151
|
Dudek I, Jesiotr M, Rzeszotarska A, Kłos K, Chciałowski A, Nowak M, Korsak J. A 63-Year-Old Man with a Diagnosis of Re-Infection with SARS-CoV-2 Nine Weeks After an Initial Hospital Admission with COVID-19 Pneumonia. AMERICAN JOURNAL OF CASE REPORTS 2022; 23:e932999. [PMID: 35073284 PMCID: PMC8800480 DOI: 10.12659/ajcr.932999] [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: 05/04/2021] [Revised: 12/08/2021] [Accepted: 11/09/2021] [Indexed: 12/04/2022]
Abstract
BACKGROUND This report describes a 63-year-old Polish man presenting with COVID-19 (Coronavirus Disease 2019) pneumonia in early 2020, before vaccines to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were available. Nine weeks following recovery from the initial infection, he tested positive again for SARS-CoV-2. CASE REPORT Man, age 63, was admitted to the Military Institute of Medicine on March 12, 2020, with body temperature 40°C, a cough, and breathlessness. On March 12, 2020, SARS-CoV-2 RNA was found in a nasopharynx smear. A chest X-ray (RTG) showed discrete areas of interstitial densities. On June 13, 2020, after 32 days of hospitalization and 2 negative real-time polymerase chain rection (RT-PCR) test results, patient was released home in good general condition. On July 23, 2020 he reported to the emergency room with fever of 39°C and general weakness. A nasopharynx smear confirmed SARS-CoV-2 infection. On admission, the patient was in moderately good condition with auscultatory changes typical for pneumonia on both sides of the chest. On the seventh day of hospitalization, the patient was transported to the Intensive Care Unit (ICU) due to drastic deterioration in respiratory function. Respiratory support with non-invasive high-flow oxygen therapy (Opti-Flow) was used. On August 20, 2020, after negative RT-PCR test results, he was discharged in good general condition. CONCLUSIONS This case of COVID-19 pneumonia presented early in the COVID-19 pandemic of 2020, and the laboratory diagnosis of the initial and subsequent SARS-CoV-2 infection relied on the laboratory methods available at that time. However, several cases of repeat SARS-CoV-2 infection have been described before the development of vaccines in late 2020.
Collapse
Affiliation(s)
- Ilona Dudek
- Department of Clinical Transfusiology, Military Institute of Medicine, Warsaw, Poland
| | - Marzena Jesiotr
- Department of Clinical Transfusiology, Military Institute of Medicine, Warsaw, Poland
| | | | - Krzysztof Kłos
- Department of Infectious Diseases and Allergology, Military Institute of Medicine, Warsaw, Poland
| | - Andrzej Chciałowski
- Department of Infectious Diseases and Allergology, Military Institute of Medicine, Warsaw, Poland
| | - Monika Nowak
- Department of Clinical Transfusiology, Military Institute of Medicine, Warsaw, Poland
| | - Jolanta Korsak
- Department of Clinical Transfusiology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
152
|
El-Shabasy RM, Nayel MA, Taher MM, Abdelmonem R, Shoueir KR, Kenawy ER. Three waves changes, new variant strains, and vaccination effect against COVID-19 pandemic. Int J Biol Macromol 2022; 204:161-168. [PMID: 35074332 PMCID: PMC8782737 DOI: 10.1016/j.ijbiomac.2022.01.118] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/12/2022]
Abstract
It has been more than one year since the first case of the coronaviruses was infected by COVID-19 in China. The world witnessed three waves of the corona virus till now, and more upcoming is expected, whereas several challenges are presented. Empirical data displayed that the features of the virus effects do vary between the three periods. The severity of the disease, differences in symptoms, attitudes of the people have been reported, although the comparative characteristics of the three waves still keep essentially indefinite. In contrast, the sense of danger toward the cries gradually decreases in most countries. This may be due to some factors, including the approved vaccines, introducing alternative plans from politicians to control and deal with the epidemic, and decreasing the mortality rates. However, the alarm voice started to rise again with the appearance of new variant strains with several mutations in the virus. Several more questions began to be asked without sufficient answers. Mutations in COVID-19 have introduced an extreme challenge in preventing and treating SARS-COV-2. The essential feature for mutations is producing new variants known by high tensmibility, disturbing the viral fitness, and enhancing the virus replication. One of the variants that has emerged recently is the Delta variant (B.1.617.2), which was firstly detected in India. In November 2021, a more ferocious mutant appeared in South Africa, also called omicron (B.1.1.529). These mutants grabbed world attention because of their higher transmissibility than the progenitor variants and spread rapidly. Several information about the virus are still confusing and remains secret. There are eight approved vaccines in the market; however, the investigation race about their effect against reinfection and their role against the new variants is still under investigation. Furthermore, this is the first time vaccinating against COVID-19, so the question remains: Will we need an annual dose of the corona vaccines, and the side effects don't been observed till now?
Collapse
Affiliation(s)
- Rehan M El-Shabasy
- Department of Chemistry, Faculty of Science, Menoufia University, 32512 Shebin El-Kom, Egypt.
| | - Mohamed A Nayel
- Department of Animal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Menoufia, Egypt
| | - Mohamed M Taher
- Department of Chemistry, Faculty of Science, Cairo University, 12613 Giza, Egypt.
| | - Rehab Abdelmonem
- Department of Industrial Pharmacy, Faculty of Pharmacy, Misr University for Science & Technology, 6th October, Egypt
| | - Kamel R Shoueir
- Institute of Nanoscience & Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt; Institut de Chimie et Procédés pour l'Énergie, l'Environnement et la Santé (ICPEES), CNRS UMR 7515-Université de Strasbourg, 25 rue Becquerel, 67087 Strasbourg, France
| | - El Refaie Kenawy
- Polymer Research Group, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
153
|
Molecularly imprinted polypyrrole based sensor for the detection of SARS-CoV-2 spike glycoprotein. Electrochim Acta 2022; 403:139581. [PMID: 34898691 PMCID: PMC8643074 DOI: 10.1016/j.electacta.2021.139581] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 12/25/2022]
Abstract
This study describes the application of a polypyrrole-based sensor for the determination of SARS-CoV-2-S spike glycoprotein. The SARS-CoV-2-S spike glycoprotein is a spike protein of the coronavirus SARS-CoV-2 that recently caused the worldwide spread of COVID-19 disease. This study is dedicated to the development of an electrochemical determination method based on the application of molecularly imprinted polymer technology. The electrochemical sensor was designed by molecular imprinting of polypyrrole (Ppy) with SARS-CoV-2-S spike glycoprotein (MIP-Ppy). The electrochemical sensors with MIP-Ppy and with polypyrrole without imprints (NIP-Ppy) layers were electrochemically deposited on a platinum electrode surface by a sequence of potential pulses. The performance of polymer layers was evaluated by pulsed amperometric detection. According to the obtained results, a sensor based on MIP-Ppy is more sensitive to the SARS-CoV-2-S spike glycoprotein than a sensor based on NIP-Ppy. Also, the results demonstrate that the MIP-Ppy layer is more selectively interacting with SARS-CoV-2-S glycoprotein than with bovine serum albumin. This proves that molecularly imprinted MIP-Ppy-based sensors can be applied for the detection of SARS-CoV-2 virus proteins.
Collapse
|
154
|
Mao Y, Wang W, Ma J, Wu S, Sun F. Reinfection rates among patients previously infected by SARS-CoV-2: systematic review and meta-analysis. Chin Med J (Engl) 2022; 135:145-152. [PMID: 34908003 PMCID: PMC8769121 DOI: 10.1097/cm9.0000000000001892] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Asymptomatic or symptomatic infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can be followed by reinfection. The protection conferred by prior infection among coronavirus disease 2019 (COVID-19) patients is unclear. We assessed the incidence of SARS-CoV-2 reinfection and the protection effect of previous infection against reinfection. METHODS We searched PubMed, EMBASE, Cochrane, Scopus, Web of Science, and ClinicalTrials.gov for publications up until the end date of May 1, 2021. The reinfection rate of recovered patients and the protection against reinfection were analyzed using meta-analysis. RESULTS Overall, 19 studies of 1096 reinfection patients were included. The pooled reinfection rate was 0.65% (95% confidence interval [CI] 0.39-0.98%). The symptomatic reinfection rate was a bit lower (0.37% [95% CI 0.11-0.78%], I2 = 99%). The reinfection rate was much higher in high-risk populations (1.59% [95% CI 0.30-3.88%], I2 = 90%). The protection against reinfection and symptomatic reinfection was similar (87.02% [95% CI 83.22-89.96%] and 87.17% [95% CI 83.09-90.26%], respectively). CONCLUSIONS The rate of reinfection with SARS-CoV-2 is relatively low. The protection against SARS-CoV-2 after natural infection is comparable to that estimated for vaccine efficacy. These data may help guide public health measures and vaccination strategies in response to the COVID-19 pandemic. High-quality clinical studies are needed to establish the relevant risk factors in recovered patients.
Collapse
Affiliation(s)
- Yinjun Mao
- Department of Pharmacy, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Weiwei Wang
- National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Jun Ma
- Institute of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Shanshan Wu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing 100191, China
| |
Collapse
|
155
|
Iwata-Yoshikawa N, Shiwa N, Sekizuka T, Sano K, Ainai A, Hemmi T, Kataoka M, Kuroda M, Hasegawa H, Suzuki T, Nagata N. A lethal mouse model for evaluating vaccine-associated enhanced respiratory disease during SARS-CoV-2 infection. SCIENCE ADVANCES 2022; 8:eabh3827. [PMID: 34995117 PMCID: PMC8741184 DOI: 10.1126/sciadv.abh3827] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 11/12/2021] [Indexed: 05/02/2023]
Abstract
One safety concern during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine development has been the vaccine-associated enhanced disease, which is characterized by eosinophilic immunopathology and T helper cell type 2 (TH2)–biased immune responses with insufficient neutralizing antibodies. In this study, we established a lethal animal model using BALB/c mice and a mouse-passaged isolate (QHmusX) from a European lineage of SARS-CoV-2. The QHmusX strain induced acute respiratory illness, associated with diffuse alveolar damage and pulmonary edema, in TH2-prone adult BALB/c mice, but not in young mice or TH1-prone C57BL/6 mice. We also showed that immunization of adult BALB/c mice with recombinant spike protein without appropriate adjuvant caused eosinophilic immunopathology with TH2-shifted immune response and insufficient neutralizing antibodies after QHmusX infection. This lethal mouse model is useful for evaluating vaccine-associated enhanced respiratory disease during SARS-CoV-2 infection and may provide new insights into the disease pathogenesis of SARS-CoV-2.
Collapse
Affiliation(s)
- Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| | - Nozomi Shiwa
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomics Center, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Kaori Sano
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| | - Takuya Hemmi
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
- Department of Biological Science and Technology, Tokyo University of Science, 125-8585 Tokyo, Japan
| | - Michiyo Kataoka
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| | - Makoto Kuroda
- Pathogen Genomics Center, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Hideki Hasegawa
- Influenza Virus Research Center, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, 208-0011 Tokyo, Japan
| |
Collapse
|
156
|
Khalili-Tanha G, Khazaei M, Soleimanpour S, Ferns GA, Avan A. The chance of COVID-19 infection after vaccination. Infect Disord Drug Targets 2022; 22:e050122199980. [PMID: 34986778 DOI: 10.2174/1871526522666220105113829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/15/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
The outbreak of COVID-19 that began in Wuhan, China, has constituted a new emerging epidemic that has spread around the world. There are some reports on illustrated the patients getting reinfected after recovering from COVID-19. Here we provide an overview of the biphasic cycle of COVID-19, genetic diversity, immune response and chance of reinfection after recovering from COVID-19. The new generation of COVID-19 is highly contagious and pathogenic infection can lead to acute respiratory distress syndrome. Whilst most patients suffer from a mild form of the disease, there is a rising concern that patients who recover from COVID-19 may be at risk of reinfection. The proportion of the infected population, is increasing worldwide; meanwhile, the rate and concern of reinfection by the recovered population are still high. Moreover, there are a few evidence on the chance of COVID-19 infection even after vaccination, which is around one per cent or less. Although the hypothesis of zero reinfections after vaccination has not been clinically proven, further studies should be performed on the recovered class in clusters to study the progression of the exposed with the re-exposed subpopulations to estimate the possibilities of reinfection and, thereby, advocate the use of these antibodies for vaccine creation.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
157
|
Stankiewicz Karita HC, Dong TQ, Johnston C, Neuzil KM, Paasche-Orlow MK, Kissinger PJ, Bershteyn A, Thorpe LE, Deming M, Kottkamp A, Laufer M, Landovitz RJ, Luk A, Hoffman R, Roychoudhury P, Magaret CA, Greninger AL, Huang ML, Jerome KR, Wener M, Celum C, Chu HY, Baeten JM, Wald A, Barnabas RV, Brown ER. Trajectory of Viral RNA Load Among Persons With Incident SARS-CoV-2 G614 Infection (Wuhan Strain) in Association With COVID-19 Symptom Onset and Severity. JAMA Netw Open 2022; 5:e2142796. [PMID: 35006245 PMCID: PMC8749477 DOI: 10.1001/jamanetworkopen.2021.42796] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPORTANCE The SARS-CoV-2 viral trajectory has not been well characterized in incident infections. These data are needed to inform natural history, prevention practices, and therapeutic development. OBJECTIVE To characterize early SARS-CoV-2 viral RNA load (hereafter referred to as viral load) in individuals with incident infections in association with COVID-19 symptom onset and severity. DESIGN, SETTING, AND PARTICIPANTS This prospective cohort study was a secondary data analysis of a remotely conducted study that enrolled 829 asymptomatic community-based participants recently exposed (<96 hours) to persons with SARS-CoV-2 from 41 US states from March 31 to August 21, 2020. Two cohorts were studied: (1) participants who were SARS-CoV-2 negative at baseline and tested positive during study follow-up, and (2) participants who had 2 or more positive swabs during follow-up, regardless of the initial (baseline) swab result. Participants collected daily midturbinate swab samples for SARS-CoV-2 RNA detection and maintained symptom diaries for 14 days. EXPOSURE Laboratory-confirmed SARS-CoV-2 infection. MAIN OUTCOMES AND MEASURES The observed SARS-CoV-2 viral load among incident infections was summarized, and piecewise linear mixed-effects models were used to estimate the characteristics of viral trajectories in association with COVID-19 symptom onset and severity. RESULTS A total of 97 participants (55 women [57%]; median age, 37 years [IQR, 27-52 years]) developed incident infections during follow-up. Forty-two participants (43%) had viral shedding for 1 day (median peak viral load cycle threshold [Ct] value, 38.5 [95% CI, 38.3-39.0]), 18 (19%) for 2 to 6 days (median Ct value, 36.7 [95% CI, 30.2-38.1]), and 31 (32%) for 7 days or more (median Ct value, 18.3 [95% CI, 17.4-22.0]). The cycle threshold value has an inverse association with viral load. Six participants (6%) had 1 to 6 days of viral shedding with censored duration. The peak mean (SD) viral load was observed on day 3 of shedding (Ct value, 33.8 [95% CI, 31.9-35.6]). Based on the statistical models fitted to 129 participants (60 men [47%]; median age, 38 years [IQR, 25-54 years]) with 2 or more SARS-CoV-2-positive swab samples, persons reporting moderate or severe symptoms tended to have a higher peak mean viral load than those who were asymptomatic (Ct value, 23.3 [95% CI, 22.6-24.0] vs 30.7 [95% CI, 29.8-31.4]). Mild symptoms generally started within 1 day of peak viral load, and moderate or severe symptoms 2 days after peak viral load. All 535 sequenced samples detected the G614 variant (Wuhan strain). CONCLUSIONS AND RELEVANCE This cohort study suggests that having incident SARS-CoV-2 G614 infection was associated with a rapid viral load peak followed by slower decay. COVID-19 symptom onset generally coincided with peak viral load, which correlated positively with symptom severity. This longitudinal evaluation of the SARS-CoV-2 G614 with frequent molecular testing serves as a reference for comparing emergent viral lineages to inform clinical trial designs and public health strategies to contain the spread of the virus.
Collapse
Affiliation(s)
| | - Tracy Q. Dong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Christine Johnston
- Division of Allergy and Infectious Diseases, University of Washington, Seattle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Kathleen M. Neuzil
- Department of Medicine, University of Maryland School of Medicine, Baltimore
| | - Michael K. Paasche-Orlow
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Medicine, Boston Medical Center, Boston, Massachusetts
| | | | - Anna Bershteyn
- Department of Population Health, New York University Grossman School of Medicine, New York
| | - Lorna E. Thorpe
- Department of Population Health, New York University Grossman School of Medicine, New York
| | - Meagan Deming
- Department of Medicine, University of Maryland School of Medicine, Baltimore
| | - Angelica Kottkamp
- Department of Medicine, New York University Grossman School of Medicine, New York
| | - Miriam Laufer
- Department of Medicine, University of Maryland School of Medicine, Baltimore
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore
| | | | - Alfred Luk
- Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Risa Hoffman
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore
| | - Pavitra Roychoudhury
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Craig A. Magaret
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Alexander L. Greninger
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Meei-Li Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Keith R. Jerome
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Mark Wener
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
- Division of Rheumatology, University of Washington, Seattle
| | - Connie Celum
- Division of Allergy and Infectious Diseases, University of Washington, Seattle
- Department of Global Health, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Helen Y. Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle
- Department of Global Health, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Jared M. Baeten
- Division of Allergy and Infectious Diseases, University of Washington, Seattle
- Department of Global Health, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Anna Wald
- Division of Allergy and Infectious Diseases, University of Washington, Seattle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Ruanne V. Barnabas
- Division of Allergy and Infectious Diseases, University of Washington, Seattle
- Department of Global Health, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Elizabeth R. Brown
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Biostatistics, University of Washington, Seattle
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
158
|
Coppola A, Buonerba C, Cardinale D, Lo Conte G, Sansone D, Rofrano G, De Vita S, Morgante M, Triassi M, Atripaldi L, Brambilla G, Sabatino R, Pierri A, Pacella D, Pizzolante A, Pierri B, Ferrucci V, Zollo M, Capasso M, Stringhini S, Ascierto PA, Roperto S, Cerino P. Durability of Humoral Immune Responses to SARS-CoV-2 in Citizens of Ariano Irpino (Campania, Italy): A Longitudinal Observational Study With an 11.5-Month Follow-Up. Front Public Health 2022; 9:801609. [PMID: 34976939 PMCID: PMC8718636 DOI: 10.3389/fpubh.2021.801609] [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: 10/25/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
As of November 17, 2021, SARS-CoV-2 (Severe Acute Respiratory Syndrome CoronaVirus 2), the causative agent of COVID-19 (COronaVIrus Disease 19), has infected ~250 million people worldwide, causing around five million deaths. Titers of anti-SARS-CoV-2 neutralizing antibodies were relatively stable for at least 9 months in a population-based study conducted in Wuhan, China, both in symptomatic and in asymptomatic individuals. In the mass screening campaign conducted in the town of Ariano Irpino (Avellino, Italy) in May, 2020, 5.7% (95% CI: 5.3-6-1) of the 13,444 asymptomatic citizens screened were positive for anti-nucleocapsid antibodies against SARS-CoV-2. Among these, 422 citizens were re-tested for anti SARS-CoV-2 antibodies in January, 2021 and/or in April, 2021 and enrolled in this longitudinal observational study. Median (interquartile range) age of the study cohort was 46 years (29–59), with 47 (11.1%) participants of minor age, while 217 (51.4%) participants were females. There was no evidence of re-infection in any of the subjects included. Presence of anti-nuclear antibodies antibodies (Elecysis, Roche) was reported in 95.7 and 93.7% of evaluable participants in January and April, 2021. Multiple logistic regression analysis used to explore associations between age, sex and seroprevalence showed that adults vs. minors had significantly lower odds of having anti-S1 antibodies (Biorad) both in January, 2021 and in April, 2021. Our findings showed that antibodies remained detectable at least 11.5 months after infection in >90% of never symptomatic cases. Further investigation is required to establish duration of immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Annachiara Coppola
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy.,Dipartimento di Medicina Sperimentale, Universita' degli studi della Campania "L. Vanvitelli", Naples, Italy
| | - Carlo Buonerba
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Davide Cardinale
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Gabriella Lo Conte
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Donato Sansone
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Giuseppe Rofrano
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Sabato De Vita
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | | | - Maria Triassi
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | | | - Gianfranco Brambilla
- Istituto Superiore di Sanità, Food Safety, Nutrition, and Veterinary Public Health Department, Rome, Italy
| | - Rocco Sabatino
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy.,Cotugno Hospital, AORN Ospedali dei Colli, Naples, Italy
| | - Andrea Pierri
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Daniela Pacella
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Antonio Pizzolante
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Biancamaria Pierri
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | | | | | | | - Silvia Stringhini
- Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy & Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Sante Roperto
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università di Napoli Federico II, Naples, Italy
| | - Pellegrino Cerino
- Centro di Referenza Nazionale per l'Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| |
Collapse
|
159
|
Tanunliong G, Liu A, Vijh R, Pidduck T, Kustra J, Márquez AC, Choi A, McLennan M, Hayden A, Kearney C, Gantt S, Krajden M, Morshed M, Jassem AN, Sekirov I. Persistence of Anti-SARS-CoV-2 Antibodies in Long Term Care Residents Over Seven Months After Two COVID-19 Outbreaks. Front Immunol 2022; 12:775420. [PMID: 35046939 PMCID: PMC8763385 DOI: 10.3389/fimmu.2021.775420] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023] Open
Abstract
Background As part of the public health outbreak investigations, serological surveys were carried out following two COVID-19 outbreaks in April 2020 and October 2020 in one long term care facility (LTCF) in British Columbia, Canada. This study describes the serostatus of the LTCF residents and monitors changes in their humoral response to SARS-CoV-2 and other human coronaviruses (HCoV) over seven months. Methods A total of 132 serum samples were collected from all 106 consenting residents (aged 54-102) post-first outbreak (N=87) and post-second outbreak (N=45) in one LTCF; 26/106 participants provided their serum following both COVID-19 outbreaks, permitting longitudinal comparisons between surveys. Health-Canada approved commercial serologic tests and a pan-coronavirus multiplexed immunoassay were used to evaluate antibody levels against the spike protein, nucleocapsid, and receptor binding domain (RBD) of SARS-CoV-2, as well as the spike proteins of HCoV-229E, HCoV-HKU1, HCoV-NL63, and HCoV-OC43. Statistical analyses were performed to describe the humoral response to SARS-CoV-2 among residents longitudinally. Findings Survey findings demonstrated that among the 26 individuals that participated in both surveys, all 10 individuals seropositive after the first outbreak continued to be seropositive following the second outbreak, with no reinfections identified among them. SARS-CoV-2 attack rate in the second outbreak was lower (28.6%) than in the first outbreak (40.2%), though not statistically significant (P>0.05). Gradual waning of anti-nucleocapsid antibodies to SARS-CoV-2 was observed on commercial (median Δ=-3.7, P=0.0098) and multiplexed immunoassay (median Δ=-169579, P=0.014) platforms; however, anti-spike and anti-receptor binding domain (RBD) antibodies did not exhibit a statistically significant decline over 7 months. Elevated antibody levels for beta-HCoVs OC43 (P<0.0001) and HKU1 (P=0.0027) were observed among individuals seropositive for SARS-CoV-2 compared to seronegative individuals. Conclusion Our study utilized well-validated serological platforms to demonstrate that humoral responses to SARS-CoV-2 persisted for at least 7 months. Elevated OC43 and HKU1 antibodies among SARS-CoV-2 seropositive individuals may be attributed to cross reaction and/or boosting of humoral response.
Collapse
Affiliation(s)
- Guadalein Tanunliong
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Aaron Liu
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rohit Vijh
- Office of the Chief Medical Health Officer, Vancouver Coastal Health, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Tamara Pidduck
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| | - Jesse Kustra
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| | - Ana Citlali Márquez
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| | - Alexandra Choi
- Office of the Chief Medical Health Officer, Vancouver Coastal Health, Vancouver, BC, Canada
| | - Meghan McLennan
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| | - Althea Hayden
- Office of the Chief Medical Health Officer, Vancouver Coastal Health, Vancouver, BC, Canada
| | | | - Soren Gantt
- Centre de Recherche de Centre Hospitalier Universitaire (CHU) Sainte-Justine, Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Mel Krajden
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| | - Muhammad Morshed
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| | - Agatha N. Jassem
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| | - Inna Sekirov
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control (BCCDC) Public Health Laboratory, Vancouver, BC, Canada
| |
Collapse
|
160
|
Abstract
Adaptive immune responses play critical roles in viral clearance and protection against re-infection, and SARS-CoV-2 is no exception. What is exceptional is the rapid characterization of the immune response to the virus performed by researchers during the first 20 months of the pandemic. This has given us a more detailed understanding of SARS-CoV-2 compared to many viruses that have been with us for a long time. Furthermore, effective COVID-19 vaccines were developed in record time, and their rollout worldwide is already making a significant difference, although major challenges remain in terms of equal access. The pandemic has engaged scientists and the public alike, and terms such as seroprevalence, neutralizing antibodies, antibody escape and vaccine certificates have become familiar to a broad community. Here, we review key findings concerning B cell and antibody (Ab) responses to SARS-CoV-2, focusing on non-severe cases and anti-spike (S) Ab responses in particular, the latter being central to protective immunity induced by infection or vaccination. The emergence of viral variants that have acquired mutations in S acutely highlights the need for continued characterization of both emerging variants and Ab responses against these during the evolving pathogen-immune system arms race.
Collapse
Affiliation(s)
- Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
161
|
Shenoy S. Gut microbiome, Vitamin D, ACE2 interactions are critical factors in immune-senescence and inflammaging: key for vaccine response and severity of COVID-19 infection. Inflamm Res 2022; 71:13-26. [PMID: 34738147 PMCID: PMC8568567 DOI: 10.1007/s00011-021-01510-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The SARS-CoV-2 pandemic continues to spread sporadically in the Unites States and worldwide. The severity and mortality excessively affected the frail elderly with co-existing medical diseases. There is growing evidence that cross-talk between the gut microbiome, Vitamin D and RAS/ACE2 system is essential for a balanced functioning of the elderly immune system and in regulating inflammation. In this review, we hypothesize that the state of gut microbiome, prior to infection determines the outcome associated with COVID-19 sepsis and may also be a critical factor in success to vaccination. METHODS Articles from PubMed/Medline searches were reviewed using a combination of terms "SARS-CoV-2, COVID-19, Inflammaging, Immune-senescence, Gut microbiome, Vitamin D, RAS/ACE2, Vaccination". CONCLUSION Evidence indicates a complex association between gut microbiota, ACE-2 expression and Vitamin D in COVID-19 severity. Status of gut microbiome is highly predictive of the blood molecular signatures and inflammatory markers and host responses to infection. Vitamin D has immunomodulatory function in innate and adaptive immune responses to viral infection. Anti-inflammatory functions of Vit D include regulation of gut microbiome and maintaining microbial diversity. It promotes growth of gut-friendly commensal strains of Bifida and Fermicutus species. In addition, Vitamin D is a negative regulator for expression of renin and interacts with the RAS/ ACE/ACE-2 signaling axis. Collectively, this triad may be the critical, link in determination of outcomes in SARS-CoV-2 infection. The presented data are empirical and informative. Further research using advanced systems biology techniques and artificial intelligence-assisted integration could assist with correlation of the gut microbiome with sepsis and vaccine responses. Modulating these factors may impact in guiding the success of vaccines and clinical outcomes in COVID-19 infections.
Collapse
Affiliation(s)
- Santosh Shenoy
- Department of Surgery, Kansas City VA Medical Center, University of Missouri Kansas City, 4801 E Linwood Blvd., Kansas City , MO, 64128, USA.
| |
Collapse
|
162
|
Kumar A, Krishna A, Singh P, Patel P. Reinfection of SARS-CoV-2 in kidney transplant recipient. INDIAN JOURNAL OF TRANSPLANTATION 2022. [DOI: 10.4103/ijot.ijot_74_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
163
|
Blain H, Tuaillon E, Gamon L, Pisoni A, Miot S, Rolland Y, Picot M, Bousquet J. Antibody response after one and two jabs of the BNT162b2 vaccine in nursing home residents: The CONsort-19 study. Allergy 2022; 77:271-281. [PMID: 34286856 PMCID: PMC8441741 DOI: 10.1111/all.15007] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022]
Abstract
Background The humoral immune response following COVID‐19 vaccination in nursing home residents is poorly known. A longitudinal study compared levels of IgG antibodies against the spike protein (S‐RBD IgG) (S‐RDB protein IgG) after one and two BNT162b2/Pfizer jabs in residents with and without prior COVID‐19. Methods In 22 French nursing homes, COVID‐19 was diagnosed with real‐time reverse‐transcriptase polymerase chain reaction (RT‐PCR) for SARS‐CoV‐2. Blood S‐RDB‐protein IgG and nucleocapsid (N) IgG protein (N‐protein IgG) were measured 21–24 days after the first jab (1,004 residents) and 6 weeks after the second (820 residents). Results In 735 residents without prior COVID‐19, 41.7% remained seronegative for S‐RDB‐protein IgG after the first jab vs. 2.1% of the 270 RT‐PCR‐positive residents (p < 0.001). After the second jab, 3% of the 586 residents without prior COVID‐19 remained seronegative. However, 26.5% had low S‐RDB‐protein IgG levels (50–1050 UA/ml) vs. 6.4% of the 222 residents with prior COVID‐19. Residents with an older infection (first wave), or with N‐protein IgG at the time of vaccination, had the highest S‐RDB‐protein IgG levels. Residents with a prior COVID‐19 infection had higher S‐RDB‐protein IgG levels after one jab than those without after two jabs. Interpretation A single vaccine jab is sufficient to reach a high humoral immune response in residents with prior COVID‐19. Most residents without prior COVID‐19 are seropositive for S‐RDB‐protein IgG after the second jab, but around 30% have low levels. Whether residents with no or low post‐vaccine S‐RDB protein IgG are at higher risk of symptomatic COVID‐19 requires further analysis.
Collapse
Affiliation(s)
- Hubert Blain
- Department of Internal Medicine and Geriatrics MUSE University Montpellier France
| | | | - Lucie Gamon
- Clinical research and epidemiology unit University hospital Montpellier France
| | | | - Stéphanie Miot
- Department of Internal Medicine and Geriatrics MUSE University Montpellier France
| | - Yves Rolland
- INSERM 1027 Gérontopôle de Toulouse Toulouse France
| | | | - Jean Bousquet
- Clinical research and epidemiology unit University hospital Montpellier France
- Department of Dermatology and Allergy Universitätsmedizin Berlin Germany
| |
Collapse
|
164
|
Abstract
After recovery from COVID-19, a person may become infected again due to reactivation of the virus inside the human body or reinfection with a genetically distinct mutant virus owing to reinfection. The COVID-19 reinfection has been recorded all around the world, albeit it is still uncommon. The reinfection with COVID-19 raises several questions about virus characteristics such as mutation, growth, functioning, and transmissibility, level and durability of immunity, diagnosis, therapy, and efficacy of vaccine(s) on genetically modified viruses and their durability and safety. This chapter focuses on various aspects of COVID-19 reinfection, including its severity, frequency, immunopathogenesis, immune responses, effect on vaccine development, Corona waves and herd immunity, management and prevention strategies. COVID-19 reinfections are often asymptomatic or mildly symptomatic, and are milder than first infections, with a few exceptions. The management of reinfection should be the same as the treatment of the first COVID-19 infection. The deep, extensive, rapid and real-time whole-genome sequencing studies, as well as an enhanced vaccination drive, and rigorous adherence to COVID-19 appropriate behavior, would be critical in limiting the severity of transmission and reinfection.
Collapse
|
165
|
Rodríguez-Grande C, Alcalá L, Estévez A, Sola-Campoy PJ, Buenestado-Serrano S, Martínez-Laperche C, Manuel de la Cueva V, Alonso R, Andrés-Zayas C, Adán-Jiménez J, Losada C, Rico-Luna C, Comas I, González-Candelas F, Catalán P, Muñoz P, Pérez-Lago L, García de Viedma D. Systematic Genomic and Clinical Analysis of Severe Acute Respiratory Syndrome Coronavirus 2 Reinfections and Recurrences Involving the Same Strain. Emerg Infect Dis 2022; 28:85-94. [PMID: 34843661 PMCID: PMC8714233 DOI: 10.3201/eid2801.211952] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Estimates of the burden of severe acute respiratory syndrome coronavirus 2 reinfections are limited by the scarcity of population-level studies incorporating genomic support. We conducted a systematic study of reinfections in Madrid, Spain, supported by genomic viral analysis and host genetic analysis, to cleanse laboratory errors and to discriminate between reinfections and recurrences involving the same strain. Among the 41,195 cases diagnosed (March 2020-March 2021), 93 (0.23%) had 2 positive reverse transcription PCR tests (55-346 days apart). After eliminating cases with specimens not stored, of suboptimal sequence quality, or belonging to different persons, we obtained valid data from 22 cases. Of those, 4 (0.01%) cases were recurrences involving the same strain; case-patients were 39-93 years of age, and 3 were immunosuppressed. Eighteen (0.04%) cases were reinfections; patients were 19-84 years of age, and most had no relevant clinical history. The second episode was more severe in 8 cases.
Collapse
|
166
|
Lund AJ, Wade KJ, Nikolakis ZL, Ivey KN, Perry BW, Pike HNC, Paull SH, Liu Y, Castoe TA, Pollock DD, Carlton EJ. Integrating genomic and epidemiologic data to accelerate progress toward schistosomiasis elimination. eLife 2022; 11:79320. [PMID: 36040013 PMCID: PMC9427098 DOI: 10.7554/elife.79320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
The global community has adopted ambitious goals to eliminate schistosomiasis as a public health problem, and new tools are needed to achieve them. Mass drug administration programs, for example, have reduced the burden of schistosomiasis, but the identification of hotspots of persistent and reemergent transmission threaten progress toward elimination and underscore the need to couple treatment with interventions that reduce transmission. Recent advances in DNA sequencing technologies make whole-genome sequencing a valuable and increasingly feasible option for population-based studies of complex parasites such as schistosomes. Here, we focus on leveraging genomic data to tailor interventions to distinct social and ecological circumstances. We consider two priority questions that can be addressed by integrating epidemiological, ecological, and genomic information: (1) how often do non-human host species contribute to human schistosome infection? and (2) what is the importance of locally acquired versus imported infections in driving transmission at different stages of elimination? These questions address processes that can undermine control programs, especially those that rely heavily on treatment with praziquantel. Until recently, these questions were difficult to answer with sufficient precision to inform public health decision-making. We review the literature related to these questions and discuss how whole-genome approaches can identify the geographic and taxonomic sources of infection, and how such information can inform context-specific efforts that advance schistosomiasis control efforts and minimize the risk of reemergence.
Collapse
Affiliation(s)
- Andrea J Lund
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado AnschutzAuroraUnited States
| | - Kristen J Wade
- Department of Biochemistry & Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
| | - Zachary L Nikolakis
- Department of Biology, University of Texas at ArlingtonArlingtonUnited States
| | - Kathleen N Ivey
- Department of Biology, University of Texas at ArlingtonArlingtonUnited States
| | - Blair W Perry
- Department of Biology, University of Texas at ArlingtonArlingtonUnited States
| | - Hamish NC Pike
- Department of Biochemistry & Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
| | - Sara H Paull
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado AnschutzAuroraUnited States
| | - Yang Liu
- Sichuan Centers for Disease Control and PreventionChengduChina
| | - Todd A Castoe
- Department of Biology, University of Texas at ArlingtonArlingtonUnited States
| | - David D Pollock
- Department of Biochemistry & Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
| | - Elizabeth J Carlton
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado AnschutzAuroraUnited States
| |
Collapse
|
167
|
Alefishat E, Jelinek HF, Mousa M, Tay GK, Alsafar HS. Immune response to SARS-CoV-2 Variants: A focus on severity, susceptibility, and preexisting immunity. J Infect Public Health 2022; 15:277-288. [PMID: 35074728 PMCID: PMC8757655 DOI: 10.1016/j.jiph.2022.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/16/2021] [Accepted: 01/09/2022] [Indexed: 01/08/2023] Open
Abstract
The heterogeneous phenotypes among patients with coronavirus disease 2019 (COVID-19) has drawn worldwide attention, especially those with severe symptoms without comorbid conditions. Immune responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative virus of COVID-19, occur mainly by the innate immune response via the interferon (IFN)-mediated pathways, and the adaptive immunity via the T lymphocyte and the antibody mediated pathways. The ability of the original Wuhan SARS-CoV-2 strain, and possibly more so with new emerging variants, to antagonize IFN-mediated antiviral responses can be behind the higher early viral load, higher transmissibility, and milder symptoms compared to SARS-CoV and are part of the continued clinical evolution of COVID-19. Since it first emerged, several variants of SARS-CoV-2 have been circulating worldwide. Variants that have the potential to elude natural or vaccine-mediated immunity are variants of concern. This review focuses on the main host factors that may explain the immune responses to SARS-CoV-2 and its variants in the context of susceptibility, severity, and preexisting immunity.
Collapse
Affiliation(s)
- Eman Alefishat
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Herbert F Jelinek
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Center of Heath Engineering Innovation, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Nuffield Department of Women's and Reproduction Health, Oxford University, Oxford, United Kingdom
| | - Guan K Tay
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Discipline of Psychiatry, Medical School, the University of Western Australia, Perth WA, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Habiba S Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Emirates Bio-Research Center, Ministry of Interior, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
168
|
Manuel-Silva J, Sonié P, Rafael A, Amorim-Alves L. Reinfection with SARS-CoV-2: An inconvenient truth? J Family Med Prim Care 2022; 11:366-369. [PMID: 35309639 PMCID: PMC8930163 DOI: 10.4103/jfmpc.jfmpc_1490_21] [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: 07/24/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 11/04/2022] Open
Abstract
Data show that antibody-related immunity against SARS-CoV-2 may not be long-lasting. We report two cases regarded as cured from COVID-19, which presented again with symptoms and a positive SARS-CoV-2 RT-PCR test. Case one, a 60-year-old male, had a biphasic presentation of symptoms compatible with COVID-19 infection, associated with a positive RT-PCR test. Case two, a 25-year-old female, had a first positive RT-PCR test during hospital screening, and months later a symptomatic presentation of COVID-19, associated with a positive RT-PCR test. All cases were immunocompetent. Anti-IgG-SARS-CoV-2 blood samples were negative in both. Elevation of analytical inflammatory markers suggested new infection in both cases. COVID-19 reinfection may be a differential diagnosis and primary care physicians should acknowledge it. Previously cured patients should be encouraged to comply with health public preventive measures.
Collapse
|
169
|
Zimba R, Romo ML, Kulkarni SG, Berry A, You W, Mirzayi C, Westmoreland DA, Parcesepe AM, Waldron L, Rane MS, Kochhar S, Robertson MM, Maroko AR, Grov C, Nash D. Patterns of SARS-CoV-2 Testing Preferences in a National Cohort in the United States: Latent Class Analysis of a Discrete Choice Experiment. JMIR Public Health Surveill 2021; 7:e32846. [PMID: 34793320 PMCID: PMC8722498 DOI: 10.2196/32846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/21/2021] [Accepted: 11/15/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Inadequate screening and diagnostic testing in the United States throughout the first several months of the COVID-19 pandemic led to undetected cases transmitting disease in the community and an underestimation of cases. Though testing supply has increased, maintaining testing uptake remains a public health priority in the efforts to control community transmission considering the availability of vaccinations and threats from variants. OBJECTIVE This study aimed to identify patterns of preferences for SARS-CoV-2 screening and diagnostic testing prior to widespread vaccine availability and uptake. METHODS We conducted a discrete choice experiment (DCE) among participants in the national, prospective CHASING COVID (Communities, Households, and SARS-CoV-2 Epidemiology) Cohort Study from July 30 to September 8, 2020. The DCE elicited preferences for SARS-CoV-2 test type, specimen type, testing venue, and result turnaround time. We used latent class multinomial logit to identify distinct patterns of preferences related to testing as measured by attribute-level part-worth utilities and conducted a simulation based on the utility estimates to predict testing uptake if additional testing scenarios were offered. RESULTS Of the 5098 invited cohort participants, 4793 (94.0%) completed the DCE. Five distinct patterns of SARS-CoV-2 testing emerged. Noninvasive home testers (n=920, 19.2% of participants) were most influenced by specimen type and favored less invasive specimen collection methods, with saliva being most preferred; this group was the least likely to opt out of testing. Fast-track testers (n=1235, 25.8%) were most influenced by result turnaround time and favored immediate and same-day turnaround time. Among dual testers (n=889, 18.5%), test type was the most important attribute, and preference was given to both antibody and viral tests. Noninvasive dual testers (n=1578, 32.9%) were most strongly influenced by specimen type and test type, preferring saliva and cheek swab specimens and both antibody and viral tests. Among hesitant home testers (n=171, 3.6%), the venue was the most important attribute; notably, this group was the most likely to opt out of testing. In addition to variability in preferences for testing features, heterogeneity was observed in the distribution of certain demographic characteristics (age, race/ethnicity, education, and employment), history of SARS-CoV-2 testing, COVID-19 diagnosis, and concern about the pandemic. Simulation models predicted that testing uptake would increase from 81.6% (with a status quo scenario of polymerase chain reaction by nasal swab in a provider's office and a turnaround time of several days) to 98.1% by offering additional scenarios using less invasive specimens, both viral and antibody tests from a single specimen, faster turnaround time, and at-home testing. CONCLUSIONS We identified substantial differences in preferences for SARS-CoV-2 testing and found that offering additional testing options would likely increase testing uptake in line with public health goals. Additional studies may be warranted to understand if preferences for testing have changed since the availability and widespread uptake of vaccines.
Collapse
Affiliation(s)
- Rebecca Zimba
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
- Department of Epidemiology and Biostatistics, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Matthew L Romo
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
- Department of Epidemiology and Biostatistics, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Sarah G Kulkarni
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Amanda Berry
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - William You
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Chloe Mirzayi
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
- Department of Epidemiology and Biostatistics, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Drew A Westmoreland
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Angela M Parcesepe
- Department of Maternal and Child Health, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, United States
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Levi Waldron
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
- Department of Epidemiology and Biostatistics, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Madhura S Rane
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Shivani Kochhar
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - McKaylee M Robertson
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Andrew R Maroko
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
- Department of Environmental, Occupational, and Geospatial Health Sciences, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Christian Grov
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
- Department of Community Health and Social Sciences, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Denis Nash
- Institute for Implementation Science in Population Health, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
- Department of Epidemiology and Biostatistics, CUNY Graduate School of Public Health & Health Policy, New York, NY, United States
| |
Collapse
|
170
|
Negi N, Maurya SP, Singh R, Das BK. An update on host immunity correlates and prospects of re-infection in COVID-19. Int Rev Immunol 2021; 41:367-392. [PMID: 34961403 PMCID: PMC8787841 DOI: 10.1080/08830185.2021.2019727] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/18/2021] [Accepted: 12/08/2021] [Indexed: 01/08/2023]
Abstract
Reinfection with SARS-CoV-2 is not frequent yet the incidence rate of it is increasing globally owing to the slow emergence of drift variants that pose a perpetual threat to vaccination strategies and have a greater propensity for disease reoccurrence. Long-term protection against SARS-CoV-2 reinfection relies on the induction of the innate as well as the adaptive immune response endowed with immune memory. However, a multitude of factors including the selection pressure, the waning immunity against SARS-CoV-2 over the first year after infection possibly favors evolution of more infectious immune escape variants, amplifying the risk of reinfection. Additionally, the correlates of immune protection, the novel SARS-CoV-2 variants of concern (VOC), the durability of the adaptive and mucosal immunity remain major challenges for the development of therapeutic and prophylactic interventions. Interestingly, a recent body of evidence indicated that the gastrointestinal (GI) tract is another important target organ for SARS-CoV-2 besides the respiratory system, potentially increasing the likelihood of reinfection by impacting the microbiome and the immune response via the gut-lung axis. In this review, we summarized the latest development in SARS-CoV-2 reinfection, and explored the untapped potential of trained immunity. We also highlighted the immune memory kinetics of the humoral and cell-mediated immune response, genetic drift of the emerging viral variants, and discussed the current challenges in vaccine development. Understanding the dynamics and the quality of immune response by unlocking the power of the innate, humoral and cell-mediated immunity during SARS-CoV-2 reinfection would open newer avenues for drug discovery and vaccine designs.
Collapse
Affiliation(s)
- Neema Negi
- Department of Chemical Sciences, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick,Limerick, Ireland
| | - Shesh Prakash Maurya
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - Ravinder Singh
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - Bimal Kumar Das
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
171
|
Konka A, Lejawa M, Gaździcka J, Bochenek A, Fronczek M, Strzelczyk JK. RT-PCR Detection of SARS-CoV-2 among Individuals from the Upper Silesian Region-Analysis of 108,516 Tests. Diagnostics (Basel) 2021; 12:diagnostics12010007. [PMID: 35054172 PMCID: PMC8774892 DOI: 10.3390/diagnostics12010007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/06/2023] Open
Abstract
Background: The COVID-19 pandemic triggered by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has left a huge mark on everyday lives, introducing restrictions and plunging the global economy. This study aimed to analyze the available epidemiological data from the register of one of the largest laboratories testing for SARS-CoV-2 in the Silesian voivodship of Poland. Methods: This analysis is based upon the epidemiological records collected between 30 March 2020, and 30 April 2021, by the Silesian Park of Medical Technology Kardio-Med Silesia (Zabrze, Poland). In addition, we performed SARS-CoV-2 variant detection in samples from patients reinfected with SARS-CoV-2. Results: Our results confirm that SARS-CoV-2 infections are more common in urban areas. Laboratory-confirmed COVID-19 cases represent 13.21% of all RT-PCR test results during the 13 months of our laboratory diagnostics for SARS-CoV-2 infections. Detection of SARS-CoV-2 variants in samples of potentially reinfected patients showed discrepancies in the results. Conclusions: Due to the higher risk of SARS-CoV-2 infection among the Upper Silesian population, the region is at greater risk of deteriorating economic situation and healthcare as compared to other areas of Poland. RT-PCR methods are inexpensive and suitable for large-scale screening, but they can be untrustworthy so detection of SARS-CoV-2 variants in samples should be confirmed by sequencing.
Collapse
Affiliation(s)
- Adam Konka
- Silesian Park of Medical Technology Kardio-Med Silesia, M. Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland; (M.L.); (A.B.); (M.F.)
- Correspondence: ; Tel.: +48-32-705-03-05
| | - Mateusz Lejawa
- Silesian Park of Medical Technology Kardio-Med Silesia, M. Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland; (M.L.); (A.B.); (M.F.)
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Jordana 38 Str., 41-808 Zabrze, Poland
| | - Jadwiga Gaździcka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Str., 41-808 Zabrze, Poland; (J.G.); (J.K.S.)
| | - Aneta Bochenek
- Silesian Park of Medical Technology Kardio-Med Silesia, M. Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland; (M.L.); (A.B.); (M.F.)
| | - Martyna Fronczek
- Silesian Park of Medical Technology Kardio-Med Silesia, M. Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland; (M.L.); (A.B.); (M.F.)
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Jordana 38 Str., 41-808 Zabrze, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Str., 41-808 Zabrze, Poland; (J.G.); (J.K.S.)
| |
Collapse
|
172
|
Omaduvie U, Bachuwa G, Ayo-Yusuf O, Vardavas C, Agaku I. State specific estimates of vaccine hesitancy among US adults. POPULATION MEDICINE 2021. [DOI: 10.18332/popmed/144224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
173
|
COVID-19 vaccination strategies depend on the underlying network of social interactions. Sci Rep 2021; 11:24051. [PMID: 34912001 PMCID: PMC8674282 DOI: 10.1038/s41598-021-03167-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Since the onset of the coronavirus disease 2019 (COVID-19) pandemic, different mitigation and management strategies limiting economic and social activities have been implemented across many countries. Despite these strategies, the virus continues to spread and mutate. As a result, vaccinations are now administered to suppress the pandemic. Current COVID-19 epidemic models need to be expanded to account for the change in behaviour of new strains, such as an increased virulence and higher transmission rate. Furthermore, models need to account for an increasingly vaccinated population. We present a network model of COVID-19 transmission accounting for different immunity and vaccination scenarios. We conduct a parameter sensitivity analysis and find the average immunity length after an infection to be one of the most critical parameters that define the spread of the disease. Furthermore, we simulate different vaccination strategies and show that vaccinating highly connected individuals first is the quickest strategy for controlling the disease.
Collapse
|
174
|
Zinn AA, Izadjoo M, Kim H, Brody RL, Roth RR, Vega A, Nguyen KK, Ngo NT, Zinn HT, Antonopoulos N, Stoltenberg RM. Rapidly Self-Sterilizing PPE Capable of Destroying 100% of Microbes in 30-60 Seconds. Front Cell Infect Microbiol 2021; 11:752899. [PMID: 34976853 PMCID: PMC8715083 DOI: 10.3389/fcimb.2021.752899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
The continued proliferation of superbugs in hospitals and the coronavirus disease 2019 (COVID-19) has created an acute worldwide demand for sustained broadband pathogen suppression in households, hospitals, and public spaces. In response, we have created a highly active, self-sterilizing copper configuration capable of inactivating a wide range of bacteria and viruses in 30-60 seconds. The highly active material destroys pathogens faster than any conventional copper configuration and acts as quickly as alcohol wipes and hand sanitizers. Unlike the latter, our copper material does not release volatile compounds or leave harmful chemical residues and maintains its antimicrobial efficacy over sustained use; it is shelf stable for years. We have performed rigorous testing in accordance with guidelines from U.S. regulatory agencies and believe that the material could offer broad spectrum, non-selective defense against most microbes via integration into masks, protective equipment, and various forms of surface coatings.
Collapse
Affiliation(s)
| | - Mina Izadjoo
- Kuprion, Inc., San Jose, CA, United States
- Integrated Pharma Services, Frederick, MD, United States
| | - Hosan Kim
- Integrated Pharma Services, Frederick, MD, United States
| | | | | | | | | | - Nhi T. Ngo
- Kuprion, Inc., San Jose, CA, United States
| | | | | | | |
Collapse
|
175
|
Najjar H, Al-Jighefee HT, Qush A, Ahmed MN, Awwad S, Kamareddine L. COVID-19 Vaccination: The Mainspring of Challenges and the Seed of Remonstrance. Vaccines (Basel) 2021; 9:1474. [PMID: 34960220 PMCID: PMC8707780 DOI: 10.3390/vaccines9121474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
As of March 2020, the time when the coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became a pandemic, our existence has been threatened and the lives of millions have been claimed. With this ongoing global issue, vaccines are considered of paramount importance in curtailing the outbreak and probably a prime gamble to bring us back to 'ordinary life'. To date, more than 200 vaccine candidates have been produced, many of which were approved by the Food and Drug Administration (FDA) for emergency use, with the research and discovery phase of their production process passed over. Capering such a chief practice in COVID-19 vaccine development, and manufacturing vaccines at an unprecedented speed brought many challenges into play and raised COVID-19 vaccine remonstrance. In this review, we highlight relevant challenges to global COVID-19 vaccine development, dissemination, and deployment, particularly at the level of large-scale production and distribution. We also delineate public perception on COVID-19 vaccination and outline the main facets affecting people's willingness to get vaccinated.
Collapse
Affiliation(s)
- Hoda Najjar
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Hadeel T. Al-Jighefee
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Abeer Qush
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Muna Nizar Ahmed
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Sara Awwad
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Layla Kamareddine
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
176
|
Hossain ME, Rahman MM, Alam MS, Sarmin M, Karim Y, Hasan M, Hoque AF, Hasan MM, Rahman MZ, Chisti MJ, Rahman M. A Case Report: Genetically Distinct Severe Acute Respiratory Syndrome Coronavirus-2 Variant Causing Reinfection. Front Microbiol 2021; 12:792514. [PMID: 34956160 PMCID: PMC8696118 DOI: 10.3389/fmicb.2021.792514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/11/2021] [Indexed: 11/29/2022] Open
Abstract
Background: The emergence of novel variants has been a great deal of international concern since the recently published data suggest that previous infections with SARS-CoV-2 may not protect an individual from new variants. We report a patient had two distinct episodes of COVID-19 with different variants of SARS-CoV-2. Methods: The nasopharyngeal samples collected from the two episodes were subjected to whole-genome sequencing and comparative genome analysis. Results: The first infection presented with mild symptoms, while the second infection presented with severe outcomes which occurred 74 days after the patient recovered from the first episode. He had elevated C-reactive protein, ferritin, and bilateral consolidation as a sign of acute infection. Genome analysis revealed that the strains from the first and second episodes belonged to two distinct Nexstrain clades 20B and 20I and Pangolin lineages B.1.1.25 and B.1.1.7, respectively. A total of 36 mutations were observed in the episode-2 strain when compared with the reference strain Wuhan-Hu-1. Among them, eight mutations were identified in the receptor-binding domain (RBD). Conclusion: Our findings concern whether the immunity acquired by natural infection or mass vaccination could confer adequate protection against the constantly evolving SARS-CoV-2. Therefore, continuous monitoring of genetic variations of SARS-CoV-2 strains is crucial for interventions such as vaccine and drug designs, treatment using monoclonal antibodies, and patient management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Mustafizur Rahman
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
177
|
Abrokwa SK, Müller SA, Méndez-Brito A, Hanefeld J, El Bcheraoui C. Recurrent SARS-CoV-2 infections and their potential risk to public health - a systematic review. PLoS One 2021; 16:e0261221. [PMID: 34882750 PMCID: PMC8659325 DOI: 10.1371/journal.pone.0261221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/27/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To inform quarantine and contact-tracing policies concerning re-positive cases-cases testing positive among those recovered. MATERIALS AND METHODS We systematically reviewed and appraised relevant literature from PubMed and Embase for the extent of re-positive cases and their epidemiological characteristics. RESULTS In 90 case reports/series, a total of 276 re-positive cases were found. Among confirmed reinfections, 50% occurred within 90 days from recovery. Four reports related onward transmission. In thirty-five observational studies, rate of re-positives ranged from zero to 50% with no onward transmissions reported. In eight reviews, pooled recurrence rate ranged from 12% to 17.7%. Probability of re-positive increased with several factors. CONCLUSION Recurrence of a positive SARS-CoV-2 test is commonly reported within the first weeks following recovery from a first infection.
Collapse
Affiliation(s)
- Seth Kofi Abrokwa
- Evidence- based Public Health, Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Sophie Alice Müller
- Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Alba Méndez-Brito
- Evidence- based Public Health, Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Johanna Hanefeld
- Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Charbel El Bcheraoui
- Evidence- based Public Health, Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
178
|
Waterlow NR, van Leeuwen E, Davies NG, Flasche S, Eggo RM. How immunity from and interaction with seasonal coronaviruses can shape SARS-CoV-2 epidemiology. Proc Natl Acad Sci U S A 2021; 118:e2108395118. [PMID: 34873059 PMCID: PMC8670441 DOI: 10.1073/pnas.2108395118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2021] [Indexed: 12/11/2022] Open
Abstract
We hypothesized that cross-protection from seasonal epidemics of human coronaviruses (HCoVs) could have affected severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission, including generating reduced susceptibility in children. To determine what the prepandemic distribution of immunity to HCoVs was, we fitted a mathematical model to 6 y of seasonal coronavirus surveillance data from England and Wales. We estimated a duration of immunity to seasonal HCoVs of 7.8 y (95% CI 6.3 to 8.1) and show that, while cross-protection between HCoV and SARS-CoV-2 may contribute to the age distribution, it is insufficient to explain the age pattern of SARS-CoV-2 infections in the first wave of the pandemic in England and Wales. Projections from our model illustrate how different strengths of cross-protection between circulating coronaviruses could determine the frequency and magnitude of SARS-CoV-2 epidemics over the coming decade, as well as the potential impact of cross-protection on future seasonal coronavirus transmission.
Collapse
Affiliation(s)
- Naomi R Waterlow
- Centre for Mathematical Modeling of Infectious Disease, London School of Hygiene and Tropical Medicine, London WC14 7HT, United Kingdom;
| | - Edwin van Leeuwen
- Centre for Mathematical Modeling of Infectious Disease, London School of Hygiene and Tropical Medicine, London WC14 7HT, United Kingdom
- Statistics, Modelling and Economics Department, UK Health Security Agency, London NW9 5EQ, United Kingdom
| | - Nicholas G Davies
- Centre for Mathematical Modeling of Infectious Disease, London School of Hygiene and Tropical Medicine, London WC14 7HT, United Kingdom
| | - Stefan Flasche
- Centre for Mathematical Modeling of Infectious Disease, London School of Hygiene and Tropical Medicine, London WC14 7HT, United Kingdom
| | - Rosalind M Eggo
- Centre for Mathematical Modeling of Infectious Disease, London School of Hygiene and Tropical Medicine, London WC14 7HT, United Kingdom
| |
Collapse
|
179
|
Kamble P, Daulatabad V, John N, John J. Synopsis of symptoms of COVID-19 during second wave of the pandemic in India. Horm Mol Biol Clin Investig 2021; 43:97-104. [PMID: 34881534 DOI: 10.1515/hmbci-2021-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022]
Abstract
COVID-19 was caused by the original coronavirus, severe acute respiratory syndrome associated coronavirus-2 (SARS CoV2), which originated in Wuhan, China. COVID-19 had a large breakout of cases in early 2020, resulting in an epidemic that turned into a pandemic. This quickly enveloped the global healthcare system. The principal testing method for COVID-19 detection, according to the WHO, is reverse transcription polymerase chain reaction (RT-PCR). Isolation of patients, quarantine, masking, social distancing, sanitizer use, and complete lockdown were all vital health-care procedures for everyone. With the 'new normal' and vaccination programmes, the number of cases and recovered patients began to rise months later. The easing of restrictions during the plateau phase resulted in a rebound of instances, which hit the people with more ferocity and vengeance towards the start of April 2021. Coronaviruses have evolved to cause respiratory, enteric, hepatic, and neurologic diseases, resulting in a wide range of diseases and symptoms such as fever, cough, myalgia or fatigue, shortness of breath, muscle ache, headache, sore throat, rhinorrhea, hemoptysis, chest pain, nausea, vomiting, diarrhoea, anosmia, and ageusia. Coronavirus infections can be mild, moderate, or severe in intensity. COVID-19 pulmonary dysfunction includes lung edoema, ground-glass opacities, surfactant depletion, and alveolar collapse. Patients who presented with gastrointestinal (GI) symptoms such as anorexia, nausea, vomiting, or diarrhoea had a higher risk of negative outcomes. COVID-19's influence on cognitive function is one of COVID-19's long-term effects. More clinical situations need to be reviewed by healthcare professionals so that an appropriate management protocol may be developed to reduce morbidity and death in future coming third/fourth wave cases.
Collapse
Affiliation(s)
- Prafull Kamble
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Telangana, India
| | - Vandana Daulatabad
- RVM Institute of Medical Sciences and Research Centre, Siddipet, Hyderabad, Telangana, India
| | - Nitin John
- Department of Physiology, All India Institute of Medical Sciences, Bibinagar, Telangana, India
| | - Jyoti John
- Department of Biochemistry, All India Institute of Medical Sciences, Nagpur, Maharashtra, India
| |
Collapse
|
180
|
Kemp F, Proverbio D, Aalto A, Mombaerts L, Fouquier d'Hérouël A, Husch A, Ley C, Gonçalves J, Skupin A, Magni S. Modelling COVID-19 dynamics and potential for herd immunity by vaccination in Austria, Luxembourg and Sweden. J Theor Biol 2021; 530:110874. [PMID: 34425136 PMCID: PMC8378986 DOI: 10.1016/j.jtbi.2021.110874] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/28/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022]
Abstract
Against the COVID-19 pandemic, non-pharmaceutical interventions have been widely applied and vaccinations have taken off. The upcoming question is how the interplay between vaccinations and social measures will shape infections and hospitalizations. Hence, we extend the Susceptible-Exposed-Infectious-Removed (SEIR) model including these elements. We calibrate it to data of Luxembourg, Austria and Sweden until 15 December 2020. Sweden results having the highest fraction of undetected, Luxembourg of infected and all three being far from herd immunity in December. We quantify the level of social interaction, showing that a level around 1/3 of before the pandemic was still required in December to keep the effective reproduction number Refft below 1, for all three countries. Aiming to vaccinate the whole population within 1 year at constant rate would require on average 1,700 fully vaccinated people/day in Luxembourg, 24,000 in Austria and 28,000 in Sweden, and could lead to herd immunity only by mid summer. Herd immunity might not be reached in 2021 if too slow vaccines rollout speeds are employed. The model thus estimates which vaccination rates are too low to allow reaching herd immunity in 2021, depending on social interactions. Vaccination will considerably, but not immediately, help to curb the infection; thus limiting social interactions remains crucial for the months to come.
Collapse
Affiliation(s)
- Françoise Kemp
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg.
| | - Daniele Proverbio
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg.
| | - Atte Aalto
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg.
| | - Laurent Mombaerts
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg.
| | - Aymeric Fouquier d'Hérouël
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg.
| | - Andreas Husch
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg.
| | - Christophe Ley
- University of Ghent, Department of Applied Mathematics, Computer Science and Statistics, Krijgslaan 281-S9, 9000 Ghent, Belgium.
| | - Jorge Gonçalves
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg; University of Cambridge, Department of Plant Sciences, Downing St, Cambridge CB2 3EA, United Kingdom.
| | - Alexander Skupin
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg; University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, United States.
| | - Stefano Magni
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, 6 Av. du Swing, 4367 Belvaux, Luxembourg.
| |
Collapse
|
181
|
Anderson RM, Vegvari C, Hollingsworth TD, Pi L, Maddren R, Ng CW, Baggaley RF. The SARS-CoV-2 pandemic: remaining uncertainties in our understanding of the epidemiology and transmission dynamics of the virus, and challenges to be overcome. Interface Focus 2021; 11:20210008. [PMID: 34956588 PMCID: PMC8504893 DOI: 10.1098/rsfs.2021.0008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
Great progress has been made over the past 18 months in scientific understanding of the biology, epidemiology and pathogenesis of SARS-CoV-2. Extraordinary advances have been made in vaccine development and the execution of clinical trials of possible therapies. However, uncertainties remain, and this review assesses these in the context of virus transmission, epidemiology, control by social distancing measures and mass vaccination and the effect on all of these on emerging variants. We briefly review the current state of the global pandemic, focussing on what is, and what is not, well understood about the parameters that control viral transmission and make up the constituent parts of the basic reproductive number R 0. Major areas of uncertainty include factors predisposing to asymptomatic infection, the population fraction that is asymptomatic, the infectiousness of asymptomatic compared to symptomatic individuals, the contribution of viral transmission of such individuals and what variables influence this. The duration of immunity post infection and post vaccination is also currently unknown, as is the phenotypic consequences of continual viral evolution and the emergence of many viral variants not just in one location, but globally, given the high connectivity between populations in the modern world. The pattern of spread of new variants is also examined. We review what can be learnt from contact tracing, household studies and whole-genome sequencing, regarding where people acquire infection, and how households are seeded with infection since they constitute a major location for viral transmission. We conclude by discussing the challenges to attaining herd immunity, given the uncertainty in the duration of vaccine-mediated immunity, the threat of continued evolution of the virus as demonstrated by the emergence and rapid spread of the Delta variant, and the logistics of vaccine manufacturing and delivery to achieve universal coverage worldwide. Significantly more support from higher income countries (HIC) is required in low- and middle-income countries over the coming year to ensure the creation of community-wide protection by mass vaccination is a global target, not one just for HIC. Unvaccinated populations create opportunities for viral evolution since the net rate of evolution is directly proportional to the number of cases occurring per unit of time. The unit for assessing success in achieving herd immunity is not any individual country, but the world.
Collapse
Affiliation(s)
- Roy M. Anderson
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Carolin Vegvari
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - T. Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Joint Universities Pandemic and Epidemiological Research (JUNIPER) consortium, University of Leicester, Leicester, UK
| | - Li Pi
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Joint Universities Pandemic and Epidemiological Research (JUNIPER) consortium, University of Leicester, Leicester, UK
| | - Rosie Maddren
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Chi Wai Ng
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | | |
Collapse
|
182
|
Dun Y, Liu C, Ripley-Gonzalez JW, Liu P, Zhou N, Gong X, You B, Du Y, Liu J, Li B, Liu S. Six-month outcomes and effect of pulmonary rehabilitation among patients hospitalized with COVID-19: a retrospective cohort study. Ann Med 2021; 53:2099-2109. [PMID: 34766857 PMCID: PMC8592619 DOI: 10.1080/07853890.2021.2001043] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 10/27/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Patients appear to maintain sequelae post-coronavirus disease 2019 (COVID-19) affecting daily life and physical health. We investigated the changes in and the effects of pulmonary rehabilitation (PR) on exercise capacity and immunology six months after COVID-19 hospitalization. METHODS This retrospective cohort reviewed 233 COVID-19 patients admitted from 17 January 2020 to 29 February 2020. Ninety-eight patients who completed 2-week and 6-month follow-ups and tests were included. Among 98 patients, 27 completed at least five sessions of PR at the First Hospital of Changsha, China, during the 6-month convalescence were allocated to the PR group; the reminder who had not performed any PR were assigned to the control group. The primary outcome was the change in six-minute walk distance (6-MWD) between the 2-week and 6-month follow-ups, which was assessed via analysis of covariance with a covariate of propensity score that adjusted for the potential confounders. Secondary outcomes were the changes in 6-MWD, SARS-CoV-2 immunoglobulins, T-lymphocytes and blood chemistry, which were evaluated via paired tests. RESULTS Participants' ages ranged from 19 to 84 years (M = 47, standard deviation (SD)=15) 45.9% identified as male. During the 6-month convalescence, 6-MWD increased 27.0%, with a mean [95% CI] of 113 [92-134] m (p < .001). SARS-CoV-2 IgG and IgM decreased 33.3% (p = .002) and 43.8% (p = .009), CD4+ T cells increased 7.9% (p = .04), and the majority of blood chemistry significantly changed. The patients in the PR group acquired a greater increase in 6-MWD than those in control (unadjusted, 194 [167-221] m, p < .001; adjusted, 123 [68-181] m, p < .001), dose-responsiveness of PR on 6-MWD was observed (p < .001). No differences in immunity variables and blood chemistry were observed between groups. CONCLUSIONS These findings suggest PR may be a strategy to promote the improvement of exercise capacity after COVID-19.
Collapse
Affiliation(s)
- Yaoshan Dun
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
- Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Chao Liu
- The First Hospital of Changsha, Changsha, China
| | - Jeffrey W. Ripley-Gonzalez
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Ping Liu
- The First Hospital of Changsha, Changsha, China
| | - Nanjiang Zhou
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Xun Gong
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Baiyang You
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
| | - Yang Du
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, China
| | - Jiyang Liu
- The First Hospital of Changsha, Changsha, China
| | - Bo Li
- The First Hospital of Changsha, Changsha, China
| | - Suixin Liu
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
183
|
Lamptey E. Should Breakthrough SARS-CoV-2 Infection Affect Our Confidence in the COVID-19 Vaccines? Infect Chemother 2021; 53:676-685. [PMID: 34979603 PMCID: PMC8731259 DOI: 10.3947/ic.2021.0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/19/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines have shown excellent clinical efficacy and effectiveness in real-world data. Although they are regarded as a means of ending the global pandemic, some individuals become infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) after immunization. This growing evidence or lack of effectiveness has led to vaccine refusal. This paper sought to help the public gain a deeper understanding of breakthrough events creating the positive perception that vaccines remain effective in ending the pandemic. Discussions vital for the general public's active participation in vaccination are expressed to create confidence or provide a potential solution to the challenge of vaccine hesitancy.
Collapse
Affiliation(s)
- Emmanuel Lamptey
- Institute of Life and Earth Sciences (Including Health and Agriculture), Pan African University, University of Ibadan, Nigeria. ,
| |
Collapse
|
184
|
Jeffery-Smith A, Rowland TAJ, Patel M, Whitaker H, Iyanger N, Williams SV, Giddings R, Thompson L, Zavala M, Aiano F, Ellis J, Lackenby A, Höschler K, Brown K, Ramsay ME, Gopal R, Chow JY, Ladhani SN, Zambon M. Reinfection with new variants of SARS-CoV-2 after natural infection: a prospective observational cohort in 13 care homes in England. THE LANCET HEALTHY LONGEVITY 2021; 2:e811-e819. [PMID: 34873592 PMCID: PMC8635459 DOI: 10.1016/s2666-7568(21)00253-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Understanding the duration of protection and risk of reinfection after natural infection is crucial to planning COVID-19 vaccination for at-risk groups, including care home residents, particularly with the emergence of more transmissible variants. We report on the duration, neutralising activity, and protection against the alpha variant of previous SARS-CoV-2 infection in care home residents and staff infected more than 6 months previously. Methods We did this prospective observational cohort surveillance in 13 care homes in Greater London, England. All staff and residents were included. Staff and residents had regular nose and throat screening for SARS-CoV-2 by RT-PCR according to national guidelines, with ad hoc testing of symptomatic individuals. From January, 2021, antigen lateral flow devices were also used, but positive tests still required RT-PCR confirmation. Staff members took the swab samples for themselves and the residents. The primary outcome was SARS-CoV-2 RT-PCR positive primary infection or reinfection in previously infected individuals, as determined by previous serological testing and screening or diagnostic RT-PCR results. Poisson regression and Cox proportional hazards models were used to estimate protective effectiveness of previous exposure. SARS-CoV-2 spike, nucleoprotein, and neutralising antibodies were assessed at multiple timepoints as part of the longitudinal follow-up. Findings Between April 10 and Aug 3, 2020, we recruited and tested 1625 individuals (933 staff and 692 residents). 248 participants were lost to follow-up (123 staff and 125 residents) and 1377 participants were included in the follow-up period to Jan 31, 2021 (810 staff and 567 residents). There were 23 reinfections (ten confirmed, eight probable, five possible) in 656 previously infected individuals (366 staff and 290 residents), compared with 165 primary infections in 721 susceptible individuals (444 staff and 277 residents). Those with confirmed reinfections had no or low neutralising antibody concentration before reinfection, with boosting of titres after reinfection. Kinetics of binding and neutralising antibodies were similar in older residents and younger staff. Interpretation SARS-CoV-2 reinfections were rare in older residents and younger staff. Protection from SARS-CoV-2 was sustained for longer than 9 months, including against the alpha variant. Reinfection was associated with no or low neutralising antibody before reinfection, but significant boosting occurred on reinfection. Funding Public Health England.
Collapse
Affiliation(s)
- Anna Jeffery-Smith
- Virus Reference Department, Public Health England, London, UK
- Blizard Institute, Queen Mary University of London, London, UK
- Correspondence to: Dr Anna Jeffery-Smith, Virus Reference Department, Public Health England, London NW9 5EQ, UK
| | | | - Monika Patel
- Virus Reference Department, Public Health England, London, UK
| | | | - Nalini Iyanger
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Sarah V Williams
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Rebecca Giddings
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Leah Thompson
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Maria Zavala
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Felicity Aiano
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Joanna Ellis
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Angie Lackenby
- Virus Reference Department, Public Health England, London, UK
| | - Katja Höschler
- Virus Reference Department, Public Health England, London, UK
| | - Kevin Brown
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Mary E Ramsay
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Robin Gopal
- Virus Reference Department, Public Health England, London, UK
| | - J Yimmy Chow
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Shamez N Ladhani
- Immunisation and Countermeasures Division, Public Health England, London, UK
- Paediatric Infectious Diseases Research Group, St. George's University of London, London, UK
| | - Maria Zambon
- Virus Reference Department, Public Health England, London, UK
| |
Collapse
|
185
|
Yinda CK, Port JR, Bushmaker T, Fischer RJ, Schulz JE, Holbrook MG, Shaia C, de Wit E, van Doremalen N, Munster VJ. Prior aerosol infection with lineage A SARS-CoV-2 variant protects hamsters from disease, but not reinfection with B.1.351 SARS-CoV-2 variant. Emerg Microbes Infect 2021; 10:1284-1292. [PMID: 34120579 PMCID: PMC8238069 DOI: 10.1080/22221751.2021.1943539] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
The circulation of SARS-CoV-2 has resulted in the emergence of variants of concern (VOCs). It is currently unclear whether the previous infection with SARS-CoV-2 provides protection against reinfection with VOCs. Here, we show that low dose aerosol exposure to hCoV-19/human/USA/WA-CDC-WA1/2020 (WA1, lineage A), resulted in a productive mild infection. In contrast, a low dose of SARS-CoV-2 via fomites did not result in productive infection in the majority of exposed hamsters and these animals remained non-seroconverted. After recovery, hamsters were re-exposed to hCoV-19/South African/KRISP-K005325/2020 (VOC B.1.351) via an intranasal challenge. Seroconverted rechallenged animals did not lose weight and shed virus for three days. They had a little infectious virus and no pathology in the lungs. In contrast, shedding, weight loss and extensive pulmonary pathology caused by B.1.351 replication were observed in the non-seroconverted animals. The rechallenged seroconverted animals did not transmit the virus to naïve sentinels via direct contact transmission, in contrast to the non-seroconverted animals. Reinfection with B.1.351 triggered an anamnestic response that boosted not only neutralizing titres against lineage A, but also titres against B.1.351. Our results confirm that aerosol exposure is a more efficient infection route than fomite exposure. Furthermore, initial infection with SARS-CoV-2 lineage A does not prevent heterologous reinfection with B.1.351 but prevents disease and onward transmission. These data suggest that previous SARS-CoV-2 exposure induces partial protective immunity. The reinfection generated a broadly neutralizing humoral response capable of effectively neutralizing B.1.351 while maintaining its ability to neutralize the virus to which the initial response was directed against.
Collapse
Affiliation(s)
- Claude Kwe Yinda
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Julia R. Port
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Trenton Bushmaker
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Robert J. Fischer
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jonathan E. Schulz
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Myndi G. Holbrook
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Vincent J. Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
186
|
Dhillon RA, Qamar MA, Gilani JA, Irfan O, Waqar U, Sajid MI, Mahmood SF. The mystery of COVID-19 reinfections: A global systematic review and meta-analysis. Ann Med Surg (Lond) 2021; 72:103130. [PMID: 34900250 PMCID: PMC8642249 DOI: 10.1016/j.amsu.2021.103130] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND As the COVID-19 pandemic rages on, reports on disparities in vaccine roll out alongside COVID-19 reinfection have been emerging. We conducted a systematic review to assess the determinants and disease spectrum of COVID-19 reinfection. MATERIALS AND METHODS A comprehensive search covering relevant databases was conducted for observational studies reporting Polymerase Chain Reaction (PCR) confirmed infection and reinfection cases. A quality assessment tool developed by the National Institute of Health (NIH) for the assessment of case series was utilized. Meta-analyses were performed using RevMan 5.3 for pooled proportions of findings in first infection and reinfection with a 95% confidence interval (CI). RESULTS Eighty-one studies reporting 577 cases were included from 22 countries. The mean age of patients was 46.2 ± 18.9 years and 179 (31.0%) cases of comorbidities were reported. The average time duration between first infection and reinfection was 63.6 ± 48.9 days. During first infection and reinfection, fever was the most common symptom (41.4% and 36.4%, respectively) whilst anti-viral therapy was the most common treatment regimen administered (44.5% and 43.0%, respectively). Comparable odds of symptomatic presentation and management were reported for the two infections. However, a higher Intensive Care Unit (ICU) admission rate was observed in reinfection compared to first infection (10 vs 3). Ten deaths were reported with respiratory failure being the most common cause of death (7/10 deaths). CONCLUSION Our findings support immunization practices given increased ICU admissions and mortality in reinfections. Our cohort serves as a guide for clinicians and authorities in devising an optimal strategy for controlling the pandemic. (249 words).
Collapse
Affiliation(s)
| | | | | | - Omar Irfan
- Amaris Consulting, Toronto, Ontario, Canada
| | - Usama Waqar
- Medical College, Aga Khan University, Karachi, Sindh, Pakistan
| | | | - Syed Faisal Mahmood
- Section of Infectious Diseases, Aga Khan University, Karachi, Sindh, Pakistan
| |
Collapse
|
187
|
Alleman TW, Vergeynst J, De Visscher L, Rollier M, Torfs E, Nopens I, Baetens JM. Assessing the effects of non-pharmaceutical interventions on SARS-CoV-2 transmission in Belgium by means of an extended SEIQRD model and public mobility data. Epidemics 2021; 37:100505. [PMID: 34649183 PMCID: PMC8487325 DOI: 10.1016/j.epidem.2021.100505] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 07/16/2021] [Accepted: 09/28/2021] [Indexed: 01/10/2023] Open
Abstract
We present a compartmental extended SEIQRD metapopulation model for SARS-CoV-2 spread in Belgium. We demonstrate the robustness of the calibration procedure by calibrating the model using incrementally larger datasets and dissect the model results by computing the effective reproduction number at home, in workplaces, in schools, and during leisure activities. We find that schools and home contacts are important transmission pathways for SARS-CoV-2 under lockdown measures. School reopening has the potential to increase the effective reproduction number from Re=0.66±0.04 (95 % CI) to Re=1.09±0.05 (95 % CI) under lockdown measures. The model accounts for the main characteristics of SARS-CoV-2 transmission and COVID-19 disease and features a detailed representation of hospitals with parameters derived from a dataset consisting of 22 136 hospitalized patients. Social contact during the pandemic is modeled by scaling pre-pandemic contact matrices with Google Community Mobility data and with effectivity-of-contact parameters inferred from hospitalization data. The calibrated social contact model with its publically available mobility data, although coarse-grained, is a cheap and readily available alternative to social-epidemiological contact studies under lockdown measures, which were not available at the start of the pandemic.
Collapse
Affiliation(s)
- Tijs W Alleman
- BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium.
| | - Jenna Vergeynst
- BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium; KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium
| | - Lander De Visscher
- KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium
| | - Michiel Rollier
- KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium
| | - Elena Torfs
- BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium
| | - Ingmar Nopens
- BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium
| | - Jan M Baetens
- KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, 9000 Gent, Belgium
| |
Collapse
|
188
|
Does infection with or vaccination against SARS-CoV-2 lead to lasting immunity? THE LANCET. RESPIRATORY MEDICINE 2021; 9:1450-1466. [PMID: 34688434 PMCID: PMC8530467 DOI: 10.1016/s2213-2600(21)00407-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/26/2021] [Accepted: 08/21/2021] [Indexed: 12/17/2022]
Abstract
Many nations are pursuing the rollout of SARS-CoV-2 vaccines as an exit strategy from unprecedented COVID-19-related restrictions. However, the success of this strategy relies critically on the duration of protective immunity resulting from both natural infection and vaccination. SARS-CoV-2 infection elicits an adaptive immune response against a large breadth of viral epitopes, although the duration of the response varies with age and disease severity. Current evidence from case studies and large observational studies suggests that, consistent with research on other common respiratory viruses, a protective immunological response lasts for approximately 5-12 months from primary infection, with reinfection being more likely given an insufficiently robust primary humoral response. Markers of humoral and cell-mediated immune memory can persist over many months, and might help to mitigate against severe disease upon reinfection. Emerging data, including evidence of breakthrough infections, suggest that vaccine effectiveness might be reduced significantly against emerging variants of concern, and hence secondary vaccines will need to be developed to maintain population-level protective immunity. Nonetheless, other interventions will also be required, with further outbreaks likely to occur due to antigenic drift, selective pressures for novel variants, and global population mobility.
Collapse
|
189
|
Gaudreault NN, Carossino M, Morozov I, Trujillo JD, Meekins DA, Madden DW, Cool K, Artiaga BL, McDowell C, Bold D, Balaraman V, Kwon T, Ma W, Henningson J, Wilson DW, Wilson WC, Balasuriya UBR, García-Sastre A, Richt JA. Experimental re-infected cats do not transmit SARS-CoV-2. Emerg Microbes Infect 2021; 10:638-650. [PMID: 33704016 PMCID: PMC8023599 DOI: 10.1080/22221751.2021.1902753] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
SARS-CoV-2 is the causative agent of COVID-19 and responsible for the current global pandemic. We and others have previously demonstrated that cats are susceptible to SARS-CoV-2 infection and can efficiently transmit the virus to naïve cats. Here, we address whether cats previously exposed to SARS-CoV-2 can be re-infected with SARS-CoV-2. In two independent studies, SARS-CoV-2-infected cats were re-challenged with SARS-CoV-2 at 21 days post primary challenge (DPC) and necropsies performed at 4, 7 and 14 days post-secondary challenge (DP2C). Sentinels were co-mingled with the re-challenged cats at 1 DP2C. Clinical signs were recorded, and nasal, oropharyngeal, and rectal swabs, blood, and serum were collected and tissues examined for histologic lesions. Viral RNA was transiently shed via the nasal, oropharyngeal and rectal cavities of the re-challenged cats. Viral RNA was detected in various tissues of re-challenged cats euthanized at 4 DP2C, mainly in the upper respiratory tract and lymphoid tissues, but less frequently and at lower levels in the lower respiratory tract when compared to primary SARS-CoV-2 challenged cats at 4 DPC. Viral RNA and antigen detected in the respiratory tract of the primary SARS-CoV-2 infected cats at early DPCs were absent in the re-challenged cats. Naïve sentinels co-housed with the re-challenged cats did not shed virus or seroconvert. Together, our results indicate that cats previously infected with SARS-CoV-2 can be experimentally re-infected with SARS-CoV-2; however, the levels of virus shed was insufficient for transmission to co-housed naïve sentinels. We conclude that SARS-CoV-2 infection in cats induces immune responses that provide partial, non-sterilizing immune protection against re-infection.
Collapse
Affiliation(s)
- Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Mariano Carossino
- Louisiana Animal Disease Diagnostic Laboratory and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Igor Morozov
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Jessie D. Trujillo
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - David A. Meekins
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Daniel W. Madden
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Konner Cool
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Bianca Libanori Artiaga
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Chester McDowell
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Dashzeveg Bold
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Velmurugan Balaraman
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Taeyong Kwon
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Wenjun Ma
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
- Department of Veterinary Pathobiology and Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Jamie Henningson
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Dennis W. Wilson
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - William C. Wilson
- Arthropod Borne Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS, USA
| | - Udeni B. R. Balasuriya
- Louisiana Animal Disease Diagnostic Laboratory and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
190
|
Campos-Ferreira D, Visani V, Córdula C, Nascimento G, Montenegro L, Schindler H, Cavalcanti I. COVID-19 challenges: From SARS-CoV-2 infection to effective point-of-care diagnosis by electrochemical biosensing platforms. Biochem Eng J 2021; 176:108200. [PMID: 34522158 PMCID: PMC8428033 DOI: 10.1016/j.bej.2021.108200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022]
Abstract
In January 2020, the World Health Organization (WHO) identified a new zoonotic virus, SARS-CoV-2, responsible for causing the COVID-19 (coronavirus disease 2019). Since then, there has been a collaborative trend between the scientific community and industry. Multidisciplinary research networks try to understand the whole SARS-CoV-2 pathophysiology and its relationship with the different grades of severity presented by COVID-19. The scientific community has gathered all the data in the quickly developed vaccines that offer a protective effect for all variants of the virus and promote new diagnostic alternatives able to have a high standard of efficiency, added to shorter response analysis time and portability. The industry enters in the context of accelerating the path taken by science until obtaining the final product. In this review, we show the principal diagnostic methods developed during the COVID-19 pandemic. However, when we observe the diagnostic tools section of an efficient infection outbreak containment report and the features required for such tools, we could observe a highlight of electrochemical biosensing platforms. Such devices present a high standard of analytical performance, are low-cost tools, easy to handle and interpret, and can be used in the most remote and low-resource regions. Therefore, probably, they are the ideal point-of-care diagnostic tools for pandemic scenarios.
Collapse
Affiliation(s)
- D. Campos-Ferreira
- Laboratório de Imunopatologia Keizo Asami – LIKA/ UFPE, Av. Prof. Moraes Rego, s/n, CEP: 506070-901 Recife, PE, Brazil,Corresponding author
| | - V. Visani
- Laboratório de Imunopatologia Keizo Asami – LIKA/ UFPE, Av. Prof. Moraes Rego, s/n, CEP: 506070-901 Recife, PE, Brazil
| | - C. Córdula
- Laboratório de Imunopatologia Keizo Asami – LIKA/ UFPE, Av. Prof. Moraes Rego, s/n, CEP: 506070-901 Recife, PE, Brazil
| | - G.A. Nascimento
- Laboratório de Imunopatologia Keizo Asami – LIKA/ UFPE, Av. Prof. Moraes Rego, s/n, CEP: 506070-901 Recife, PE, Brazil,Centro Acadêmico do Agreste - CAA/UFPE, Av. Marielle Franco, s/n - Km 59 - Bairro Nova Caruaru, CEP: 55.014-900 Caruaru, PE, Brazil
| | - L.M.L. Montenegro
- Fundação Oswaldo Cruz (Fiocruz), Centro de Pesquisas Instituto Aggeu Magalhães (IAM), Av. Professor Moraes Rego s/n, CEP: 50670-901 Recife, PE, Brazil
| | - H.C. Schindler
- Fundação Oswaldo Cruz (Fiocruz), Centro de Pesquisas Instituto Aggeu Magalhães (IAM), Av. Professor Moraes Rego s/n, CEP: 50670-901 Recife, PE, Brazil
| | - I.M.F. Cavalcanti
- Laboratório de Imunopatologia Keizo Asami – LIKA/ UFPE, Av. Prof. Moraes Rego, s/n, CEP: 506070-901 Recife, PE, Brazil,Centro Acadêmico de Vitória – CAV/UFPE, R. Alto do Reservatório, CEP: 55 612-440 Vitória de Santo Antão, PE, Brazil
| |
Collapse
|
191
|
Gularte JS, da Silva MS, Demoliner M, Hansen AW, Heldt FH, Silveira F, Filippi M, Pereira VMDAG, da Silva FP, Mallmann L, Fink P, Laux JL, Weber MN, de Almeida PR, Fleck JD, Spilki FR. Reinfection cases by closely related SARS-CoV-2 lineages in Southern Brazil. Braz J Microbiol 2021; 52:1881-1885. [PMID: 34562232 PMCID: PMC8475897 DOI: 10.1007/s42770-021-00621-0] [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: 07/23/2021] [Accepted: 09/19/2021] [Indexed: 11/25/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the pandemic that started in late 2019 and still affects people's lives all over the world. Lack of protective immunity after primary infection has been involved with reported reinfection cases by SARS-CoV-2. In this study, we described two cases of reinfection caused by non-VOC (Variants of Concern) strains in southern Brazil, being one patient a healthcare worker. The four samples previously positive for SARS-CoV-2 by real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) were sequenced by a high-performance platform and the genomic analysis confirmed that lineages responsible for infections were B.1.91 and B.1.1.33 (patient 1), and B.1.1.33 and B.1.1.28 (patient 2). The interval between the two positive RT-qPCR for patients 1 and 2 was 45 and 61 days, respectively. This data shows that patients may be reinfected even by very closely related SARS-CoV-2 lineages.
Collapse
Affiliation(s)
- Juliana Schons Gularte
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil.
| | - Mariana Soares da Silva
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Meriane Demoliner
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Alana Witt Hansen
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Fágner Henrique Heldt
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Flávio Silveira
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Micheli Filippi
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Vyctoria Malayhka de Abreu Góes Pereira
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Francini Pereira da Silva
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Larissa Mallmann
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Pietra Fink
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Jéssica Luísa Laux
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Matheus Nunes Weber
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Paula Rodrigues de Almeida
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Juliane Deise Fleck
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| | - Fernando Rosado Spilki
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, Nº 2755, Prédio Vermelho, Piso 1, sala 103, Vila Nova, Novo Hamburgo, Rio Grande do Sul, CEP 93525-075, Brazil
| |
Collapse
|
192
|
Zarkesh K, Entezar-Almahdi E, Ghasemiyeh P, Akbarian M, Bahmani M, Roudaki S, Fazlinejad R, Mohammadi-Samani S, Firouzabadi N, Hosseini M, Farjadian F. Drug-based therapeutic strategies for COVID-19-infected patients and their challenges. Future Microbiol 2021; 16:1415-1451. [PMID: 34812049 PMCID: PMC8610072 DOI: 10.2217/fmb-2021-0116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging epidemic-prone diseases have introduced numerous health and economic challenges in recent years. Given current knowledge of COVID-19, herd immunity through vaccines alone is unlikely. In addition, vaccination of the global population is an ongoing challenge. Besides, the questions regarding the prevalence and the timing of immunization are still under investigation. Therefore, medical treatment remains essential in the management of COVID-19. Herein, recent advances from beginning observations of COVID-19 outbreak to an understanding of the essential factors contributing to the spread and transmission of COVID-19 and its treatment are reviewed. Furthermore, an in-depth discussion on the epidemiological aspects, clinical symptoms and most efficient medical treatment strategies to mitigate the mortality and spread rates of COVID-19 is presented.
Collapse
Affiliation(s)
- Khatereh Zarkesh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elaheh Entezar-Almahdi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Ghasemiyeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Akbarian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Bahmani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrzad Roudaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rahil Fazlinejad
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Hosseini
- Department of Manufacturing & Industrial Engineering, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
193
|
Olanipekun T, Abe T, Effoe V, Westney G, Snyder R. Changes in COVID-19 vaccine acceptance rate among recovered critically Ill patients: A 12-month follow-up study. Vaccine 2021; 39:7074-7081. [PMID: 34756611 PMCID: PMC8542446 DOI: 10.1016/j.vaccine.2021.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 08/17/2021] [Accepted: 10/09/2021] [Indexed: 11/28/2022]
Abstract
Introduction We surveyed a cohort of patients who recovered from severe SARS-CoV-2 infection to determine the COVID-19 vaccination rate. We also compared the willingness to accept COVID-19 vaccine before and after its availability to assess changes in perception and attitude towards vaccination. Materials and Methods Recovered patients with severe hypoxemic respiratory failure from SARS-CoV-2 infection treated in the ICU at Grady Memorial Hospital, Atlanta, Georgia between April 1, 2020, and June 30, 2020 were followed up over a 1-year period to assess vaccine acceptability and acceptance rates, and changes in perception towards COVID-19 vaccination before and after vaccine availability. Results A total of 98 and 93 patients completed the initial and follow up surveys respectively. During the initial survey, 41% of the patients intended to receive vaccination, 46% responded they would not accept a vaccine against COVID-19 even if it were proven to be ‘safe and effective ‘and 13% undecided. During the follow up survey, 44% of the study cohort had received at least one dose of a COVID-19 vaccine. Major reasons provided by respondents for not accepting COVID-19 vaccine were lack of trust in the effectiveness of the vaccine, pharmaceutical companies, government, vaccine technology, fear of side effects and perceived immunity against COVID-19. Respondents were more likely to be vaccinated if recommended by their physicians (OR 6.4, 95% CI 2.8–8.3), employers (OR 2.5, 95% CI 1.9–5.8), and family and friends (OR 1.6, 95% CI 1.1–4.5). Conclusion We found a suboptimal COVID-19 vaccination rate in a cohort of patients who recovered from severe infection. COVID-19 vaccine information and recommendation by healthcare providers, employers, and family and friends may improve vaccination uptake.
Collapse
Affiliation(s)
- Titilope Olanipekun
- Department of Hospital Medicine, Covenant Health System, Knoxville, TN, USA; Department of Internal Medicine, Morehouse School of Medicine, Atlanta, GA, USA.
| | - Temidayo Abe
- Department of Internal Medicine, Morehouse School of Medicine, Atlanta, GA, USA
| | - Valery Effoe
- Department of Internal Medicine, Morehouse School of Medicine, Atlanta, GA, USA; Division of Cardiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Gloria Westney
- Department of Internal Medicine, Morehouse School of Medicine, Atlanta, GA, USA; Division of Pulmonary and Critical Care Medicine, Atlanta, GA, USA
| | - Richard Snyder
- Department of Internal Medicine, Morehouse School of Medicine, Atlanta, GA, USA; Division of Pulmonary and Critical Care Medicine, Atlanta, GA, USA
| |
Collapse
|
194
|
Kow CS, Javed A, Ramachandram D, Hasan SS. Clinical outcomes of sofosbuvir-based antivirals in patients with COVID-19: a systematic review and meta-analysis of randomized trials. Expert Rev Anti Infect Ther 2021; 20:567-575. [PMID: 34719324 DOI: 10.1080/14787210.2022.2000861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Several randomized trials have evaluated the effects of sofosbuvir-based direct-acting antivirals on the clinical outcomes in patients with COVID-19. METHODS A systematic literature search with no language restrictions was performed on electronic databases and preprint repositories to identify eligible randomized trials published up to 8 July 2021. A random-effects model was used to estimate the pooled odds ratio (OR) for outcomes of interest with the use of sofosbuvir combined with direct-acting antiviral agents relative to the nonuse of sofosbuvir-based direct-acting antiviral agents at 95% confidence intervals (CI). RESULTS The meta-analysis of 11 trials (n = 2,161) revealed statistically significant reduction in the odds of mortality (pooled odds ratio = 0.59; 95% confidence interval 0.36 to 0.99) but no statistically significant difference in the odds of development of composite endpoint of severe illness (pooled odds ratio = 0.79; 95% confidence interval 0.43 to 1.44) with the administration of sofosbuvir-based direct-acting antiviral agents among patients with COVID-19, relative to non-administration of sofosbuvir-based direct-acting antiviral agents.Subgroup analysis with seven trials involving sofosbuvir-daclatasvir revealed no significant mortality benefit (pooled odds ratio = 0.77; 95% confidence interval 0.48 to 1.22). CONCLUSION Sofosbuvir-based direct-acting antiviral agents have no protective effects against the development of severe illness in patients with COVID-19 with the current dosing regimen. Whether sofosbuvir-based direct-acting antiviral agents could offer mortality benefits would require further investigations.
Collapse
Affiliation(s)
- Chia Siang Kow
- School of Pharmacy, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Amaan Javed
- University College of Medical Sciences, University of Delhi, Delhi, India
| | | | - Syed Shahzad Hasan
- School of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| |
Collapse
|
195
|
Sciscent BY, Eisele CD, Ho L, King SD, Jain R, Golamari RR. COVID-19 reinfection: the role of natural immunity, vaccines, and variants. J Community Hosp Intern Med Perspect 2021; 11:733-739. [PMID: 34804382 PMCID: PMC8604456 DOI: 10.1080/20009666.2021.1974665] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The COVID-19 pandemic has altered innumerable lives. Although recent mass vaccinations offer a glimmer of hope, the rising death toll and new variants continue to dominate the current scenario. As we begin to understand more about SARS-CoV-2 infections, the territory of reinfections with COVID-19 remains unexplored. In this review, we will discuss several aspects of reinfection: (a) How is COVID-19 reinfection characterized? (b) Does prior literature differentiate between reinfection and reactivation? (c) What SARS-CoV-2 strains do the vaccines target and can they protect against new strains? Larger and longer timeline studies are needed to understand reinfection risks. With the ongoing distribution of the SARS-CoV-2 vaccines to provide protection, the understanding of the possibility for SARS-CoV-2 reinfection remains critical. Abbreviations CDC: Centers for Disease ControlSARS-CoV-2: Severe acute respiratory syndrome coronavirus 2COVID-19: Coronavirus disease 2019RT-PCR: Reverse Transcription Polymerase Chain ReactionPASC: Post-Acute Sequelae of SARS-CoV-2 infection
Collapse
Affiliation(s)
- Bao Y Sciscent
- The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| | - Caroline D Eisele
- The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| | - Lisa Ho
- The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| | - Steven D King
- The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| | - Rohit Jain
- Department of Hospital Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Reshma R Golamari
- Department of Hospital Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
196
|
Second SARS-CoV-2 infections twelve months after initial infections in Australia, confirmed by genomic analysis. Med J Aust 2021; 216:199-201. [PMID: 34808695 PMCID: PMC9011572 DOI: 10.5694/mja2.51352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/03/2022]
|
197
|
Kumar N, Oke J, Nahmias-Biran BH. Activity-based epidemic propagation and contact network scaling in auto-dependent metropolitan areas. Sci Rep 2021; 11:22665. [PMID: 34811414 PMCID: PMC8608855 DOI: 10.1038/s41598-021-01522-w] [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: 11/18/2020] [Accepted: 10/26/2021] [Indexed: 01/03/2023] Open
Abstract
We build on recent work to develop a fully mechanistic, activity-based and highly spatio-temporally resolved epidemiological model which leverages person-trajectories obtained from an activity-based model calibrated for two full-scale prototype cities, consisting of representative synthetic populations and mobility networks for two contrasting auto-dependent city typologies. We simulate the propagation of the COVID-19 epidemic in both cities to analyze spreading patterns in urban networks across various activity types. Investigating the impact of the transit network, we find that its removal dampens disease propagation significantly, suggesting that transit restriction is more critical for mitigating post-peak disease spreading in transit dense cities. In the latter stages of disease spread, we find that the greatest share of infections occur at work locations. A statistical analysis of the resulting activity-based contact networks indicates that transit contacts are scale-free, work contacts are Weibull distributed, and shopping or leisure contacts are exponentially distributed. We validate our simulation results against existing case and mortality data across multiple cities in their respective typologies. Our framework demonstrates the potential for tracking epidemic propagation in urban networks, analyzing socio-demographic impacts and assessing activity- and mobility-specific implications of both non-pharmaceutical and pharmaceutical intervention strategies.
Collapse
Affiliation(s)
- Nishant Kumar
- ETH Zurich, Future Resilient Systems, Singapore-ETH Centre, Singapore, 138602, Singapore
| | - Jimi Oke
- Department of Civil and Environmental Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Bat-Hen Nahmias-Biran
- Department of Civil Engineering, Ariel University, Ariel, 40700, Israel.
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
198
|
Borges MASB, Guilarde AO, Tomich LGMDM, Turchi MD. Clinical practice in COVID-19: The most frequently asked questions to infectious diseases specialists. Braz J Infect Dis 2021; 25:101648. [PMID: 34793712 PMCID: PMC8572702 DOI: 10.1016/j.bjid.2021.101648] [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: 06/30/2021] [Revised: 09/28/2021] [Accepted: 10/17/2021] [Indexed: 12/15/2022] Open
Abstract
Since the emergence of the disease caused by the severe respiratory syndrome coronavirus 2 (SARS-CoV-2) - COVID-19 - in late December 2019, a vast number of publications on the subject appeared in peer-reviewed journals and preprints. Despite the significant amount of available information, infectious disease physicians are requested to solve questions from colleagues, patients, and relatives on a daily basis. Here, we aim to describe the evidence supporting the answers for frequently asked questions, based on a literature review. We created a web-based questionnaire which was distributed to a group of 70 infectious disease specialists and medical residents, asking what questions and issues they most frequently faced. The 10 most frequent questions guided the topics for a narrative review. We provide evidence and consensus-based information on subjects such as infection and transmission, isolation, management of COVID-19 confirmed cases, reinfection, clinical-therapeutic management, vaccination, and antibodies post-infection/vaccination. Correctly clarifying doubts and providing clear information to physicians, patients, and family members helps to better manage COVID-19 in the community and the hospital settings.
Collapse
Affiliation(s)
- Moara Alves Santa Bárbara Borges
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil; Serviço de Infectologia, Hospital das Clínicas da Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Adriana Oliveira Guilarde
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil; Serviço de Infectologia, Hospital das Clínicas da Universidade Federal de Goiás, Goiânia, GO, Brazil; Centro de Reabilitação e Readaptação Dr. Henrique Santillo-CRER, Goiânia, GO, Brazil
| | - Lísia Gomes Martins de Moura Tomich
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil; Hospital Estadual de Doenças Tropicais, Goiânia, GO, Brazil; Universidade de Rio Verde - campus Aparecida de Goiânia, Aparecida de Goiânia, GO, Brazil
| | - Marília Dalva Turchi
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| |
Collapse
|
199
|
Klein RC, Fabres Klein MH, Barbosa LG, Gonzaga Knnup LV, Rodrigues Venâncio LP, Lima JB, Araújo-Santos T. Identifying Inconclusive Data in the SARS-CoV-2 Molecular Diagnostic Using Nucleocapsid Phosphoprotein Gene as Target. Arch Pathol Lab Med 2021; 146:272-277. [PMID: 34797908 DOI: 10.5858/arpa.2021-0423-sa] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT The gold standard test to identify the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in coronavirus disease 2019 (COVID-19) patients is the real-time reverse transcription quantitative polymerase chain reaction (RT-qPCR), but the inconclusive data and presence of false positive diagnosis remain the major problem of this approach. OBJECTIVE To compare the fitness of two primers sets to the SARS-CoV-2 nucleocapsid phosphoprotein (NP) gene in the molecular diagnosis of COVID-19, we verify the inconclusive data and confidence of high cycle threshold (Ct) values in the SARS-CoV-2 detection. DESIGN The 970 patient samples were tested using United States Centers for Disease Control and Prevention protocol. We compared the fitness of two primers sets to two different regions of NP gene. In addition, we check the consistency of positive samples with high Ct values by retesting extracted SARS-CoV-2 RNA or by second testing of patients. RESULTS The N1 and N2 displayed similar fitness during testing with no differences between Ct values. Then, we verified security range Cts related to positive diagnostic with Ct above 34 failing in 21/32 (65.6%) after retesting of samples. The samples patients with Ct above 34.89 that were doubly positive revealed a low sensitivity (52.4%) and specificity (63.6%) of the test in samples with Ct above 34. CONCLUSIONS It is secure to use one primer set to the NP gene to identify SARS-CoV-2 in samples. However, samples with high Ct values may be considered inconclusive and retested to avoid false positive diagnosis.
Collapse
Affiliation(s)
- Raphael Contelli Klein
- Infectious Agents and Vectors Research Group at the Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Mary Hellen Fabres Klein
- Infectious Agents and Vectors Research Group at the Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Larissa Gomes Barbosa
- Infectious Agents and Vectors Research Group at the Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Lívia Vasconcelos Gonzaga Knnup
- Infectious Agents and Vectors Research Group at the Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Larissa Paola Rodrigues Venâncio
- Infectious Agents and Vectors Research Group at the Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Jonilson Berlink Lima
- Infectious Agents and Vectors Research Group at the Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Théo Araújo-Santos
- Infectious Agents and Vectors Research Group at the Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| |
Collapse
|
200
|
Loretelli C, Abdelsalam A, D'Addio F, Ben Nasr M, Assi E, Usuelli V, Maestroni A, Seelam AJ, Ippolito E, Di Maggio S, Loreggian L, Radovanovic D, Vanetti C, Yang J, El Essawy B, Rossi A, Pastore I, Montefusco L, Lunati ME, Bolla AM, Biasin M, Antinori S, Santus P, Riva A, Zuccotti G, Galli M, Rusconi S, Fiorina P. PD-1 blockade counteracts post-COVID-19 immune abnormalities and stimulates the anti-SARS-CoV-2 immune response. JCI Insight 2021; 6:146701. [PMID: 34784300 PMCID: PMC8783674 DOI: 10.1172/jci.insight.146701] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
A substantial proportion of patients who have recovered from coronavirus disease-2019 (COVID-19) experience COVID-19–related symptoms even months after hospital discharge. We extensively immunologically characterized patients who recovered from COVID-19. In these patients, T cells were exhausted, with increased PD-1+ T cells, as compared with healthy controls. Plasma levels of IL-1β, IL-1RA, and IL-8, among others, were also increased in patients who recovered from COVID-19. This altered immunophenotype was mirrored by a reduced ex vivo T cell response to both nonspecific and specific stimulation, revealing a dysfunctional status of T cells, including a poor response to SARS-CoV-2 antigens. Altered levels of plasma soluble PD-L1, as well as of PD1 promoter methylation and PD1-targeting miR–15-5p, in CD8+ T cells were also observed, suggesting abnormal function of the PD-1/PD-L1 immune checkpoint axis. Notably, ex vivo blockade of PD-1 nearly normalized the aforementioned immunophenotype and restored T cell function, reverting the observed post–COVID-19 immune abnormalities; indeed, we also noted an increased T cell–mediated response to SARS-CoV-2 peptides. Finally, in a neutralization assay, PD-1 blockade did not alter the ability of T cells to neutralize SARS-CoV-2 spike pseudotyped lentivirus infection. Immune checkpoint blockade ameliorates post–COVID-19 immune abnormalities and stimulates an anti–SARS-CoV-2 immune response.
Collapse
Affiliation(s)
- Cristian Loretelli
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Ahmed Abdelsalam
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Francesca D'Addio
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, United States of America
| | - Emma Assi
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Vera Usuelli
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Anna Maestroni
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Andy Joe Seelam
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Elio Ippolito
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Stefania Di Maggio
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Lara Loreggian
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Dejan Radovanovic
- Division of Respiratory Diseases, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Claudia Vanetti
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Jun Yang
- Institute of Organ Transplantation, Tongji Hospital and Medical College,, Wuhan, China
| | | | - Antonio Rossi
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Laura Montefusco
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | | | - Andrea M Bolla
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Spinello Antinori
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Pierachille Santus
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Massimo Galli
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Stefano Rusconi
- Infectious Diseases Unit, Legnano General Hospital, ASST Ovest Milanese, Legnano, Italy
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, United States of America
| |
Collapse
|