARS-CoV-2-specific enzyme-linked immunosorbent assays and pseudovirus neutralizations in a subset of reinfection (n = 16) and convalescent samples (n = 32). Univariate, multivariate, and time to event analyses were used to identify associations. RESULTS Individuals with reinfection had more frequent testing at shorter intervals compared with the convalescents. Unstable housing was associated with more than 2-fold greater chance of reinfection. Preexisting comorbidities and COVID-19 severity after the initial infection were not associated with reinfection. SARS-CoV-2 immunoglobulin G levels and pseudovirus neutralization were not different within the early weeks after primary infection and at a timepoint at least 90 days later in the 2 groups. In the convalescents, but not in those with reinfection, the late as compared with early humoral responses were significantly higher. CONCLUSIONS Reinfection associates with unstable housing, which is likely a marker for virus exposure, and reinfection occurs in the presence of SARS-CoV-2 antibodies.
Collapse
Affiliation(s)
- David J Bean
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Janet Monroe
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jacquelyn Turcinovic
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yvetane Moreau
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - John H Connor
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Manish Sagar
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
85
|
Vo V, Tillett RL, Papp K, Shen S, Gu R, Gorzalski A, Siao D, Markland R, Chang CL, Baker H, Chen J, Schiller M, Betancourt WQ, Buttery E, Pandori M, Picker MA, Gerrity D, Oh EC. Use of wastewater surveillance for early detection of Alpha and Epsilon SARS-CoV-2 variants of concern and estimation of overall COVID-19 infection burden. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 835:155410. [PMID: 35469875 PMCID: PMC9026949 DOI: 10.1016/j.scitotenv.2022.155410] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 05/14/2023]
Abstract
A decline in diagnostic testing for SARS-CoV-2 is expected to delay the tracking of COVID-19 variants of concern and interest in the United States. We hypothesize that wastewater surveillance programs provide an effective alternative for detecting emerging variants and assessing COVID-19 incidence, particularly when clinical surveillance is limited. Here, we analyzed SARS-CoV-2 RNA in wastewater from eight locations across Southern Nevada between March 2020 and April 2021. Trends in SARS-CoV-2 RNA concentrations (ranging from 4.3 log10 gc/L to 8.7 log10 gc/L) matched trends in confirmed COVID-19 incidence, but wastewater surveillance also highlighted several limitations with the clinical data. Amplicon-based whole genome sequencing (WGS) of 86 wastewater samples identified the B.1.1.7 (Alpha) and B.1.429 (Epsilon) lineages in December 2020, but clinical sequencing failed to identify the variants until January 2021, thereby demonstrating that 'pooled' wastewater samples can sometimes expedite variant detection. Also, by calibrating fecal shedding (11.4 log10 gc/infection) and wastewater surveillance data to reported seroprevalence, we estimate that ~38% of individuals in Southern Nevada had been infected by SARS-CoV-2 as of April 2021, which is significantly higher than the 10% of individuals confirmed through clinical testing. Sewershed-specific ascertainment ratios (i.e., X-fold infection undercounts) ranged from 1.0 to 7.7, potentially due to demographic differences. Our data underscore the growing application of wastewater surveillance in not only the identification and quantification of infectious agents, but also the detection of variants of concern that may be missed when diagnostic testing is limited or unavailable.
Collapse
Affiliation(s)
- Van Vo
- Laboratory of Neurogenetics and Precision Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Richard L Tillett
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Katerina Papp
- Southern Nevada Water Authority, P.O. Box 99954, Las Vegas, NV, 89193, USA
| | - Shirley Shen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Richard Gu
- Laboratory of Neurogenetics and Precision Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | | | - Danielle Siao
- Nevada State Public Health Laboratory, Reno, NV 89597, USA
| | - Rayma Markland
- Southern Nevada Public Health Laboratory of the Southern Nevada Health District, Las Vegas, NV 89106, USA
| | - Ching-Lan Chang
- Laboratory of Neurogenetics and Precision Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Hayley Baker
- Laboratory of Neurogenetics and Precision Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Martin Schiller
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Walter Q Betancourt
- Water & Energy Sustainable Technology (WEST) Center, University of Arizona, 2959 W. Calle Agua Nueva, Tucson, AZ 85745, USA
| | - Erin Buttery
- Southern Nevada Public Health Laboratory of the Southern Nevada Health District, Las Vegas, NV 89106, USA
| | - Mark Pandori
- Nevada State Public Health Laboratory, Reno, NV 89597, USA
| | - Michael A Picker
- Southern Nevada Public Health Laboratory of the Southern Nevada Health District, Las Vegas, NV 89106, USA
| | - Daniel Gerrity
- Southern Nevada Water Authority, P.O. Box 99954, Las Vegas, NV, 89193, USA.
| | - Edwin C Oh
- Laboratory of Neurogenetics and Precision Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; Department of Internal Medicine, UNLV School of Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA.
| |
Collapse
|
86
|
Isnaini N, Mardian Y, Lokida D, Budiono F, Butar-Butar DP, Arlinda D, Salim G, Kosasih H, Wulan WN, Perodin J, Neal A, Lane HC, Karyana M. Mild reinfection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Delta variant: First case report from Indonesia. Front Med (Lausanne) 2022; 9:906469. [PMID: 35935779 PMCID: PMC9355687 DOI: 10.3389/fmed.2022.906469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/27/2022] [Indexed: 12/21/2022] Open
Abstract
Background Reinfection with SARS-CoV-2 has been well documented, yet little is known about the degree of protection a previous infection provides against reinfection, especially against Variants of Concern (VOC). Case presentation Here we describe a case of an unvaccinated 49-year-old man who experienced two sequential SARS-CoV-2 infections with two different variants, as evidenced by genomic sequencing. The first episode was caused by the Pango lineage B.1.466.2 and resulted in severe COVID-19 with 5 days in an intensive care unit (ICU). The second episode occurred approximately 6 months later, during the Delta surge in Indonesia. Genomic analysis showed that the second infection was caused by the Delta variant (Pango lineage B.1.617.2) and resulted in mild disease that did not require hospitalization. No SARS-CoV-2 nucleic acid was detected between the two episodes, but both binding and neutralizing antibodies to SARS-CoV-2 were detected prior to the reinfection, with the second infection leading to an increase in the levels of antibody. Conclusion We confirmed that the patient experienced a reinfection instead of persistent viral shedding from the first infection based on epidemiological, clinical, serological, and genomic analyses. Our case supports the hypothesis that SARS-CoV-2 reinfection may occur once antibody titers decrease or following the emergence of a new variant. The milder presentation in the patient’s second infection deserves further investigation to provide a clear picture of the role of post-infection immunity in altering the course of subsequent disease.
Collapse
Affiliation(s)
| | - Yan Mardian
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Dewi Lokida
- Tangerang District Hospital, Tangerang, Indonesia.,Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | | | - Deni P Butar-Butar
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Dona Arlinda
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia.,National Institute of Health Research and Development, Ministry of Health, Jakarta, Indonesia
| | - Gustiani Salim
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Herman Kosasih
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | - Wahyu Nawang Wulan
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia
| | | | - Aaron Neal
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Muhammad Karyana
- Indonesia Research Partnership on Infectious Disease, Jakarta, Indonesia.,National Institute of Health Research and Development, Ministry of Health, Jakarta, Indonesia
| |
Collapse
|
87
|
Okendo J. Characterization of the expressed RNA variants from young patients with critical and non-critical SARS-CoV-2 infection. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022; 23:115. [PMID: 37521832 PMCID: PMC9362069 DOI: 10.1186/s43042-022-00327-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/27/2022] [Indexed: 11/10/2022] Open
Abstract
Background Since the COVID-19 outbreak emerged, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has continuously evolved into variants with underlying mutations associated with increased transmissibility, potential escape from neutralizing antibodies, and disease severity. Although intensive research is ongoing worldwide to understand the emergence of SARS-CoV-2 variants, there is a lack of information on what constitutes the expressed RNA variants in critical and non-critical comorbidity-free young patients. The study sought to characterize the expressed RNA variants from young patients with critical and non-critical forms of SARS-CoV-2 infection. Methodology The bulk ribonucleic acid (RNA) sequencing data with the identifier GSE172114 were downloaded from the Gene Expression Omnibus (GEO) database. The study participants were divided into critical, n = 46, and non-critical, n = 23. FastQC version 0.11.9 and Cutadapt version 3.7 were used to assess the read quality and perform adapter trimming, respectively. Spliced Transcripts Alignment to a Reference (STAR) version 2.7.10a was used to align reads to the human (hg38) reference genome. Genome Analysis Tool Kit (GATK) best practice was followed to call variants using the rnavar pipeline, part of the nf-core pipelines. Results Our research demonstrates that critical and non-critical SARS-CoV-2-infected individuals are characterized by a unique set of expressed RNA variants. The expressed gene variants are enriched on the innate immune response, specifically neutrophil-mediated immune response. On the other hand, the expressed gene variants are involved in both innate and cellular immune responses. Conclusion Deeply phenotyped comorbidity-free young patients with critical and non-critical SARS-CoV-2 infection are characterized by a unique set of expressed RNA variants. The findings in this study can inform the patient classification process in health facilities globally when admitting young patients infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Javan Okendo
- Systems and Chemical Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road Observatory, Cape Town, 7925 South Africa
| |
Collapse
|
88
|
Yang J, Han M, Wang L, Wang L, Xu T, Wu L, Ma J, Wong G, Liu W, Gao GF, Bi Y. Relatively rapid evolution rates of SARS-CoV-2 spike gene at the primary stage of massive vaccination. BIOSAFETY AND HEALTH 2022; 4:228-233. [PMID: 35856045 PMCID: PMC9277989 DOI: 10.1016/j.bsheal.2022.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022] Open
Abstract
A series of stringent non-pharmacological and pharmacological interventions were implemented to contain the pandemic but the pandemic continues. Moreover, vaccination breakthrough infection and reinfection in convalescent coronavirus disease 2019 (COVID-19) cases have been reported. Further, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants emerged with mutations in spike (S) gene, the target of most current vaccines. Importantly, the mutations exhibit a trend of immune escape from the vaccination. Herein the scientific question that if the vaccination drives genetic or antigenic drifts of SARS-CoV-2 remains elusive. We performed correlation analyses to uncover the impacts of wide vaccination on epidemiological characteristics of COVID-19. In addition, we investigated the evolutionary dynamics and genetic diversity of SARS-CoV-2 under immune pressure by utilizing the Bayesian phylodynamic inferences and the lineage entropy calculation respectively. We found that vaccination coverage was negatively related to the infections, severe cases, and deaths of COVID-19 respectively. With the increasing vaccination coverage, the lineage diversity of SARS-CoV-2 dampened, but the rapid mutation rates of the S gene were identified, and the vaccination could be one of the explanations for driving mutations in S gene. Moreover, new epidemics resurged in several countries with high vaccination coverage, questioning their current pandemic control strategies. Hence, integrated vaccination and non-pharmacological interventions are critical to control the pandemic. Furthermore, novel vaccine preparation should enhance its capabilities to curb both disease severity and infection possibility.
Collapse
Affiliation(s)
- Jing Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Han
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Liang Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Likui Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Tianrui Xu
- Department of Anatomy and Cell Biology, McGill University, Montreal H3A0G4, Canada
| | - Linhuan Wu
- Microbial Resource and Big Data Center, Chinese National Microbiology Data Center (NMDC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Juncai Ma
- Microbial Resource and Big Data Center, Chinese National Microbiology Data Center (NMDC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Gary Wong
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - George F. Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuhai Bi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China,Corresponding author: CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
89
|
Nguyen DC, Lamothe PA, Woodruff MC, Saini AS, Faliti CE, Sanz I, Lee FE. COVID-19 and plasma cells: Is there long-lived protection? Immunol Rev 2022; 309:40-63. [PMID: 35801537 PMCID: PMC9350162 DOI: 10.1111/imr.13115] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infection with SARS-CoV-2, the etiology of the ongoing COVID-19 pandemic, has resulted in over 450 million cases with more than 6 million deaths worldwide, causing global disruptions since early 2020. Memory B cells and durable antibody protection from long-lived plasma cells (LLPC) are the mainstay of most effective vaccines. However, ending the pandemic has been hampered by the lack of long-lived immunity after infection or vaccination. Although immunizations offer protection from severe disease and hospitalization, breakthrough infections still occur, most likely due to new mutant viruses and the overall decline of neutralizing antibodies after 6 months. Here, we review the current knowledge of B cells, from extrafollicular to memory populations, with a focus on distinct plasma cell subsets, such as early-minted blood antibody-secreting cells and the bone marrow LLPC, and how these humoral compartments contribute to protection after SARS-CoV-2 infection and immunization.
Collapse
Affiliation(s)
- Doan C. Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Pedro A. Lamothe
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Matthew C. Woodruff
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ankur S. Saini
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Caterina E. Faliti
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ignacio Sanz
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Frances Eun‐Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
90
|
Goto K, Komatsu K, Sekizuka T, Ebisawa H, Ootake M, Honda M, Nagata N, Yoshida D, Yanaoka T, Kimura H, Kuroda M. Detection of SARS-CoV-2 Genome for over 100 Days after COVID-19 Onset. Jpn J Infect Dis 2022; 75:620-622. [PMID: 35908868 DOI: 10.7883/yoken.jjid.2021.841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019, is spreading globally. In general, the viral genome becomes undetectable within a couple of weeks after infection. Herein, we report a case of long-term detection of the SARS-CoV-2 genome from the same individual for 106 days. Whole genome sequencing was performed on specimens taken at the onset of the disease and at 2 months after onset, and the B.1.1.7 lineage was detected in both samples. Comparison of the full-length sequences revealed a single-base difference and no amino acid mutations. This is the first case in Japan where the virus was detected over a long time, and the full-length sequences were compared.
Collapse
Affiliation(s)
- Keiko Goto
- Department of Virology, Ibaraki Prefectural Institute of Public Health, Japan
| | - Kenichi Komatsu
- Department of Internal Medicine, Ibaraki Western Medical Center, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomics Center, National Institute of Infectious Diseases, Japan
| | - Hiromi Ebisawa
- Department of Internal Medicine, Ibaraki Western Medical Center, Japan
| | | | | | - Noriko Nagata
- Department of Virology, Ibaraki Prefectural Institute of Public Health, Japan
| | - Daisuke Yoshida
- Department of Virology, Ibaraki Prefectural Institute of Public Health, Japan
| | - Toshikazu Yanaoka
- Department of Virology, Ibaraki Prefectural Institute of Public Health, Japan
| | - Hirokazu Kimura
- Department of Health Science, Gunma Paz University Graduate School, Japan
| | - Makoto Kuroda
- Pathogen Genomics Center, National Institute of Infectious Diseases, Japan
| |
Collapse
|
91
|
Li H, Zhao X, Li J, Zheng H, Zhao Y, Yang J, Zhou J, Yang F, Chen Y, Zuo Y, Lai Q, Long H, Li Y, Jin W, Shi H, Liu L. Nasal Mucosa Exploited by SARS-CoV-2 for Replicating and Shedding during Reinfection. Viruses 2022; 14:v14081608. [PMID: 35893674 PMCID: PMC9394478 DOI: 10.3390/v14081608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
Reinfection risk is a great concern with regard to the COVID-19 pandemic because a large proportion of the population has recovered from an initial infection, and previous reports found that primary exposure to SARS-CoV-2 protects against reinfection in rhesus macaques without viral presence and pathological injury; however, a high possibility for reinfection at the current stage of the pandemic has been proven. We found the reinfection of SARS-CoV-2 in Syrian hamsters with continuous viral shedding in the upper respiratory tracts and few injuries in the lung, and nasal mucosa was exploited by SARS-CoV-2 for replication and shedding during reinfection; meanwhile, no viral replication or enhanced damage was observed in the lower respiratory tracts. Consistent with the mild phenotype in the reinfection, increases in mRNA levels in cytokines and chemokines in the nasal mucosa but only slight increases in the lung were found. Notably, the high levels of neutralizing antibodies in serum could not prevent reinfection in hamsters but may play roles in benefitting the lung recovery and symptom relief of COVID-19. In summary, Syrian hamsters could be reinfected by SARS-CoV-2 with mild symptoms but with obvious viral shedding and replication, and both convalescent and vaccinated patients should be wary of the transmission and reinfection of SARS-CoV-2.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Longding Liu
- Correspondence: ; Tel.: +86-871-6833-5905; Fax: +86-871-6833-4483
| |
Collapse
|
92
|
Modelling the interplay of SARS-CoV-2 variants in the United Kingdom. Sci Rep 2022; 12:12372. [PMID: 35859100 PMCID: PMC9296900 DOI: 10.1038/s41598-022-16147-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/05/2022] [Indexed: 12/28/2022] Open
Abstract
Many COVID-19 vaccines are proving to be highly effective to prevent severe disease and to diminish infections. Their uneven geographical distribution favors the appearance of new variants of concern, as the highly transmissible Delta variant, affecting particularly non-vaccinated people. It is important to device reliable models to analyze the spread of the different variants. A key factor is to consider the effects of vaccination as well as other measures used to contain the pandemic like social behaviour. The stochastic geographical model presented here, fulfills these requirements. It is based on an extended compartmental model that includes various strains and vaccination strategies, allowing to study the emergence and dynamics of the new COVID-19 variants. The model conveniently separates the parameters related to the disease from the ones related to social behavior and mobility restrictions. We applied the model to the United Kingdom by using available data to fit the recurrence of the currently prevalent variants. Our computer simulations allow to describe the appearance of periodic waves and the features that determine the prevalence of certain variants. They also provide useful predictions to help planning future vaccination boosters. We stress that the model could be applied to any other country of interest.
Collapse
|
93
|
Fels JM, Khan S, Forster R, Skalina KA, Sirichand S, Fox AS, Bergman A, Mitchell WB, Wolgast LR, Szymczak WA, Bortz RH, Dieterle ME, Florez C, Haslwanter D, Jangra RK, Laudermilch E, Wirchnianski AS, Barnhill J, Goldman DL, Khine H, Goldstein DY, Daily JP, Chandran K, Kelly L. Genomic surveillance of SARS-CoV-2 during the first year of the pandemic in the Bronx enabled clinical and epidemiological inference. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006211. [PMID: 35831070 PMCID: PMC9528964 DOI: 10.1101/mcs.a006211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
The Bronx was an early epicenter of the COVID-19 pandemic in the USA. We conducted temporal genomic surveillance of 104 SARS-CoV-2 genomes across the Bronx from March October 2020. Although the local structure of SARS-CoV-2 lineages mirrored those of New York City and New York State, temporal sampling revealed a dynamic and changing landscape of SARS-CoV-2 genomic diversity. Mapping the trajectories of mutations, we found that while some became 'endemic' to the Bronx, other, novel mutations rose in prevalence in the late summer/early fall. Geographically resolved genomes enabled us to distinguish between cases of reinfection and persistent infection in two pediatric patients. We propose that limited, targeted, temporal genomic surveillance has clinical and epidemiological utility in managing the ongoing COVID pandemic.
Collapse
Affiliation(s)
| | | | | | - Karin A Skalina
- Montefiore Medical Center/Albert Einstein College of Medicine
| | | | - Amy S Fox
- Montefiore Medical Center/Albert Einstein College of Medicine
| | | | | | - Lucia R Wolgast
- Montefiore Medical Center/Albert Einstein College of Medicine
| | | | | | | | - Catalina Florez
- Albert Einstein College of Medicine and United States Military Academy at West Point
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Wu CS, Chiang HM, Chen Y, Chen CY, Chen HF, Su WC, Wang WJ, Chou YC, Chang WC, Wang SC, Hung MC. Prospects of Coffee Leaf against SARS-CoV-2 Infection. Int J Biol Sci 2022; 18:4677-4689. [PMID: 35874948 PMCID: PMC9305275 DOI: 10.7150/ijbs.76058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
In the current climate, many countries are in dire need of effective preventive methods to curb the Severe Acute Respiratory Syndrome Coronavirus Type 2 (SARS-CoV-2) pandemic. The purpose of this research is to screen and explore natural plant extracts that have the potential to against SARS-CoV-2 and provide alternative options for SARS-CoV-2 prevention and hand sanitizer or spray-like disinfectants. We first used Spike-ACE2 ELISA and TMPRSS2 fluorescence resonance energy transfer (FRET) assays to screen extracts from agricultural by-products from Taiwan with the potential to impede SARS-CoV-2 infection. Next, the SARS-CoV-2 pseudo-particles (Vpp) infection assay was tested to validate the effectiveness. We identified an extract from coffee leaf (Coffea Arabica), a natural plant that effectively inhibited wild-type SARS-CoV-2, and five Variants of Concern (Alpha, Beta, Gamma, Delta, and Omicron strain) from entering host cells. In an attempt to apply coffee leaf extract for hand sanitizer or spray-like disinfectants, we designed a skin-like gelatin membrane experiment. We showed that the high concentration of coffee leaf extract on the skin surface could block SARS-CoV-2 into cells more potently than 75% Ethanol, a standard disinfectant to inactivate SARS-CoV-2. Finally, LC-HRMS analysis was used to identify compounds such as caffeine, chlorogenic acid (CGA), quinic acid, and mangiferin that are associated with an anti-SARS-CoV-2 activity. Our results demonstrated that coffee leaf extract, an agricultural by-product effectively inhibits SARS-CoV-2 Vpp infection through an ACE2-dependent mechanism and may be utilized to develop products against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Chen-Shiou Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
| | - Hsiu-Mei Chiang
- Department of Cosmeceutics, China Medical University, Taichung 406040, Taiwan
| | - Yeh Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Institute of New Drug Development, China Medical University, Taichung 406040, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan
| | - Chung-Yu Chen
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan
| | - Hsiao-Fan Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
| | - Wen-Chi Su
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- International Master's Program of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404332, Taiwan
| | - Wei-Jan Wang
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung 406040, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei 115024, Taiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung 404332, Taiwan
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung 404332, Taiwan
- Department of Biotechnology, Asia University, Taichung, 41354 Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung 404332, Taiwan
- Department of Biotechnology, Asia University, Taichung, 41354 Taiwan
| |
Collapse
|
95
|
Genomic evidence for divergent co-infections of co-circulating SARS-CoV-2 lineages. Comput Struct Biotechnol J 2022; 20:4015-4024. [PMID: 35915661 PMCID: PMC9330581 DOI: 10.1016/j.csbj.2022.07.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 12/15/2022] Open
|
96
|
An Outbreak of SARS-CoV-2 Reinfection in a Long-Term Care Facility in South Korea. J Infect Public Health 2022; 15:966-969. [PMID: 35932619 PMCID: PMC9329134 DOI: 10.1016/j.jiph.2022.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/03/2022] [Accepted: 07/23/2022] [Indexed: 11/20/2022] Open
Abstract
We report a cluster of 12 cases of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection in a long-term care facility in South Korea. There were two outbreaks of SARS-CoV-2 infection in the facility at the beginning and end of October 2021, respectively. All residents in the facility were screened for SARS-CoV-2 infection using RT-PCR as part of the investigation of the second outbreak. Twelve residents, who had infection confirmed during the first outbreak, were found to be re-positive for RT-PCR test at the second outbreak. 8 Of 12 RT-PCR re-positive cases were confirmed as reinfections based on investigation through the whole genome sequencing, viral culture, and serological analysis, despite of the short interval between the first and second outbreaks (29–33 days) and a history of full vaccination for 7 of the 12 re-positive cases. This study suggests that decreased immunity and underlying health condition in older adults makes them susceptible to reinfection, highlighting the importance of prevention and control measures regardless of vaccination status in long-term care settings.
Collapse
|
97
|
Chowdhury D, Fremed MA, Dean P, Glickstein JS, Robinson J, Rellosa N, Thacker D, Soma D, Briskin SM, Asplund C, Johnson J, Snyder C. Return to Activity After SARS-CoV-2 Infection: Cardiac Clearance for Children and Adolescents. Sports Health 2022; 14:460-465. [PMID: 34427496 PMCID: PMC9214892 DOI: 10.1177/19417381211039746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
CONTEXT Sedentary behavior and inability to participate in organized physical activity has negatively affected the physical and mental health of children and adolescents; however, cardiac injury and associated risk for sudden cardiac death with return to activity remains a major concern. Guidelines have been proposed for return to activities; however, these fail to address the needs of younger children and those participating in more casual activities. Guidance is needed for primary care providers to facilitate safe return to everyday activity and sports and to help direct appropriate laboratory, electrocardiographic, and anatomical assessment. EVIDENCE ACQUISITION Review of computerized databases of available literature on SARS-CoV-2 infection in children and postinfection sequelae, risk factors for sudden cardiac death, and previous return to play recommendations. STUDY DESIGN Clinical guidelines based on available evidence and expert consensus. LEVEL OF EVIDENCE Level 4. RESULTS In this report, we review the literature on return to activity after SARS-CoV-2 infection and propose recommendations for cardiac clearance for children and adolescents. Though severe disease and cardiac injury is less common in children than in adults, it can occur. Several diagnostic modalities such as electrocardiography, echocardiography, cardiac magnetic resonance imaging, and serologic testing may be useful in the cardiac evaluation of children after SARS-CoV-2 infection. CONCLUSION Gradual return to activity is possible in most children and adolescents after SARS-CoV-2 infection and many of these patients can be cleared by their primary care providers. Providing education on surveillance for cardiopulmonary symptoms with return to sports can avoid unnecessary testing and delays in clearance.
Collapse
Affiliation(s)
- Devyani Chowdhury
- Devyani Chowdhury, MD, Cardiology Care for Children, Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, 1834 Oregon Pike 20, Lancaster, PA 17601 ()
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Simon V, Kota V, Bloomquist RF, Hanley HB, Forgacs D, Pahwa S, Pallikkuth S, Miller LG, Schaenman J, Yeaman MR, Manthei D, Wolf J, Gaur AH, Estepp JH, Srivastava K, Carreño JM, Cuevas F, Ellebedy AH, Gordon A, Valdez R, Cobey S, Reed EF, Kolhe R, Thomas PG, Schultz-Cherry S, Ross TM, Krammer F. PARIS and SPARTA: Finding the Achilles' Heel of SARS-CoV-2. mSphere 2022; 7:e0017922. [PMID: 35586986 PMCID: PMC9241545 DOI: 10.1128/msphere.00179-22] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 12/05/2022] Open
Abstract
To understand reinfection rates and correlates of protection for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we established eight different longitudinal cohorts in 2020 under the umbrella of the PARIS (Protection Associated with Rapid Immunity to SARS-CoV-2)/SPARTA (SARS SeroPrevalence And Respiratory Tract Assessment) studies. Here, we describe the PARIS/SPARTA cohorts, the harmonized assays and analysis that are performed across the cohorts, as well as case definitions for SARS-CoV-2 infection and reinfection that have been established by the team of PARIS/SPARTA investigators. IMPORTANCE Determining reinfection rates and correlates of protection against SARS-CoV-2 infection induced by both natural infection and vaccination is of high significance for the prevention and control of coronavirus disease 2019 (COVID-19). Furthermore, understanding reinfections or infection after vaccination and the role immune escape plays in these scenarios will inform the need for updates of the current SARS-CoV-2 vaccines and help update guidelines suitable for the postpandemic world.
Collapse
Affiliation(s)
- Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vamsi Kota
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Ryan F. Bloomquist
- Department of Restorative Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hannah B. Hanley
- Center for Vaccine and Immunology, University of Georgia, Athens, Georgia, USA
| | - David Forgacs
- Center for Vaccine and Immunology, University of Georgia, Athens, Georgia, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Loren G. Miller
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Joanna Schaenman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michael R. Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - David Manthei
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Joshua Wolf
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Aditya H. Gaur
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeremie H. Estepp
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Frans Cuevas
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - PARIS/SPARTA Study Group,
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Restorative Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois, USA
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ali H. Ellebedy
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Riccardo Valdez
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois, USA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Restorative Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Paul G. Thomas
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ted M. Ross
- Center for Vaccine and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
99
|
Wallace S, Hall V, Charlett A, Kirwan PD, Cole M, Gillson N, Atti A, Timeyin J, Foulkes S, Taylor-Kerr A, Andrews N, Shrotri M, Rokadiya S, Oguti B, Vusirikala A, Islam J, Zambon M, Brooks TJG, Ramsay M, Brown CS, Chand M, Hopkins S. Impact of prior SARS-CoV-2 infection and COVID-19 vaccination on the subsequent incidence of COVID-19: a multicentre prospective cohort study among UK healthcare workers - the SIREN (Sarscov2 Immunity & REinfection EvaluatioN) study protocol. BMJ Open 2022; 12:e054336. [PMID: 35768083 PMCID: PMC9240450 DOI: 10.1136/bmjopen-2021-054336] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Understanding the effectiveness and durability of protection against SARS-CoV-2 infection conferred by previous infection and COVID-19 is essential to inform ongoing management of the pandemic. This study aims to determine whether prior SARS-CoV-2 infection or COVID-19 vaccination in healthcare workers protects against future infection. METHODS AND ANALYSIS This is a prospective cohort study design in staff members working in hospitals in the UK. At enrolment, participants are allocated into cohorts, positive or naïve, dependent on their prior SARS-CoV-2 infection status, as measured by standardised SARS-CoV-2 antibody testing on all baseline serum samples and previous SARS-CoV-2 test results. Participants undergo monthly antibody testing and fortnightly viral RNA testing during follow-up and based on these results may move between cohorts. Any results from testing undertaken for other reasons (eg, symptoms, contact tracing) or prior to study entry will also be captured. Individuals complete enrolment and fortnightly questionnaires on exposures, symptoms and vaccination. Follow-up is 12 months from study entry, with an option to extend follow-up to 24 months.The primary outcome of interest is infection with SARS-CoV-2 after previous SARS-CoV-2 infection or COVID-19 vaccination during the study period. Secondary outcomes include incidence and prevalence (both RNA and antibody) of SARS-CoV-2, viral genomics, viral culture, symptom history and antibody/neutralising antibody titres. ETHICS AND DISSEMINATION The study was approved by the Berkshire Research Ethics Committee, Health Research Authority (IRAS ID 284460, REC reference 20/SC/0230) on 22 May 2020; the vaccine amendment was approved on 12 January 2021. Participants gave informed consent before taking part in the study.Regular reports to national and international expert advisory groups and peer-reviewed publications ensure timely dissemination of findings to inform decision making. TRIAL REGISTRATION NUMBER ISRCTN11041050.
Collapse
Affiliation(s)
- Sarah Wallace
- National Infection Service, UK Health Security Agency, London, UK
| | - Victoria Hall
- National Infection Service, UK Health Security Agency, London, UK
| | - Andre Charlett
- Statistics, Modelling and Economics Unit, UK Health Security Agency, London, UK
| | - Peter D Kirwan
- National Infection Service, UK Health Security Agency, London, UK
- MRC Biostatistics Unit, Cambridge, UK
| | - Michele Cole
- National Infection Service, UK Health Security Agency, London, UK
| | - Natalie Gillson
- National Infection Service, UK Health Security Agency, London, UK
| | - Ana Atti
- National Infection Service, UK Health Security Agency, London, UK
| | - Jean Timeyin
- National Infection Service, UK Health Security Agency, London, UK
| | - Sarah Foulkes
- National Infection Service, UK Health Security Agency, London, UK
| | | | - Nick Andrews
- Statistics, Modelling and Economics Unit, UK Health Security Agency, London, UK
| | | | - Sakib Rokadiya
- National Infection Service, UK Health Security Agency, London, UK
| | - Blanche Oguti
- National Infection Service, UK Health Security Agency, London, UK
| | | | - Jasmin Islam
- National Infection Service, UK Health Security Agency, London, UK
| | - Maria Zambon
- National Infection Service, UK Health Security Agency, London, UK
| | - Tim J G Brooks
- National Infection Service, UK Health Security Agency, London, UK
| | - Mary Ramsay
- National Infection Service, UK Health Security Agency, London, UK
| | - Colin S Brown
- National Infection Service, UK Health Security Agency, London, UK
| | - Meera Chand
- National Infection Service, UK Health Security Agency, London, UK
| | - Susan Hopkins
- National Infection Service, UK Health Security Agency, London, UK
| |
Collapse
|
100
|
Yuguero O, Companys M, Guzmán M, Maciel R, Llobet C, López A, Olles R, Pujol V, Ruiz MJ, Saura M, Vidal C, Godoy P. Epidemiological and clinical characteristics of SARS-CoV-2 reinfections in a Spanish region. SAGE Open Med 2022; 10:20503121221108556. [PMID: 35784672 PMCID: PMC9244920 DOI: 10.1177/20503121221108556] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/04/2022] [Indexed: 11/15/2022] Open
Abstract
Objective: We aimed to assess the prevalence and clinical characteristics of SARS-CoV-2 reinfections in a Spanish region. Methods: This is a retrospective observational study in all patients with SARS-CoV-2 infections in the Lleida health region from 1 March to 30 November 2020. Reinfections were classified as patients with positive SARS-CoV-2 PCR tests separated by at least 90 days plus a negative test result between both infection episodes. Primary and secondary outcomes: The primary outcome was the percentage of SARS-CoV-2 reinfections among all SARS-CoV-2 infections detected during our study period. Secondary outcomes were the clinical and sociodemographic characteristics of patients with SARS-CoV-2 reinfections. Results: Of the 27,758 patients diagnosed with SARS-CoV-2 infection in the study period, 14 were identified as coronavirus reinfection (0.050%). Of the reinfected sample, 12 patients (85.7%) were women. The median age was 41.5 years. Two patients died in the second coronavirus episode. Conclusion: The reinfection rate of SARS-CoV-2 in the Spanish region Lleida was relatively low during the observational period in 2020 (less than 1%). These data are in line with the notion that previous SARS-CoV-2 infections may offer a significant protection by so called natural immunity.
Collapse
Affiliation(s)
- Oriol Yuguero
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
- Faculty of Medicine, University of Lleida, Lleida, Spain
| | - Maria Companys
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Marianela Guzmán
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Rita Maciel
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Cecília Llobet
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Aurora López
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Rebeca Olles
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Violant Pujol
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Maria José Ruiz
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Mireia Saura
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Carmel Vidal
- ER LAB, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Pere Godoy
- Faculty of Medicine, University of Lleida, Lleida, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|