401
|
Ortega MA, García-Montero C, Fraile-Martinez O, Colet P, Baizhaxynova A, Mukhtarova K, Alvarez-Mon M, Kanatova K, Asúnsolo A, Sarría-Santamera A. Recapping the Features of SARS-CoV-2 and Its Main Variants: Status and Future Paths. J Pers Med 2022; 12:995. [PMID: 35743779 PMCID: PMC9225183 DOI: 10.3390/jpm12060995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/06/2022] [Accepted: 06/16/2022] [Indexed: 12/14/2022] Open
Abstract
Over the two years that we have been experiencing the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) pandemic, our challenges have been the race to develop vaccines and the difficulties in fighting against new variants due to the rapid ability of the virus to evolve. In this sense, different organizations have identified and classified the different variants that have been emerging, distinguishing between variants of concern (VOC), variants of interest (VOI), or variants under monitoring (VUM). The following review aims to describe the latest updates focusing on VOC and already de-escalated variants, as well as to describe the impact these have had on the global situation. Understanding the intrinsic properties of SARS-CoV-2 and its interaction with the immune system and vaccination is essential to make out the underlying mechanisms that have led to the appearance of these variants, helping to determine the next steps for better public management of this pandemic.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Paolo Colet
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Ardak Baizhaxynova
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Kymbat Mukhtarova
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (M.A.O.); (C.G.-M.); (O.F.-M.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Kaznagul Kanatova
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| | - Angel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Antonio Sarría-Santamera
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan; (P.C.); (A.B.); (K.M.); (K.K.)
| |
Collapse
|
402
|
Fleming-Dutra KE, Britton A, Shang N, Derado G, Link-Gelles R, Accorsi EK, Smith ZR, Miller J, Verani JR, Schrag SJ. Association of Prior BNT162b2 COVID-19 Vaccination With Symptomatic SARS-CoV-2 Infection in Children and Adolescents During Omicron Predominance. JAMA 2022; 327:2210-2219. [PMID: 35560036 PMCID: PMC9107063 DOI: 10.1001/jama.2022.7493] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Efficacy of 2 doses of the BNT162b2 COVID-19 vaccine (Pfizer-BioNTech) against COVID-19 was high in pediatric trials conducted before the SARS-CoV-2 Omicron variant emerged. Among adults, estimated vaccine effectiveness (VE) of 2 BNT162b2 doses against symptomatic Omicron infection was reduced compared with prior variants, waned rapidly, and increased with a booster. OBJECTIVE To evaluate the association of symptomatic infection with prior vaccination with BNT162b2 to estimate VE among children and adolescents during Omicron variant predominance. DESIGN, SETTING, AND PARTICIPANTS A test-negative, case-control analysis was conducted using data from 6897 pharmacy-based, drive-through SARS-CoV-2 testing sites across the US from a single pharmacy chain in the Increasing Community Access to Testing platform. This analysis included 74 208 tests from children 5 to 11 years of age and 47 744 tests from adolescents 12 to 15 years of age with COVID-19-like illness who underwent SARS-CoV-2 nucleic acid amplification testing from December 26, 2021, to February 21, 2022. EXPOSURES Two BNT162b2 doses 2 weeks or more before SARS-CoV-2 testing vs no vaccination for children; 2 or 3 doses 2 weeks or more before testing vs no vaccination for adolescents (who are recommended to receive a booster dose). MAIN OUTCOMES AND MEASURES Symptomatic infection. The adjusted odds ratio (OR) for the association of prior vaccination and symptomatic SARS-CoV-2 infection was used to estimate VE: VE = (1 - OR) × 100%. RESULTS A total of 30 999 test-positive cases and 43 209 test-negative controls were included from children 5 to 11 years of age, as well as 22 273 test-positive cases and 25 471 test-negative controls from adolescents 12 to 15 years of age. The median age among those with included tests was 10 years (IQR, 7-13); 61 189 (50.2%) were female, 75 758 (70.1%) were White, and 29 034 (25.7%) were Hispanic/Latino. At 2 to 4 weeks after dose 2, among children, the adjusted OR was 0.40 (95% CI, 0.35-0.45; estimated VE, 60.1% [95% CI, 54.7%-64.8%]) and among adolescents, the OR was 0.40 (95% CI, 0.29-0.56; estimated VE, 59.5% [95% CI, 44.3%-70.6%]). During month 2 after dose 2, among children, the OR was 0.71 (95% CI, 0.67-0.76; estimated VE, 28.9% [95% CI, 24.5%-33.1%]) and among adolescents, the OR was 0.83 (95% CI, 0.76-0.92; estimated VE, 16.6% [95% CI, 8.1%-24.3%]). Among adolescents, the booster dose OR 2 to 6.5 weeks after the dose was 0.29 (95% CI, 0.24-0.35; estimated VE, 71.1% [95% CI, 65.5%-75.7%]). CONCLUSIONS AND RELEVANCE Among children and adolescents, estimated VE for 2 doses of BNT162b2 against symptomatic infection was modest and decreased rapidly. Among adolescents, the estimated effectiveness increased after a booster dose.
Collapse
Affiliation(s)
| | - Amadea Britton
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
- Epidemic Intelligence Service, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Nong Shang
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Gordana Derado
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Ruth Link-Gelles
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Emma K. Accorsi
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
- Epidemic Intelligence Service, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Zachary R. Smith
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Joseph Miller
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Jennifer R. Verani
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Stephanie J. Schrag
- US Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| |
Collapse
|
403
|
Affiliation(s)
- Sophie E Katz
- Department of Pediatrics, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kathryn Edwards
- Department of Pediatrics, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
404
|
Lewis NM, Self WH, Gaglani M, Ginde AA, Douin DJ, Keipp Talbot H, Casey JD, Mohr NM, Zepeski A, Ghamande SA, McNeal TA, Shapiro NI, Gibbs KW, Files DC, Hager DN, Shehu A, Prekker ME, Erickson HL, Gong MN, Mohamed A, Johnson NJ, Srinivasan V, Steingrub JS, Peltan ID, Brown SM, Martin ET, Monto AS, Khan A, Busse LW, ten Lohuis CC, Duggal A, Wilson JG, Gordon AJ, Qadir N, Chang SY, Mallow C, Rivas C, Babcock HM, Kwon JH, Exline MC, Lauring AS, Halasa N, Chappell JD, Grijalva CG, Rice TW, Rhoads JP, Jones ID, Stubblefield WB, Baughman A, Womack KN, Lindsell CJ, Hart KW, Zhu Y, Adams K, Patel MM, Tenforde MW. Effectiveness of the Ad26.COV2.S (Johnson & Johnson) Coronavirus Disease 2019 (COVID-19) Vaccine for Preventing COVID-19 Hospitalizations and Progression to High Disease Severity in the United States. Clin Infect Dis 2022; 75:S159-S166. [PMID: 35675695 PMCID: PMC9214149 DOI: 10.1093/cid/ciac439] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Indexed: 01/19/2023] Open
Abstract
Background . Adults in the United States (US) began receiving the adenovirus vector coronavirus disease 2019 (COVID-19) vaccine, Ad26.COV2.S (Johnson & Johnson [Janssen]), in February 2021. We evaluated Ad26.COV2.S vaccine effectiveness (VE) against COVID-19 hospitalization and high disease severity during the first 10 months of its use. Methods . In a multicenter case-control analysis of US adults (≥18 years) hospitalized 11 March to 15 December 2021, we estimated VE against susceptibility to COVID-19 hospitalization (VEs), comparing odds of prior vaccination with a single dose Ad26.COV2.S vaccine between hospitalized cases with COVID-19 and controls without COVID-19. Among hospitalized patients with COVID-19, we estimated VE against disease progression (VEp) to death or invasive mechanical ventilation (IMV), comparing odds of prior vaccination between patients with and without progression. Results . After excluding patients receiving mRNA vaccines, among 3979 COVID-19 case-patients (5% vaccinated with Ad26.COV2.S) and 2229 controls (13% vaccinated with Ad26.COV2.S), VEs of Ad26.COV2.S against COVID-19 hospitalization was 70% (95% confidence interval [CI]: 63-75%) overall, including 55% (29-72%) among immunocompromised patients, and 72% (64-77%) among immunocompetent patients, for whom VEs was similar at 14-90 days (73% [59-82%]), 91-180 days (71% [60-80%]), and 181-274 days (70% [54-81%]) postvaccination. Among hospitalized COVID-19 case-patients, VEp was 46% (18-65%) among immunocompetent patients. Conclusions . The Ad26.COV2.S COVID-19 vaccine reduced the risk of COVID-19 hospitalization by 72% among immunocompetent adults without waning through 6 months postvaccination. After hospitalization for COVID-19, vaccinated immunocompetent patients were less likely to require IMV or die compared to unvaccinated immunocompetent patients.
Collapse
Affiliation(s)
- Nathaniel M. Lewis
- Corresponding Author Nathaniel M. Lewis, Influenza Prevention and Control Team, Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Mailstop 46 24/7, Atlanta, Georgia, 30329 ()
| | - Wesley H. Self
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Manjusha Gaglani
- Baylor Scott & White Health, Temple, Texas, USA,Texas A&M University College of Medicine, Temple, Texas, USA
| | - Adit A. Ginde
- University of Colorado School of Medicine, Aurora, Colorado, USA
| | - David J. Douin
- University of Colorado School of Medicine, Aurora, Colorado, USA
| | - H. Keipp Talbot
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | | | | | | - Kevin W. Gibbs
- Wake Forest University Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - D. Clark Files
- Wake Forest University Baptist Medical Center, Winston-Salem, North Carolina, USA
| | | | - Arber Shehu
- Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | | | | - Michelle N. Gong
- Montefiore Healthcare Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Amira Mohamed
- Montefiore Healthcare Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | | | | - Ithan D. Peltan
- Intermountain Medical Center and University of Utah, Salt Lake City, Utah, USA
| | - Samuel M. Brown
- Intermountain Medical Center and University of Utah, Salt Lake City, Utah, USA
| | - Emily T. Martin
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Arnold S. Monto
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Akram Khan
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | | | | | | | | | | | - Nida Qadir
- David Geffen School of Medicine at UCLA, Ronald Reagan-UCLA Medical Center, Los Angeles, California, USA
| | - Steven Y. Chang
- David Geffen School of Medicine at UCLA, Ronald Reagan-UCLA Medical Center, Los Angeles, California, USA
| | | | | | | | | | | | - Adam S. Lauring
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Natasha Halasa
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | - Todd W. Rice
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Ian D. Jones
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | | | | - Yuwei Zhu
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | |
Collapse
|
405
|
Hajjaji N, Lepoutre K, Lakhdar S, Bécourt S, Bellier C, Kaczmarek E, Broyelle A, Giscard S, Lartigau E. 16 Months Follow Up of Patients’ Behavior and Mild COVID-19 Patterns in a Large Cohort of Cancer Patients During the Pandemic. Front Oncol 2022; 12:901426. [PMID: 35747798 PMCID: PMC9209649 DOI: 10.3389/fonc.2022.901426] [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: 03/21/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Acute severe forms of COVID-19 infection are more likely in cancer patients and growing attention has been given to the persistent symptoms of the disease after severe COVID-19. However, mild illness is the dominant clinical presentation of COVID-19 infection. To investigate patients’ behavior and the short- and longer-term pattern of the disease in cancer patients with mild COVID infection, a longitudinal online survey was conducted for 16 months during the pandemic in a large cohort of cancer patients from a French COVID-19 hot spot. An online questionnaire was administered at three time points between the first wave of the pandemic in France and the fourth wave. The questionnaire was completed by 1415 to 2224 patients, which queried patients’ demographics, their behavior, and COVID infection patterns. Seventy percent of the patients were female, and 40% had a comorbid condition. More than one-third of the participants had breast cancer, and half were survivors. The rate of infection was 30% during wave 1 and 10% in wave 4; most patients had a mild COVID-19 infection. Twenty-five percent of infected patients during wave 4 did not seek medical advice. At wave 4, 87% of the patients received at least one dose of vaccine. Systematic compliance to shielding measures decreased over time. The short-term pattern of mild COVID changed between wave 1 and wave 4. Twenty-two percent of infected patients experienced persistent signs for more than 6 months with a negative impact on sleep, social behavior, and increased consumption of stress-relieving drugs. Our results showed a high prevalence of long-lasting symptoms in cancer patients with mild COVID-19 infection and inadequate behavior toward the disease and prevention measures among patients.
Collapse
Affiliation(s)
- Nawale Hajjaji
- Medical Oncology department, Oscar Lambret Cancer Center, Lille, France
- Inserm, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Univ. Lille, Lille, France
- *Correspondence: Nawale Hajjaji,
| | | | - Sarra Lakhdar
- Medical Oncology department, Oscar Lambret Cancer Center, Lille, France
| | - Stéphanie Bécourt
- Medical Oncology department, Oscar Lambret Cancer Center, Lille, France
| | - Charlotte Bellier
- Medical Oncology department, Oscar Lambret Cancer Center, Lille, France
| | - Emilie Kaczmarek
- Medical Oncology department, Oscar Lambret Cancer Center, Lille, France
| | - Antonin Broyelle
- Medical Oncology department, Oscar Lambret Cancer Center, Lille, France
| | | | - Eric Lartigau
- Radiotherapy department, Oscar Lambret Cancer Center, Lille, France
| |
Collapse
|
406
|
Maslo C, Toubkin M. Characteristics and Outcomes of Hospitalized Patients in South Africa During the COVID-19 Omicron Wave-Reply. JAMA 2022; 327:2148. [PMID: 35670790 DOI: 10.1001/jama.2022.5575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
| | - Mande Toubkin
- Netcare Ltd South Africa, Johannesburg, South Africa
| |
Collapse
|
407
|
Richterman A, Behrman A, Brennan PJ, O’Donnell JA, Snider CK, Chaiyachati KH. Durability of Severe Acute Respiratory Syndrome Coronavirus 2 Messenger RNA Booster Vaccine Protection Against Omicron Among Healthcare Workers With a Vaccine Mandate. Clin Infect Dis 2022; 76:e319-e326. [PMID: 35666508 PMCID: PMC9214168 DOI: 10.1093/cid/ciac454] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/25/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant has spread rapidly throughout the world since being identified in South Africa in November 2021. Few studies have assessed primary series and booster vaccine effectiveness against Omicron among US healthcare workers. METHODS We conducted a test-negative case-control design to estimate BNT162b2 and mRNA1273 primary vaccination and booster effectiveness against SARS-CoV-2 infection and symptomatic coronavirus disease 2019 during an Omicron surge among employees of the University of Pennsylvania Health System. The study period was between 1 July 2021 and 5 April 2022. We defined the Delta period as 1 July to 12 December 2021 and the Omicron period as beginning 12 December 21. RESULTS Our sample included 14 520 tests (2776 [19%] positive)-7422 (506 [7%] positive) during Delta and 7098 (2270 [32%] positive) during Omicron. Benchmarked against Delta, the vaccine effectiveness of 2 vaccine doses was lower during Omicron, with no significant protection against infection. Booster doses added significant protection, although they also showed reduced effectiveness during Omicron. Compared with findings in employees who had received 2 vaccine doses, 3 doses of BNT162b2 had a relative effectiveness of 50% (95% confidence interval, 42%-56%) during Omicron, relative to 78% (63%-87%) during Delta; 3 doses of mRNA1273 had a relative effectiveness of 56% (45%-65%) during Omicron, relative to 96% (82%-99%) during Delta. Restricting the sample to symptomatic tests yielded similar results to our primary analysis. After initial waning in BNT162b2 booster protection against infection, it remained largely stable for ≥16 weeks after vaccination. CONCLUSIONS Our findings provide a strong rationale for boosters among healthcare workers in the Omicron era.
Collapse
Affiliation(s)
- Aaron Richterman
- Correspondence: Aaron Richterman, Hospital of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA, 19104 ()
| | - Amy Behrman
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,The University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Patrick J Brennan
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,The University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Judith A O’Donnell
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,The University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Christopher K Snider
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,The University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Krisda H Chaiyachati
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,The University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| |
Collapse
|
408
|
Chen Z, Li J, Li T, Fan T, Meng C, Li C, Kang J, Chai L, Hao Y, Tang Y, Al-Hartomy OA, Wageh S, Al-Sehemi AG, Luo Z, Yu J, Shao Y, Li D, Feng S, Liu WJ, He Y, Ma X, Xie Z, Zhang H. A CRISPR/Cas12a empowered surface plasmon resonance platform for rapid and specific diagnosis of the Omicron variant of SARS-CoV-2. Natl Sci Rev 2022; 9:nwac104. [PMID: 35992231 PMCID: PMC9385456 DOI: 10.1093/nsr/nwac104] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 11/14/2022] Open
Abstract
The outbreak of the COVID-19 pandemic was partially due to the challenge of identifying asymptomatic and presymptomatic carriers of the virus, and thus highlights a strong motivation for diagnostics with high sensitivity that can be rapidly deployed. On the other hand, several concerning SARS-CoV-2 variants, including Omicron, are required to be identified as soon as the samples are identified as ‘positive’. Unfortunately, a traditional PCR test does not allow their specific identification. Herein, for the first time, we have developed MOPCS (Methodologies of Photonic CRISPR Sensing), which combines an optical sensing technology-surface plasmon resonance (SPR) with the ‘gene scissors’ clustered regularly interspaced short palindromic repeat (CRISPR) technique to achieve both high sensitivity and specificity when it comes to measurement of viral variants. MOPCS is a low-cost, CRISPR/Cas12a-system-empowered SPR gene-detecting platform that can analyze viral RNA, without the need for amplification, within 38 min from sample input to results output, and achieve a limit of detection of 15 fM. MOPCS achieves a highly sensitive analysis of SARS-CoV-2, and mutations appear in variants B.1.617.2 (Delta), B.1.1.529 (Omicron) and BA.1 (a subtype of Omicron). This platform was also used to analyze some recently collected patient samples from a local outbreak in China, identified by the Centers for Disease Control and Prevention. This innovative CRISPR-empowered SPR platform will further contribute to the fast, sensitive and accurate detection of target nucleic acid sequences with single-base mutations.
Collapse
Affiliation(s)
- Zhi Chen
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
| | - Jinfeng Li
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
- Shenzhen International Institute for Biomedical Research , Shenzhen 518116 , Guangdong , China
| | - Tianzhong Li
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
- Shenzhen International Institute for Biomedical Research , Shenzhen 518116 , Guangdong , China
| | - Taojian Fan
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
| | - Changle Meng
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
| | - Chaozhou Li
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
| | - Jianlong Kang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
| | - Luxiao Chai
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
| | - Yabin Hao
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
- Shenzhen Han's Tech Limited Company. Shenzhen 518000 , China
| | - Yuxuan Tang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
- Shenzhen Metasensing Tech Limited Company. Shenzhen 518000 , China
| | - Omar A Al-Hartomy
- Department of Physics, Faculty of Science, King Abdulaziz University , Jeddah 21589, Saudi Arabia
| | - Swelm Wageh
- Department of Physics, Faculty of Science, King Abdulaziz University , Jeddah 21589, Saudi Arabia
| | - Abdullah G Al-Sehemi
- Research Center for Advanced Materials Science (RCAMS), King Khalid University , Abha 61413, Saudi Arabia
- Department of Chemistry, College of Science, King Khalid University , Abha 61413, Saudi Arabia
| | - Zhiguang Luo
- Zhongmin (Shenzhen) intelligent ecology Co. , Ltd., Shenzhen 518055 , China
| | - Jiangtian Yu
- Shenzhen International Institute for Biomedical Research , Shenzhen 518116 , Guangdong , China
| | - Yonghong Shao
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University , Shenzhen 518060 , China
| | - Defa Li
- Department of Laboratory Medicine, Shenzhen Children's Hospital , Shenzhen 518038 , China
| | - Shuai Feng
- Optoelectronics Research Center, School of Science, Minzu University of China , Beijing 100081 , China
| | - William J Liu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing 102206 , China
- Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences , Beijing 102206 , China
| | - Yaqing He
- Shenzhen Center for Disease Control and Prevention , Shenzhen 518055 , China
| | - Xiaopeng Ma
- Department of Respiratory, Shenzhen Children's Hospital , Shenzhen 518038 , China
| | - Zhongjian Xie
- Institute of Pediatrics, Shenzhen Children's Hospital , Shenzhen 518038 , China
| | - Han Zhang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education; Shenzhen Institute of Translational Medicine; Department of Otolaryngology, Shenzhen Second People's Hospital; the First Affiliated Hospital; Institute of Microscale Optoelectronics, Shenzhen University , Shenzhen 518060 , China
| |
Collapse
|
409
|
Lundberg AL, Lorenzo-Redondo R, Hultquist JF, Hawkins CA, Ozer EA, Welch SB, Prasad PVV, Achenbach CJ, White JI, Oehmke JF, Murphy RL, Havey RJ, Post LA. Overlapping Delta and Omicron Outbreaks During the COVID-19 Pandemic: Dynamic Panel Data Estimates. JMIR Public Health Surveill 2022; 8:e37377. [PMID: 35500140 PMCID: PMC9169703 DOI: 10.2196/37377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/25/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The Omicron variant of SARS-CoV-2 is more transmissible than prior variants of concern (VOCs). It has caused the largest outbreaks in the pandemic, with increases in mortality and hospitalizations. Early data on the spread of Omicron were captured in countries with relatively low case counts, so it was unclear how the arrival of Omicron would impact the trajectory of the pandemic in countries already experiencing high levels of community transmission of Delta. OBJECTIVE The objective of this study is to quantify and explain the impact of Omicron on pandemic trajectories and how they differ between countries that were or were not in a Delta outbreak at the time Omicron occurred. METHODS We used SARS-CoV-2 surveillance and genetic sequence data to classify countries into 2 groups: those that were in a Delta outbreak (defined by at least 10 novel daily transmissions per 100,000 population) when Omicron was first sequenced in the country and those that were not. We used trend analysis, survival curves, and dynamic panel regression models to compare outbreaks in the 2 groups over the period from November 1, 2021, to February 11, 2022. We summarized the outbreaks in terms of their peak rate of SARS-CoV-2 infections and the duration of time the outbreaks took to reach the peak rate. RESULTS Countries that were already in an outbreak with predominantly Delta lineages when Omicron arrived took longer to reach their peak rate and saw greater than a twofold increase (2.04) in the average apex of the Omicron outbreak compared to countries that were not yet in an outbreak. CONCLUSIONS These results suggest that high community transmission of Delta at the time of the first detection of Omicron was not protective, but rather preluded larger outbreaks in those countries. Outbreak status may reflect a generally susceptible population, due to overlapping factors, including climate, policy, and individual behavior. In the absence of strong mitigation measures, arrival of a new, more transmissible variant in these countries is therefore more likely to lead to larger outbreaks. Alternately, countries with enhanced surveillance programs and incentives may be more likely to both exist in an outbreak status and detect more cases during an outbreak, resulting in a spurious relationship. Either way, these data argue against herd immunity mitigating future outbreaks with variants that have undergone significant antigenic shifts.
Collapse
Affiliation(s)
- Alexander L Lundberg
- Buehler Center for Health Policy and Economics, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
- Department of Emergency Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Center for Pathogen Genomics and Microbial Evolution, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Center for Pathogen Genomics and Microbial Evolution, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Claudia A Hawkins
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Center for Global Communicable and Emerging Infectious Diseases, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Egon A Ozer
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Center for Pathogen Genomics and Microbial Evolution, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Sarah B Welch
- Buehler Center for Health Policy and Economics, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
- Department of Emergency Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - P V Vara Prasad
- Sustainable Intensification Innovation Lab, Kansas State University, Manhattan, KS, United States
| | - Chad J Achenbach
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Robert J Havey, MD Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Janine I White
- Buehler Center for Health Policy and Economics, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - James F Oehmke
- Buehler Center for Health Policy and Economics, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Robert L Murphy
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Robert J Havey, MD Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Robert J Havey
- Robert J Havey, MD Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Medicine, General Internal Medicine and Geriatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lori A Post
- Buehler Center for Health Policy and Economics, Robert J Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
- Department of Emergency Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
410
|
Amanatidou E, Gkiouliava A, Pella E, Serafidi M, Tsilingiris D, Vallianou NG, Karampela I, Dalamaga M. Breakthrough infections after COVID-19 vaccination: Insights, perspectives and challenges. Metabol Open 2022; 14:100180. [PMID: 35313532 PMCID: PMC8928742 DOI: 10.1016/j.metop.2022.100180] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 12/19/2022] Open
Abstract
Vaccination programs against SARS-CoV-2 constitute the mainstay of public health interventions against the global COVID-19 pandemic. Currently available vaccines have shown 90% or better rates of protection against severe disease and mortality. Barely a year after vaccines became available, the Omicron variant and its unprecedented speed of transmission has posed a new challenge. Overall, Omicron presents increased immune escape, transmissibility, and decreased pathogenicity. Vaccines do not offer a full protection against SARS-CoV-2 acquisition, since "breakthrough" infections may occur in fully vaccinated individuals, who may in turn spread the virus to others. Breakthrough infections may be causally related to the viral profile (viral variant and load, incubation period, transmissibility, pathogenicity, immune evasion), immunity characteristics (mucosal versus systemic immunity, duration of immunity, etc.), host determinants (age, comorbidities, immune status, immunosuppressive drugs) and vaccination properties (platform, antigen dose, dose number, dose interval, route of administration). Determining the rate of breakthrough infections may be challenging and necessitates the conduction of population-based studies regarding vaccine effectiveness as well as neutralizing antibody testing, a surrogate of immune protection. In this review, we analyze the causes of breakthrough infections, their clinical consequences (severity of infection and transmission), methods of determining their incidence as well as challenges and perspectives. Long COVID as well as multi-inflammatory syndrome in adolescents may be significantly reduced in breakthrough infections. The need for universal pancoranavirus vaccines that would aim at protecting against a plethora of SARS-CoV-2 variants as well as emerging variants is discussed. Finally, novel vaccine strategies, such as nasal vaccines, may confer robust mucosal and systemic protection, reducing efficiently transmission.
Collapse
Affiliation(s)
- Evropi Amanatidou
- Laboratory of Biomathematics, School of Medicine, University of Thessaly, Larissa, Greece
| | - Anna Gkiouliava
- Laboratory of Biomathematics, School of Medicine, University of Thessaly, Larissa, Greece
| | - Eva Pella
- Laboratory of Biomathematics, School of Medicine, University of Thessaly, Larissa, Greece
| | - Maria Serafidi
- Laboratory of Biomathematics, School of Medicine, University of Thessaly, Larissa, Greece
| | - Dimitrios Tsilingiris
- First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, Chaidari, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
411
|
Liu X, Munro APS, Feng S, Janani L, Aley PK, Babbage G, Baxter D, Bula M, Cathie K, Chatterjee K, Dejnirattisai W, Dodd K, Enever Y, Qureshi E, Goodman AL, Green CA, Harndahl L, Haughney J, Hicks A, van der Klaauw AA, Kwok J, Libri V, Llewelyn MJ, McGregor AC, Minassian AM, Moore P, Mughal M, Mujadidi YF, Holliday K, Osanlou O, Osanlou R, Owens DR, Pacurar M, Palfreeman A, Pan D, Rampling T, Regan K, Saich S, Serafimova T, Saralaya D, Screaton GR, Sharma S, Sheridan R, Sturdy A, Supasa P, Thomson EC, Todd S, Twelves C, Read RC, Charlton S, Hallis B, Ramsay M, Andrews N, Lambe T, Nguyen-Van-Tam JS, Cornelius V, Snape MD, Faust SN. Persistence of immunogenicity after seven COVID-19 vaccines given as third dose boosters following two doses of ChAdOx1 nCov-19 or BNT162b2 in the UK: Three month analyses of the COV-BOOST trial. J Infect 2022; 84:795-813. [PMID: 35405168 PMCID: PMC8993491 DOI: 10.1016/j.jinf.2022.04.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVES To evaluate the persistence of immunogenicity three months after third dose boosters. METHODS COV-BOOST is a multicentre, randomised, controlled, phase 2 trial of seven COVID-19 vaccines used as a third booster dose. The analysis was conducted using all randomised participants who were SARS-CoV-2 naïve during the study. RESULTS Amongst the 2883 participants randomised, there were 2422 SARS-CoV-2 naïve participants until D84 visit included in the analysis with median age of 70 (IQR: 30-94) years. In the participants who had two initial doses of ChAdOx1 nCov-19 (Oxford-AstraZeneca; hereafter referred to as ChAd), schedules using mRNA vaccines as third dose have the highest anti-spike IgG at D84 (e.g. geometric mean concentration of 8674 ELU/ml (95% CI: 7461-10,085) following ChAd/ChAd/BNT162b2 (Pfizer-BioNtech, hearafter referred to as BNT)). However, in people who had two initial doses of BNT there was no significant difference at D84 in people given ChAd versus BNT (geometric mean ratio (GMR) of 0.95 (95%CI: 0.78, 1.15). Also, people given Ad26.COV2.S (Janssen; hereafter referred to as Ad26) as a third dose had significantly higher anti-spike IgG at D84 than BNT (GMR of 1.20, 95%CI: 1.01,1.43). Responses at D84 between people who received BNT (15 μg) or BNT (30 μg) after ChAd/ChAd or BNT/BNT were similar, with anti-spike IgG GMRs of half-BNT (15 μg) versus BNT (30 μg) ranging between 0.74-0.86. The decay rate of cellular responses were similar between all the vaccine schedules and doses. CONCLUSIONS 84 days after a third dose of COVID-19 vaccine the decay rates of humoral response were different between vaccines. Adenoviral vector vaccine anti-spike IgG concentrations at D84 following BNT/BNT initial doses were similar to or even higher than for a three dose (BNT/BNT/BNT) schedule. Half dose BNT immune responses were similar to full dose responses. While high antibody tires are desirable in situations of high transmission of new variants of concern, the maintenance of immune responses that confer long-lasting protection against severe disease or death is also of critical importance. Policymakers may also consider adenoviral vector, fractional dose of mRNA, or other non-mRNA vaccines as third doses.
Collapse
Affiliation(s)
- Xinxue Liu
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Alasdair P S Munro
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Shuo Feng
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Leila Janani
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Gavin Babbage
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Marcin Bula
- NIHR Liverpool and Broadgreen Clinical Research Facility, Liverpool, UK
| | - Katrina Cathie
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Krishna Chatterjee
- NIHR Cambridge Clinical Research Facility, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Wanwisa Dejnirattisai
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kate Dodd
- NIHR Liverpool and Broadgreen Clinical Research Facility, Liverpool, UK
| | | | - Ehsaan Qureshi
- NIHR/Wellcome Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Anna L Goodman
- Department of Infection, Guy's and St Thomas' NHS Foundation Trust, London, UK; MRC Clinical Trials Unit, University College London, London, UK
| | - Christopher A Green
- NIHR/Wellcome Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Linda Harndahl
- Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | - John Haughney
- Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, Glasgow, UK
| | | | - Agatha A van der Klaauw
- Wellcome-MRC Institute of Metabolic Science, Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK
| | - Jonathan Kwok
- Cancer Research UK Oxford Centre, University of Oxford, Oxford, UK
| | - Vincenzo Libri
- NIHR UCLH Clinical Research Facility and NIHR UCLH Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | | | - Alastair C McGregor
- Department of Infectious Diseases and Tropical Medicine, London Northwest University Healthcare, London, UK
| | - Angela M Minassian
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | - Kyra Holliday
- NIHR Leeds Clinical Research Facility, Leeds Teaching Hospitals Trust and University of Leeds, Leeds, UK
| | - Orod Osanlou
- Public Health Wales, Betsi Cadwaladr University Health Board, Bangor University, Bangor, UK
| | | | - Daniel R Owens
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Mihaela Pacurar
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Adrian Palfreeman
- University Hospitals of Leicester NHS Trust, University of Leicester, Leicester, UK
| | - Daniel Pan
- University Hospitals of Leicester NHS Trust, University of Leicester, Leicester, UK
| | - Tommy Rampling
- NIHR UCLH Clinical Research Facility and NIHR UCLH Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | - Karen Regan
- Bradford Institute for Health Research and Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Stephen Saich
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Teona Serafimova
- Department of Infection, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Dinesh Saralaya
- Bradford Institute for Health Research and Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Gavin R Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sunil Sharma
- University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Ray Sheridan
- Royal Devon and Exeter Hospital NHS Foundation Trust, Exeter, UK
| | - Ann Sturdy
- Department of Infectious Diseases and Tropical Medicine, London Northwest University Healthcare, London, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Emma C Thomson
- Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, Glasgow, UK; MRC University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Shirley Todd
- Royal Devon and Exeter Hospital NHS Foundation Trust, Exeter, UK
| | - Chris Twelves
- NIHR Leeds Clinical Research Facility, Leeds Teaching Hospitals Trust and University of Leeds, Leeds, UK
| | - Robert C Read
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | | | | | - Mary Ramsay
- UK Health Security Agency, Colindale, London, UK
| | - Nick Andrews
- UK Health Security Agency, Colindale, London, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
412
|
Alshukairi AN, Al-Omari A, Al-Tawfiq JA, El-Kafrawy SA, El-Daly MM, Hassan AM, Faizo AA, Alandijany TA, Dada A, Saeedi MF, Alhamlan FS, Al Hroub MK, Qushmaq I, Azhar EI. Active viral shedding in a vaccinated hospitalized patient infected with the delta variant (B.1.617.2) of SARS-CoV-2 and challenges of de-isolation. J Infect Public Health 2022; 15:628-630. [PMID: 35576779 PMCID: PMC9047479 DOI: 10.1016/j.jiph.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 12/02/2022] Open
Abstract
In the era of SARS-CoV-2 variants and COVID-19 vaccination, the duration of infectious viral shedding and isolation in post vaccine breakthrough infections is challenging and depends on disease severity. The current study described a case of SARS-CoV-2 Delta variant pneumonia requiring hospitalization. The patient received two doses of BNT162b2 COVID-19 vaccines, and he had positive SARS-CoV-2 viral cultures 12 days post symptom onset. The time between the second dose of vaccine and the breakthrough infection was 6 months. While immunosuppression is a known risk factor for prolonged infectious viral shedding, age and time between vaccination and breakthrough infection are important risk factors that warrant further studies.
Collapse
Affiliation(s)
- Abeer N Alshukairi
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia; College of Medicine, AlFaisal University, Riyadh, Saudi Arabia.
| | - Awad Al-Omari
- College of Medicine, AlFaisal University, Riyadh, Saudi Arabia; Department of Critical Care, Dr Sulaiman Al Habib Medical Group, Riyadh, Saudi Arabia
| | - Jaffar A Al-Tawfiq
- Infectious Disease Unit, Specialty Internal Medicine, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia; Infectious Disease Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Infectious Disease Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Sherif A El-Kafrawy
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| | - Mai M El-Daly
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| | - Ahmed M Hassan
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arwa A Faizo
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| | - Thamir A Alandijany
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| | - Ashraf Dada
- College of Medicine, AlFaisal University, Riyadh, Saudi Arabia; Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Mohammed F Saeedi
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Fatma S Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohammad K Al Hroub
- Department of Infection Control and Hospital Epidemiology, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Ismael Qushmaq
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Esam I Azhar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| |
Collapse
|
413
|
Canaday DH, Oyebanji OA, White E, Keresztesy D, Payne M, Wilk D, Carias L, Aung H, St Denis K, Sheehan ML, Berry SD, Cameron CM, Cameron MJ, Wilson BM, Balazs AB, King CL, Gravenstein S. COVID-19 vaccine booster dose needed to achieve Omicron-specific neutralisation in nursing home residents. EBioMedicine 2022; 80:104066. [PMID: 35605428 PMCID: PMC9122310 DOI: 10.1016/j.ebiom.2022.104066] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Nursing home (NH) residents have borne a disproportionate share of SARS-CoV-2 morbidity and mortality. Vaccines have limited hospitalisation and death from earlier variants in this vulnerable population. With the rise of Omicron and future variants, it is vital to sustain and broaden vaccine-induced protection. We examined the effect of boosting with BNT162b2 mRNA vaccine on humoral immunity and Omicron-specific neutralising activity among NH residents and healthcare workers (HCWs). METHODS We longitudinally enrolled 85 NH residents (median age 77) and 48 HCWs (median age 51), and sampled them after the initial vaccination series; and just before and 2 weeks after booster vaccination. Anti-spike, anti-receptor binding domain (RBD) and neutralisation titres to the original Wuhan strain and neutralisation to the Omicron strain were obtained. FINDINGS Booster vaccination significantly increased vaccine-specific anti-spike, anti-RBD, and neutralisation levels above the pre-booster levels in NH residents and HCWs, both in those with and without prior SARS-CoV-2 infection. Omicron-specific neutralisation activity was low after the initial 2 dose series with only 28% of NH residents' and 28% HCWs' titres above the assay's lower limit of detection. Omicron neutralising activity following the booster lifted 86% of NH residents and 93% of HCWs to the detectable range. INTERPRETATION With boosting, the vast majority of HCWs and NH residents developed detectable Omicron-specific neutralising activity. These data provide immunologic evidence that strongly supports booster vaccination to broaden neutralising activity and counter waning immunity in the hope it will better protect this vulnerable, high-risk population against the Omicron variant. FUNDING NIH AI129709-03S1, U01 CA260539-01, CDC 200-2016-91773, and VA BX005507-01.
Collapse
Affiliation(s)
- David H Canaday
- Case Western Reserve University School of Medicine, Cleveland, OH; Geriatric Research, Education and Clinical Center, Cleveland VA.
| | | | - Elizabeth White
- Department of Health Services, Policy, and Practice, Brown University School of Public Health, Providence, RI
| | | | - Michael Payne
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Dennis Wilk
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Lenore Carias
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Htin Aung
- Case Western Reserve University School of Medicine, Cleveland, OH
| | | | | | - Sarah D Berry
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA
| | - Cheryl M Cameron
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Mark J Cameron
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Brigid M Wilson
- Geriatric Research, Education and Clinical Center, Cleveland VA
| | | | | | - Stefan Gravenstein
- Department of Health Services, Policy, and Practice, Brown University School of Public Health, Providence, RI; Center on Innovation in Long-Term Services and Supports, Providence Veterans Administration Medical Center, Providence, RI; Division of Geriatrics and Palliative Medicine, Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
414
|
Salerno G, Gentile G, De Luca O, Costanzi G, Cirelli G, Di Simone Di Giuseppe B, Marcellini L, Anibaldi P, Marcolongo A, Simmaco M, Borro M. Age-Related Dynamics of Serum Anti-Spike IgG Ab After the Third Dose of BNT162b2 Vaccine in a Naive Health Care Workers Population. Viral Immunol 2022; 35:386-389. [PMID: 35648037 DOI: 10.1089/vim.2022.0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The kinetics of postvaccination serum anti-Spike IgG concentration were determined in 1,541 health care workers (Sant'Andrea Hospital of Rome, Italy) with no prior infection by SARS-COV-2. Anti-Spike IgG were measured at 3, 12, and 24 weeks after the completion of the primary vaccine cycle (two doses of the BNT162b2 vaccine by Biontech/Pfizer) and 3 weeks apart a third BNT162b2 dose. Stratification of the study population by age (decades from 21-30 to 61-70) highlighted that 24 weeks after cycle completion all age groups had an order of magnitude reduction in serum IgG titers. Considering older adults (age 61-70), they had significantly lower serum IgG titers at each time point compared with younger people, except after the booster dose, which induced similar and elevated IgG titers despite the age.
Collapse
Affiliation(s)
- Gerardo Salerno
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Rome, Italy.,Laboratory of Clinical Biochemistry, Sant'Andrea University Hospital, Rome, Italy
| | - Giovanna Gentile
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Rome, Italy.,Laboratory of Clinical Biochemistry, Sant'Andrea University Hospital, Rome, Italy
| | - Ottavia De Luca
- Laboratory of Clinical Biochemistry, Sant'Andrea University Hospital, Rome, Italy
| | - Giuseppe Costanzi
- Laboratory of Clinical Biochemistry, Sant'Andrea University Hospital, Rome, Italy
| | - Gloria Cirelli
- Occupational Health Department, Sant'Andrea University Hospital, Rome, Italy
| | | | - Laura Marcellini
- Occupational Health Department, Sant'Andrea University Hospital, Rome, Italy
| | - Paolo Anibaldi
- Hospital Direction and Clinical Department, Sant'Andrea University Hospital, Rome, Italy
| | - Adriano Marcolongo
- Hospital Direction and Clinical Department, Sant'Andrea University Hospital, Rome, Italy
| | - Maurizio Simmaco
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Rome, Italy.,Laboratory of Clinical Biochemistry, Sant'Andrea University Hospital, Rome, Italy
| | - Marina Borro
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Rome, Italy.,Laboratory of Clinical Biochemistry, Sant'Andrea University Hospital, Rome, Italy
| |
Collapse
|
415
|
Marra AR, Miraglia JL, Malheiros DT, Guozhang Y, Teich VD, da Silva Victor E, Rebello Pinho JR, Cypriano A, Vieira LW, Polonio M, Ornelas RH, de Oliveira SM, Borges Junior FA, Oler SCC, Schettino GDPP, de Oliveira KG, Ferraz Santana RA, de Mello Malta F, Amgarten D, Boechat AL, Trecenti NMZ, Kobayashi T, Salinas JL, Edmond MB, Rizzo LV. Effectiveness of Heterologous Coronavirus Disease 2019 (COVID-19) Vaccine Booster Dosing in Brazilian Healthcare Workers, 2021. Clin Infect Dis 2022; 76:e360-e366. [PMID: 35639918 PMCID: PMC9213833 DOI: 10.1093/cid/ciac430] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/12/2022] [Accepted: 05/24/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Little is currently known about vaccine effectiveness (VE) for either 2 doses of Oxford-AstraZeneca (ChAdOx1) viral vector vaccine or CoronaVac (Instituto Butantan) inactivated viral vaccine followed by a third dose of mRNA vaccine (Pfizer/BioNTech) among healthcare workers (HCWs). METHODS We conducted a retrospective cohort study among HCWs (aged ≥18 years) working in a private healthcare system in Brazil from January to December 2021. VE was defined as 1 - incidence rate ratio (IRR), with IRR determined using Poisson models with the occurrence of laboratory-confirmed coronavirus disease 2019 (COVID-19) infection as the outcome, adjusting for age, sex, and job type. We compared those receiving viral vector or inactivated viral primary series (2 doses) with those who received an mRNA booster. RESULTS A total of 11 427 HCWs met the inclusion criteria. COVID-19 was confirmed in 31.5% of HCWs receiving 2 doses of CoronaVac vaccine versus 0.9% of HCWs receiving 2 doses of CoronaVac vaccine with mRNA booster (P < .001) and 9.8% of HCWs receiving 2 doses of ChAdOx1 vaccine versus 1% among HCWs receiving 2 doses of ChAdOx1 vaccine with mRNA booster (P < .001). In the adjusted analyses, the estimated VE was 92.0% for 2 CoronaVac vaccines plus mRNA booster and 60.2% for 2 ChAdOx1 vaccines plus mRNA booster, when compared with those with no mRNA booster. Of 246 samples screened for mutations, 191 (77.6%) were Delta variants. CONCLUSIONS While 2 doses of ChAdOx1 or CoronaVac vaccines prevent COVID-19, the addition of a Pfizer/BioNTech booster provided significantly more protection.
Collapse
Affiliation(s)
- Alexandre R Marra
- Correspondence: A. R. Marra, University of Iowa, Department of Internal Medicine, 200 Hawkins Dr, C51-GH, Iowa City, IA 52242 (; )
| | | | | | - Yang Guozhang
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | | | | | | | - Miria Polonio
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | - Takaaki Kobayashi
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | | | - Michael B Edmond
- West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | | |
Collapse
|
416
|
Protection against Omicron conferred by mRNA primary vaccine series, boosters, and prior infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.05.26.22275639. [PMID: 35665013 PMCID: PMC9164448 DOI: 10.1101/2022.05.26.22275639] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
B ackground Prisons and jails are high-risk settings for Covid-19 transmission, morbidity, and mortality. We evaluate protection conferred by prior infection and vaccination against the SARS-CoV-2 Omicron variant within the California state prison system. M ethods We employed a test-negative design to match resident and staff cases during the Omicron wave (December 24, 2021-April 14, 2022) to controls according to a case's test-week as well as demographic, clinical, and carceral characteristics. We estimated protection against infection using conditional logistic regression, with exposure status defined by vaccination, stratified by number of mRNA doses received, and prior infection, stratified by periods before or during Delta variant predominance. R esults We matched 15,783 resident and 8,539 staff cases to 180,169 resident and 90,409 staff controls. Among cases, 29.7% and 2.2% were infected before or during the emergence of the Delta variant, respectively; 30.6% and 36.3% were vaccinated with two or three doses, respectively. Estimated protection from Omicron infection for two and three doses were 14.9% (95% Confidence Interval [CI], 12.3-19.7%) and 43.2% (42.2-47.4%) for those without known prior infections, 47.8% (95% CI, 46.6-52.8%) and 61.3% (95% CI, 60.7-64.8%) for those infected before the emergence of Delta, and 73.1% (95% CI, 69.8-80.1%) and 86.8% (95% CI, 82.1-92.7) for those infected during the period of Delta predominance. C onclusion A third mRNA dose provided significant, additional protection over two doses, including among individuals with prior infection. Our findings suggest that vaccination should remain a priority-even in settings with high levels of transmission and prior infection.
Collapse
|
417
|
Jung MK, Jeong SD, Noh JY, Kim DU, Jung S, Song JY, Jeong HW, Park SH, Shin EC. BNT162b2-induced memory T cells respond to the Omicron variant with preserved polyfunctionality. Nat Microbiol 2022; 7:909-917. [PMID: 35577972 DOI: 10.1038/s41564-022-01123-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/07/2022] [Indexed: 12/26/2022]
Abstract
The Omicron variant (B.1.1.529) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) extensively escapes neutralizing antibodies elicited by SARS-CoV-2 infection or vaccination. In the present study, we investigated whether BNT162b2 messenger RNA vaccine-induced memory T cells functionally respond to the Omicron spike protein. Experiments were performed using samples from healthcare workers who were immunized with two or three doses of the BNT162b2 mRNA vaccine and individuals with prior SARS-CoV-2 infection who were immunized with two doses of the BNT162b2 vaccine. Vaccine-induced memory T cells exhibited substantial responses to the Omicron spike protein, with no difference between healthcare workers with two versus three vaccine doses. In individuals with prior infection, two-dose vaccination robustly boosted memory T cells that responded to the Omicron spike protein and the SARS-CoV-2 wild-type (lineage B) spike protein. Importantly, polyfunctionality was preserved in vaccine-induced memory T cells responding to the Omicron spike protein. The present findings indicate that BNT162b2-induced memory T cells substantially respond to the Omicron variant with preserved polyfunctionality.
Collapse
Affiliation(s)
- Min Kyung Jung
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science, Daejeon, Republic of Korea
| | - Seong Dong Jeong
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science, Daejeon, Republic of Korea
| | - Ji Yun Noh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.,Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Dong-Uk Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sungmin Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Hye Won Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea. .,Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea. .,Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea.
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea. .,The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| | - Eui-Cheol Shin
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science, Daejeon, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
418
|
Real-World Effectiveness of the mRNA COVID-19 Vaccines in Japan: A Case–Control Study. Vaccines (Basel) 2022; 10:vaccines10050779. [PMID: 35632535 PMCID: PMC9145554 DOI: 10.3390/vaccines10050779] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/25/2022] Open
Abstract
The real-world effectiveness of the coronavirus disease 2019 (COVID-19) vaccines in Japan remains unclear. This case–control study evaluated the vaccine effectiveness (VE) of two doses of mRNA vaccine, BNT162b2 or mRNA-1273, against the delta (B.1.617.2) variant in the Japanese general population in the period June–September 2021. Individuals in close contact with COVID-19 patients were tested using polymerase chain reaction (PCR). A self-administered questionnaire evaluated vaccination status, demographic data, underlying medical conditions, lifestyle, personal protective health behaviors, and living environment. Two vaccine doses were reported by 11.6% of cases (n = 389) and 35.2% of controls (n = 179). Compared with controls, cases were younger and had a lower proportion who always performed handwashing for ≥20 s, a higher proportion of alcohol consumers, and a lower proportion of individuals living in single-family homes or with commuting family members. After adjusting for these confounding factors and day of PCR testing by multivariate logistic regression analysis, the VE in the period June–July (delta variant proportion 45%) was 92% and 79% in the period August–September (delta variant proportion 89%). The adjusted VE for homestay, hotel-based isolation and quarantine, and hospitalization was 78%, 77%, and 97%, respectively. Despite declining slightly, VE against hospitalization remained robust for ~3 months after the second dose. Vaccination policymaking will require longer-term monitoring of VE against new variants.
Collapse
|
419
|
Mohsin M, Mahmud S. Omicron SARS-CoV-2 variant of concern: A review on its transmissibility, immune evasion, reinfection, and severity. Medicine (Baltimore) 2022; 101:e29165. [PMID: 35583528 PMCID: PMC9276130 DOI: 10.1097/md.0000000000029165] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/07/2022] [Indexed: 01/04/2023] Open
Abstract
Omicron, the new Covid-19 variant, has already become dominant in many countries and is spreading at an unprecedented speed. The objective of this study was to review the existing literature on Omicron's transmissibility, immune evasion, reinfection, and severity.A literature search was performed using "PubMed," "Web of Science," "Scopus," "ScienceDirect," "Google Scholar," "medRxiv," and "bioRxiv." Data were extracted from articles that reported at least one of the following: transmissibility, immune evasion, reinfection, and severity related to Omicron.We found that Omicron spread faster than any other variant. This higher transmissibility can be ascribed to its extraordinary ability to evade the immunity developed by both vaccination and previous infections. However, we found that infections by Omicron were significantly less severe than those caused by Delta and other previous variants. We observed a significantly lower incidence of hospitalization, intensive care unit admission, and mechanical ventilator use in Omicron infections than in Delta or other variants. A substantially shorter median hospital stay and lower fatality rate were also observed in the Omicron infections. Despite Omicron's higher potential to evade immunity, vaccines and booster shots were found to be still significantly effective in protecting against severe Covid-19 infections.Omicrons may be less severe than other variants of concern. However, its immune evasiveness and rapid spread pose an enormous threat to the global healthcare system.
Collapse
Affiliation(s)
- Md Mohsin
- Applied Statistics, Institute of Statistical Research and Training, University of Dhaka, Dhaka, Bangladesh
| | - Sultan Mahmud
- International Centre for Diarrhoeal Disease Research, Bangladesh (ICDDR’B), 68 Shaheed Tajuddin Ahmed Ave, Dhaka, Bangladesh
| |
Collapse
|
420
|
Chaguza C, Coppi A, Earnest R, Ferguson D, Kerantzas N, Warner F, Young HP, Breban MI, Billig K, Koch RT, Pham K, Kalinich CC, Ott IM, Fauver JR, Hahn AM, Tikhonova IR, Castaldi C, De Kumar B, Pettker CM, Warren JL, Weinberger DM, Landry ML, Peaper DR, Schulz W, Vogels CBF, Grubaugh ND. Rapid emergence of SARS-CoV-2 Omicron variant is associated with an infection advantage over Delta in vaccinated persons. MED 2022; 3:325-334.e4. [PMID: 35399324 PMCID: PMC8983481 DOI: 10.1016/j.medj.2022.03.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/25/2021] [Accepted: 03/29/2022] [Indexed: 11/30/2022]
Abstract
Background The SARS-CoV-2 Omicron variant became a global concern due to its rapid spread and displacement of the dominant Delta variant. We hypothesized that part of Omicron's rapid rise was based on its increased ability to cause infections in persons that are vaccinated compared to Delta. Methods We analyzed nasal swab PCR tests for samples collected between December 12 and 16, 2021, in Connecticut when the proportion of Delta and Omicron variants was relatively equal. We used the spike gene target failure (SGTF) to classify probable Delta and Omicron infections. We fitted an exponential curve to the estimated infections to determine the doubling times for each variant. We compared the test positivity rates for each variant by vaccination status, number of doses, and vaccine manufacturer. Generalized linear models were used to assess factors associated with odds of infection with each variant among persons testing positive for SARS-CoV-2. Findings For infections with high virus copies (Ct < 30) among vaccinated persons, we found higher odds that they were infected with Omicron compared to Delta, and that the odds increased with increased number of vaccine doses. Compared to unvaccinated persons, we found significant reduction in Delta positivity rates after two (43.4%-49.1%) and three vaccine doses (81.1%), while we only found a significant reduction in Omicron positivity rates after three doses (62.3%). Conclusion The rapid rise in Omicron infections was likely driven by Omicron's escape from vaccine-induced immunity. Funding This work was supported by the Centers for Disease Control and Prevention (CDC).
Collapse
Affiliation(s)
- Chrispin Chaguza
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Andreas Coppi
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Rebecca Earnest
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - David Ferguson
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Nicholas Kerantzas
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Frederick Warner
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - H Patrick Young
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Mallery I Breban
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Kendall Billig
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Robert Tobias Koch
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Kien Pham
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Chaney C Kalinich
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Isabel M Ott
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Joseph R Fauver
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anne M Hahn
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Irina R Tikhonova
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | | | - Bony De Kumar
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | - Christian M Pettker
- Quality and Safety, Yale New Haven Health, New Haven, CT, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Joshua L Warren
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Marie L Landry
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
- Clinical Virology Laboratory, Yale New Haven Hospital, New Haven, CT, USA
| | - David R Peaper
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Wade Schulz
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Chantal B F Vogels
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
421
|
Chaguza C, Coppi A, Earnest R, Ferguson D, Kerantzas N, Warner F, Young HP, Breban MI, Billig K, Koch RT, Pham K, Kalinich CC, Ott IM, Fauver JR, Hahn AM, Tikhonova IR, Castaldi C, De Kumar B, Pettker CM, Warren JL, Weinberger DM, Landry ML, Peaper DR, Schulz W, Vogels CBF, Grubaugh ND. Rapid emergence of SARS-CoV-2 Omicron variant is associated with an infection advantage over Delta in vaccinated persons. MED 2022; 3:325-334.e4. [PMID: 35399324 DOI: 10.1101/2022.01.22.22269660] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/25/2021] [Accepted: 03/29/2022] [Indexed: 05/22/2023]
Abstract
BACKGROUND The SARS-CoV-2 Omicron variant became a global concern due to its rapid spread and displacement of the dominant Delta variant. We hypothesized that part of Omicron's rapid rise was based on its increased ability to cause infections in persons that are vaccinated compared to Delta. METHODS We analyzed nasal swab PCR tests for samples collected between December 12 and 16, 2021, in Connecticut when the proportion of Delta and Omicron variants was relatively equal. We used the spike gene target failure (SGTF) to classify probable Delta and Omicron infections. We fitted an exponential curve to the estimated infections to determine the doubling times for each variant. We compared the test positivity rates for each variant by vaccination status, number of doses, and vaccine manufacturer. Generalized linear models were used to assess factors associated with odds of infection with each variant among persons testing positive for SARS-CoV-2. FINDINGS For infections with high virus copies (Ct < 30) among vaccinated persons, we found higher odds that they were infected with Omicron compared to Delta, and that the odds increased with increased number of vaccine doses. Compared to unvaccinated persons, we found significant reduction in Delta positivity rates after two (43.4%-49.1%) and three vaccine doses (81.1%), while we only found a significant reduction in Omicron positivity rates after three doses (62.3%). CONCLUSION The rapid rise in Omicron infections was likely driven by Omicron's escape from vaccine-induced immunity. FUNDING This work was supported by the Centers for Disease Control and Prevention (CDC).
Collapse
Affiliation(s)
- Chrispin Chaguza
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Andreas Coppi
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Rebecca Earnest
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - David Ferguson
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Nicholas Kerantzas
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Frederick Warner
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - H Patrick Young
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Mallery I Breban
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Kendall Billig
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Robert Tobias Koch
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Kien Pham
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Chaney C Kalinich
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Isabel M Ott
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Joseph R Fauver
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anne M Hahn
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Irina R Tikhonova
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | | | - Bony De Kumar
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | - Christian M Pettker
- Quality and Safety, Yale New Haven Health, New Haven, CT, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Joshua L Warren
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Marie L Landry
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
- Clinical Virology Laboratory, Yale New Haven Hospital, New Haven, CT, USA
| | - David R Peaper
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Wade Schulz
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Chantal B F Vogels
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
422
|
Yoon SK, Hegmann KT, Thiese MS, Burgess JL, Ellingson K, Lutrick K, Olsho LEW, Edwards LJ, Sokol B, Caban-Martinez AJ, Schaefer-Solle N, Jones JM, Tyner H, Hunt A, Respet K, Gaglani M, Dunnigan K, Rose S, Naleway A, Groom H, Kuntz J, Fowlkes AL, Thompson MG, Yoo YM. Protection with a Third Dose of mRNA Vaccine against SARS-CoV-2 Variants in Frontline Workers. N Engl J Med 2022; 386:1855-1857. [PMID: 35385628 PMCID: PMC9006784 DOI: 10.1056/nejmc2201821] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Angela Hunt
- St. Luke's Regional Health Care System, Duluth, MN
| | | | | | | | | | | | | | | | | | | | - Young M Yoo
- Centers for Disease Control and Prevention, Atlanta, GA
| |
Collapse
|
423
|
Zhang J, Chen N, Zhao D, Zhang J, Hu Z, Tao Z. Clinical Characteristics of COVID-19 Patients Infected by the Omicron Variant of SARS-CoV-2. Front Med (Lausanne) 2022; 9:912367. [PMID: 35615088 PMCID: PMC9125333 DOI: 10.3389/fmed.2022.912367] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 12/19/2022] Open
Abstract
Background Currently, as the omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) surges amid the coronavirus disease 2019 (COVID-19) pandemic, its clinical characteristics with intrinsic severity and the protection from vaccination have been understudied. Methods We reported 169 COVID-19 patients that were infected with the omicron variant of SARS-CoV-2 and hospitalized in Suzhou, China, from February to March 2022, with their demographic information, medical/immunization history, clinical symptom, and hematological profile. At the same time, patients with none/partial (one-dose), full (two-dose) and three-dose vaccination were also compared to assess the vaccine effectiveness. Findings For the omicron COVID-19 patients included in this study, their median age was 33.0 [interquartile range (IQR): 24.0-45.5], 53.3% were male and the median duration from illness onset to hospitalization was 2 days. Hypertension, bronchitis, and diabetes were the leading comorbidities among patients. While the common clinical symptoms included cough, fever, expectoration, and fatigue, etc., asymptomatic patients took up a significant portion (46.7%). For hematological parameters, most values revealed the alleviated pathogenicity induced by the omicron variant infection. No critically ill or deceased patients due to COVID-19 infection were reported in this study. Interpretation Our results supported that the viremic effect of the omicron variant became milder than the previous circulating variants, while full vaccination or booster shot was greatly desired for an effective protection against clinical severity.
Collapse
Affiliation(s)
- Jianguo Zhang
- Department of Emergency Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, China
| | - Nan Chen
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Daguo Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinhui Zhang
- Department of Critical Care Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, China
| | - Zhenkui Hu
- Department of Critical Care Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, China
| | - Zhimin Tao
- Department of Emergency Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, China
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
424
|
Prasad N, Derado G, Nanduri SA, Reses HE, Dubendris H, Wong E, Soe MM, Li Q, Dollard P, Bagchi S, Edwards J, Shang N, Budnitz D, Bell J, Verani JR, Benin A, Link-Gelles R, Jernigan J, Pilishvili T. Effectiveness of a COVID-19 Additional Primary or Booster Vaccine Dose in Preventing SARS-CoV-2 Infection Among Nursing Home Residents During Widespread Circulation of the Omicron Variant - United States, February 14-March 27, 2022. MMWR. MORBIDITY AND MORTALITY WEEKLY REPORT 2022; 71:633-637. [PMID: 35511708 PMCID: PMC9098239 DOI: 10.15585/mmwr.mm7118a4] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nursing home residents have experienced disproportionally high levels of COVID-19-associated morbidity and mortality and were prioritized for early COVID-19 vaccination (1). Following reported declines in vaccine-induced immunity after primary series vaccination, defined as receipt of 2 primary doses of an mRNA vaccine (BNT162b2 [Pfizer-BioNTech] or mRNA-1273 [Moderna]) or 1 primary dose of Ad26.COV2 (Johnson & Johnson [Janssen]) vaccine (2), CDC recommended that all persons aged ≥12 years receive a COVID-19 booster vaccine dose.* Moderately to severely immunocompromised persons, a group that includes many nursing home residents, are also recommended to receive an additional primary COVID-19 vaccine dose.† Data on vaccine effectiveness (VE) of an additional primary or booster dose against infection with SARS-CoV-2 (the virus that causes COVID-19) among nursing home residents are limited, especially against the highly transmissible B.1.1.529 and BA.2 (Omicron) variants. Weekly COVID-19 surveillance and vaccination coverage data among nursing home residents, reported by skilled nursing facilities (SNFs) to CDC's National Healthcare Safety Network (NHSN)§ during February 14-March 27, 2022, when the Omicron variant accounted for >99% of sequenced isolates, were analyzed to estimate relative VE against infection for any COVID-19 additional primary or booster dose compared with primary series vaccination. After adjusting for calendar week and variability across SNFs, relative VE of a COVID-19 additional primary or booster dose was 46.9% (95% CI = 44.8%-48.9%). These findings indicate that among nursing home residents, COVID-19 additional primary or booster doses provide greater protection against Omicron variant infection than does primary series vaccination alone. All immunocompromised nursing home residents should receive an additional primary dose, and all nursing home residents should receive a booster dose, when eligible, to protect against COVID-19. Efforts to keep nursing home residents up to date with vaccination should be implemented in conjunction with other COVID-19 prevention strategies, including testing and vaccination of nursing home staff members and visitors.
Collapse
|
425
|
Ng TI, Correia I, Seagal J, DeGoey DA, Schrimpf MR, Hardee DJ, Noey EL, Kati WM. Antiviral Drug Discovery for the Treatment of COVID-19 Infections. Viruses 2022; 14:961. [PMID: 35632703 PMCID: PMC9143071 DOI: 10.3390/v14050961] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a recently emerged human coronavirus. COVID-19 vaccines have proven to be successful in protecting the vaccinated from infection, reducing the severity of disease, and deterring the transmission of infection. However, COVID-19 vaccination faces many challenges, such as the decline in vaccine-induced immunity over time, and the decrease in potency against some SARS-CoV-2 variants including the recently emerged Omicron variant, resulting in breakthrough infections. The challenges that COVID-19 vaccination is facing highlight the importance of the discovery of antivirals to serve as another means to tackle the pandemic. To date, neutralizing antibodies that block viral entry by targeting the viral spike protein make up the largest class of antivirals that has received US FDA emergency use authorization (EUA) for COVID-19 treatment. In addition to the spike protein, other key targets for the discovery of direct-acting antivirals include viral enzymes that are essential for SARS-CoV-2 replication, such as RNA-dependent RNA polymerase and proteases, as judged by US FDA approval for remdesivir, and EUA for Paxlovid (nirmatrelvir + ritonavir) for treating COVID-19 infections. This review presents an overview of the current status and future direction of antiviral drug discovery for treating SARS-CoV-2 infections, covering important antiviral targets such as the viral spike protein, non-structural protein (nsp) 3 papain-like protease, nsp5 main protease, and the nsp12/nsp7/nsp8 RNA-dependent RNA polymerase complex.
Collapse
Affiliation(s)
- Teresa I. Ng
- Virology Drug Discovery, AbbVie Inc., North Chicago, IL 60064, USA;
| | - Ivan Correia
- Department of Cell and Protein Sciences, Drug Discovery Science and Technology, AbbVie Inc., Worcester, MA 01605, USA;
| | - Jane Seagal
- Department of Biologics Discovery, Drug Discovery Science and Technology, AbbVie Inc., Worcester, MA 01605, USA;
| | - David A. DeGoey
- Department of Centralized Medicinal Chemistry, Drug Discovery Science and Technology, AbbVie Inc., North Chicago, IL 60064, USA; (D.A.D.); (M.R.S.); (D.J.H.)
| | - Michael R. Schrimpf
- Department of Centralized Medicinal Chemistry, Drug Discovery Science and Technology, AbbVie Inc., North Chicago, IL 60064, USA; (D.A.D.); (M.R.S.); (D.J.H.)
| | - David J. Hardee
- Department of Centralized Medicinal Chemistry, Drug Discovery Science and Technology, AbbVie Inc., North Chicago, IL 60064, USA; (D.A.D.); (M.R.S.); (D.J.H.)
| | - Elizabeth L. Noey
- Department of Structural Biology, Drug Discovery Science and Technology, AbbVie Inc., North Chicago, IL 60064, USA;
| | - Warren M. Kati
- Virology Drug Discovery, AbbVie Inc., North Chicago, IL 60064, USA;
| |
Collapse
|
426
|
Bunch CM, Moore EE, Moore HB, Neal MD, Thomas AV, Zackariya N, Zhao J, Zackariya S, Brenner TJ, Berquist M, Buckner H, Wiarda G, Fulkerson D, Huff W, Kwaan HC, Lankowicz G, Laubscher GJ, Lourens PJ, Pretorius E, Kotze MJ, Moolla MS, Sithole S, Maponga TG, Kell DB, Fox MD, Gillespie L, Khan RZ, Mamczak CN, March R, Macias R, Bull BS, Walsh MM. Immuno-Thrombotic Complications of COVID-19: Implications for Timing of Surgery and Anticoagulation. Front Surg 2022; 9:889999. [PMID: 35599794 PMCID: PMC9119324 DOI: 10.3389/fsurg.2022.889999] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/05/2022] [Indexed: 12/12/2022] Open
Abstract
Early in the coronavirus disease 2019 (COVID-19) pandemic, global governing bodies prioritized transmissibility-based precautions and hospital capacity as the foundation for delay of elective procedures. As elective surgical volumes increased, convalescent COVID-19 patients faced increased postoperative morbidity and mortality and clinicians had limited evidence for stratifying individual risk in this population. Clear evidence now demonstrates that those recovering from COVID-19 have increased postoperative morbidity and mortality. These data-in conjunction with the recent American Society of Anesthesiologists guidelines-offer the evidence necessary to expand the early pandemic guidelines and guide the surgeon's preoperative risk assessment. Here, we argue elective surgeries should still be delayed on a personalized basis to maximize postoperative outcomes. We outline a framework for stratifying the individual COVID-19 patient's fitness for surgery based on the symptoms and severity of acute or convalescent COVID-19 illness, coagulopathy assessment, and acuity of the surgical procedure. Although the most common manifestation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is COVID-19 pneumonitis, every system in the body is potentially afflicted by an endotheliitis. This endothelial derangement most often manifests as a hypercoagulable state on admission with associated occult and symptomatic venous and arterial thromboembolisms. The delicate balance between hyper and hypocoagulable states is defined by the local immune-thrombotic crosstalk that results commonly in a hemostatic derangement known as fibrinolytic shutdown. In tandem, the hemostatic derangements that occur during acute COVID-19 infection affect not only the timing of surgical procedures, but also the incidence of postoperative hemostatic complications related to COVID-19-associated coagulopathy (CAC). Traditional methods of thromboprophylaxis and treatment of thromboses after surgery require a tailored approach guided by an understanding of the pathophysiologic underpinnings of the COVID-19 patient. Likewise, a prolonged period of risk for developing hemostatic complications following hospitalization due to COVID-19 has resulted in guidelines from differing societies that recommend varying periods of delay following SARS-CoV-2 infection. In conclusion, we propose the perioperative, personalized assessment of COVID-19 patients' CAC using viscoelastic hemostatic assays and fluorescent microclot analysis.
Collapse
Affiliation(s)
- Connor M. Bunch
- Department of Emergency Medicine, Henry Ford Hospital, Detroit, MI, United States
| | - Ernest E. Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, Denver, CO, United States
| | - Hunter B. Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, Denver, CO, United States
| | - Matthew D. Neal
- Pittsburgh Trauma Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Anthony V. Thomas
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
| | - Nuha Zackariya
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
| | - Jonathan Zhao
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Sufyan Zackariya
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Toby J. Brenner
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Margaret Berquist
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Hallie Buckner
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Grant Wiarda
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Daniel Fulkerson
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
- Department of Neurosurgery, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Wei Huff
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
- Department of Neurosurgery, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Hau C. Kwaan
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Genevieve Lankowicz
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | | | | | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Maritha J. Kotze
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University and National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa
| | - Muhammad S. Moolla
- Division of General Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Sithembiso Sithole
- Division of General Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Tongai G. Maponga
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Douglas B. Kell
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mark D. Fox
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
| | - Laura Gillespie
- Department of Quality Assurance and Performance Improvement, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Rashid Z. Khan
- Department of Hematology, Michiana Hematology Oncology, Mishawaka, IN, United States
| | - Christiaan N. Mamczak
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
- Department of Orthopaedic Trauma, Memorial Hospital South Bend, South Bend, IN, United States
| | - Robert March
- Department of Cardiothoracic Surgery, St. Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Rachel Macias
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
- Department of Plastic and Reconstructive Surgery, St. Joseph Regional Medical Center, Mishawaka, IN, United States
| | - Brian S. Bull
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Mark M. Walsh
- Indiana University School of Medicine South Bend Campus, Notre Dame, IN, United States
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, United States
| |
Collapse
|
427
|
Bestvina CM, Whisenant JG, Torri V, Cortellini A, Wakelee H, Peters S, Roca E, De Toma A, Hirsch FR, Mamdani H, Halmos B, Arrieta O, Metivier AC, Fidler MJ, Rogado J, Presley CJ, Mascaux C, Genova C, Blaquier JB, Addeo A, Finocchiaro G, Khan H, Mazieres J, Morgillo F, Bar J, Aujayeb A, Mountzios G, Scotti V, Grosso F, Geraedts E, Zhumagaliyeva AN, Horn L, Garassino MC, Baena J. COVID-19 Outcomes, Patient Vaccination Status, and Cancer-Related Delays during the Omicron Wave: A Brief Report from the TERAVOLT Analysis. JTO Clin Res Rep 2022; 3:100335. [PMID: 35619644 PMCID: PMC9119707 DOI: 10.1016/j.jtocrr.2022.100335] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/01/2022] [Indexed: 11/08/2022] Open
Abstract
Introduction The Thoracic Centers International coronavirus disease 2019 (COVID-19) Collaboration (TERAVOLT) registry found approximately 30% mortality in patients with thoracic malignancies during the initial COVID-19 surges. Data from South Africa suggested a decrease in severity and mortality with the Omicron wave. Our objective was to assess mortality of patients with thoracic malignancies with the Omicron-predominant wave and evaluate efficacy of vaccination. Methods A prospective, multicenter observational study was conducted. A total of 28 institutions contributed data from January 14, 2022, to February 4, 2022. Inclusion criteria were any thoracic cancer and a COVID-19 diagnosis on or after November 1, 2021. End points included mortality, hospitalization, symptomatic COVID-19 infection, asymptomatic COVID-19 infection, and delay in cancer therapy. Analysis was done through contingency tables and a multivariable logistic model. Results We enrolled a total of 346 patients. Median age was 65 years, 52.3% were female, 74.2% were current or former smokers, 86% had NSCLC, 72% had stage IV at time of COVID-19 diagnosis, and 66% were receiving cancer therapy. Variant was unknown for 70%; for those known, Omicron represented 82%. Overall mortality was 3.2%. Using multivariate analysis, COVID-19 vaccination with booster compared with no vaccination had a protective effect on hospitalization or death (OR = 0.30, confidence interval: 0.15–0.57, p = 0.0003), whereas vaccination without booster did not (OR = 0.64, confidence interval: 0.33–1.24, p = 0.1864). Cancer care was delayed in 56.4% of the patients. Conclusions TERAVOLT found reduced patient mortality with the most recent COVID-19 surge. COVID-19 vaccination with booster improved outcomes of hospitalization or death. Delays in cancer therapy remain an issue, which has the potential to worsen cancer-related mortality.
Collapse
|
428
|
Affiliation(s)
- Paul A Offit
- From the Children's Hospital of Philadelphia, Philadelphia
| |
Collapse
|
429
|
SARS-CoV-2 Omicron variant: recent progress and future perspectives. Signal Transduct Target Ther 2022; 7:141. [PMID: 35484110 PMCID: PMC9047469 DOI: 10.1038/s41392-022-00997-x] [Citation(s) in RCA: 296] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/27/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Since the outbreak of the coronavirus disease 2019 (COVID-19) pandemic, there have been a few variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), one of which is the Omicron variant (B.1.1.529). The Omicron variant is the most mutated SARS-CoV-2 variant, and its high transmissibility and immune evasion ability have raised global concerns. Owing to its enhanced transmissibility, Omicron has rapidly replaced Delta as the dominant variant in several regions. However, recent studies have shown that the Omicron variant exhibits reduced pathogenicity due to altered cell tropism. In addition, Omicron exhibits significant resistance to the neutralizing activity of vaccines, convalescent serum, and most antibody therapies. In the present review, recent advances in the molecular and clinical characteristics of the infectivity, pathogenicity, and immune evasion of Omicron variant was summarized, and potential therapeutic applications in response to Omicron infection were discussed. Furthermore, we highlighted potential response to future waves and strategies to end the pandemic.
Collapse
|
430
|
Gagne M, Moliva JI, Foulds KE, Andrew SF, Flynn BJ, Werner AP, Wagner DA, Teng IT, Lin BC, Moore C, Jean-Baptiste N, Carroll R, Foster SL, Patel M, Ellis M, Edara VV, Maldonado NV, Minai M, McCormick L, Honeycutt CC, Nagata BM, Bock KW, Dulan CNM, Cordon J, Flebbe DR, Todd JPM, McCarthy E, Pessaint L, Van Ry A, Narvaez B, Valentin D, Cook A, Dodson A, Steingrebe K, Nurmukhambetova ST, Godbole S, Henry AR, Laboune F, Roberts-Torres J, Lorang CG, Amin S, Trost J, Naisan M, Basappa M, Willis J, Wang L, Shi W, Doria-Rose NA, Zhang Y, Yang ES, Leung K, O'Dell S, Schmidt SD, Olia AS, Liu C, Harris DR, Chuang GY, Stewart-Jones G, Renzi I, Lai YT, Malinowski A, Wu K, Mascola JR, Carfi A, Kwong PD, Edwards DK, Lewis MG, Andersen H, Corbett KS, Nason MC, McDermott AB, Suthar MS, Moore IN, Roederer M, Sullivan NJ, Douek DC, Seder RA. mRNA-1273 or mRNA-Omicron boost in vaccinated macaques elicits similar B cell expansion, neutralizing responses, and protection from Omicron. Cell 2022; 185:1556-1571.e18. [PMID: 35447072 PMCID: PMC8947944 DOI: 10.1016/j.cell.2022.03.038] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 01/06/2023]
Abstract
SARS-CoV-2 Omicron is highly transmissible and has substantial resistance to neutralization following immunization with ancestral spike-matched vaccines. It is unclear whether boosting with Omicron-matched vaccines would enhance protection. Here, nonhuman primates that received mRNA-1273 at weeks 0 and 4 were boosted at week 41 with mRNA-1273 or mRNA-Omicron. Neutralizing titers against D614G were 4,760 and 270 reciprocal ID50 at week 6 (peak) and week 41 (preboost), respectively, and 320 and 110 for Omicron. 2 weeks after the boost, titers against D614G and Omicron increased to 5,360 and 2,980 for mRNA-1273 boost and 2,670 and 1,930 for mRNA-Omicron, respectively. Similar increases against BA.2 were observed. Following either boost, 70%-80% of spike-specific B cells were cross-reactive against WA1 and Omicron. Equivalent control of virus replication in lower airways was observed following Omicron challenge 1 month after either boost. These data show that mRNA-1273 and mRNA-Omicron elicit comparable immunity and protection shortly after the boost.
Collapse
Affiliation(s)
- Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shayne F Andrew
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne P Werner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danielle A Wagner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher Moore
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nazaire Jean-Baptiste
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robin Carroll
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephanie L Foster
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mit Patel
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Madison Ellis
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Venkata-Viswanadh Edara
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nahara Vargas Maldonado
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Lauren McCormick
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Caitlyn N M Dulan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jamilet Cordon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dillon R Flebbe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John-Paul M Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth McCarthy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | - Saule T Nurmukhambetova
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sucheta Godbole
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Farida Laboune
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jesmine Roberts-Torres
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cynthia G Lorang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shivani Amin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jessica Trost
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mursal Naisan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacquelyn Willis
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Shi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole A Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kwanyee Leung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sijy O'Dell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen D Schmidt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cuiping Liu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Darcy R Harris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | - Kai Wu
- Moderna Inc., Cambridge, MA 02139, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | - Kizzmekia S Corbett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ian N Moore
- Division of Pathology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
431
|
Padoan A, Cosma C, Della Rocca F, Barbaro F, Santarossa C, Dall'Olmo L, Galla L, Cattelan A, Cianci V, Basso D, Plebani M. A cohort analysis of SARS-CoV-2 anti-spike protein receptor binding domain (RBD) IgG levels and neutralizing antibodies in fully vaccinated healthcare workers. Clin Chem Lab Med 2022; 60:1110-1115. [PMID: 35473824 DOI: 10.1515/cclm-2022-0322] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The waning of humoral immunity after COVID-19 vaccine booster (third dose) has not yet been fully evaluated. This study updates data on anti-SARS-CoV-2 spike protein receptor binding domain (S-RBD) binding antibodies (bAb) and neutralizing antibodies (NAb) levels in individuals with homologous vaccination 3-4 months after receiving the booster dose. METHODS Fifty-five healthcare workers (HCW) from Padova University-Hospital were asked to collect serum samples for determining antibodies (Ab) at 12 (t12) and 28 (t28) days, at 6 months (t6m) after their first Comirnaty/BNT162b2 inoculation, and 3-4 months after receiving the 3rd homologous booster dose. HCW were monitored weekly for SARS-CoV-2 infection. Ab titers were measured by two chemiluminescent immunoassays, one targeting the S-RBD immunoglobulin G (IgG), and one surrogate viral neutralization test (sVNT), measuring NAb. RESULTS Twenty of the HCW had natural COVID-19 infection (COVID+) at different times, before either the first or the second vaccination. Median S-RBD IgG and NAb levels and their interquartile ranges 3-4 months after the 3rd dose were 1,076 (529-3,409) kBAU/L and 15.8 (11.3-38.3) mg/L, respectively, for COVID-, and 1,373 (700-1,373) kBAU/L and 21 (12.8-53.9) mg/L, respectively, for COVID+. At multivariate regression analyses, with age and gender included as covariates, S-RBD IgG bAb and sVNT NAb levels were closely associated with the time interval between serological determination and the 3rd vaccine dose (log10 _coeff=-0.013, p=0.012 and log10 _coeff=-0.010, p=0.025) for COVID+, whereas no such association was found in COVID- individuals. CONCLUSIONS The third booster dose increases anti-SARS-CoV-2 Ab levels, elevated levels persisting for up to 3-4 months. Waning of Ab levels appears to be less pronounced for COVID+ individuals.
Collapse
Affiliation(s)
- Andrea Padoan
- Department of Medicine - DIMED, University of Padova, Padova, Italy.,Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | | | - Francesco Barbaro
- Unit of Infectious and Tropical Diseases, University-Hospital of Padova, Padova, Italy
| | | | - Luigi Dall'Olmo
- Department of Surgical Oncological and Gastroenterological Sciences - DISCOG, University of Padova, Padova, Italy.,Veneto Institute of Oncology, IOV-IRCCS, Padova, Italy
| | - Luisa Galla
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Annamaria Cattelan
- Unit of Infectious and Tropical Diseases, University-Hospital of Padova, Padova, Italy
| | - Vito Cianci
- Unit of Infectious and Tropical Diseases, University-Hospital of Padova, Padova, Italy
| | - Daniela Basso
- Department of Medicine - DIMED, University of Padova, Padova, Italy.,Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Mario Plebani
- Department of Medicine - DIMED, University of Padova, Padova, Italy.,Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| |
Collapse
|
432
|
Sharma A, Oda G, Holodniy M. Effectiveness of Messenger RNA-based Vaccines During the Emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 Omicron Variant. Clin Infect Dis 2022; 75:2186-2192. [PMID: 35475889 PMCID: PMC9129111 DOI: 10.1093/cid/ciac325] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND We evaluated the effectiveness of mRNA-based vaccines following emergence of SARS-CoV-2 Omicron variant. METHODS Recipients of a third dose of BNT162b2 or mRNA-1273 ≥180 days after the primary series were matched to primary-series recipients and unvaccinated persons. Participants were followed from 1 December 2021 to 12 March 2022. Outcomes were documented SARS-CoV-2 infection, COVID-19 hospitalization, and COVID-19 death. Effectiveness was calculated from 100-day risks estimated with the Kaplan-Meier estimator. RESULTS BNT162b2 and mRNA-1273 groups included 221 267 and 187 507 third-dose recipients, respectively, matched to equal numbers of primary-series recipients and unvaccinated persons. Compared with no vaccination, effectiveness of a third dose of BNT162b2 was 47.8% (95% confidence interval [CI], 45.2-50.3), 81.8% (95% CI, 79.2-84.2), and 89.6% (95% CI, 85.0-93.6) against infection, hospitalization, and death, respectively. Effectiveness of a third dose of BNT162b2 compared with the primary series was 30.1% (95% CI, 26.2-33.7), 61.4% (95% CI, 55.0-67.1), and 78.8% (95% CI, 67.9-87.5) against infection, hospitalization, and death, respectively. Effectiveness of a third dose of mRNA-1273 compared with no vaccination was 61.9% (95% CI, 59.4-64.4), 87.9% (95% CI, 85.3-90.2), and 91.4% (95% CI, 86.4-95.6) against infection, hospitalization, and death, respectively. Effectiveness of a third dose of mRNA-1273 compared with the primary series was 37.1% (95% CI, 32.2-41.7), 63.5% (95% CI, 53.7-71.6), and 75.0% (95% CI, 55.4-88.0) against infection, hospitalization, and death, respectively. CONCLUSIONS BNT162b2 and mRNA-1273 were effective against COVID-19 following emergence of Omicron variant. A third dose provided additional protection over the primary series.
Collapse
Affiliation(s)
- Aditya Sharma
- US Department of Veterans Affairs, Public Health Program, Washington, DC and Palo Alto, CA, USA,Corresponding Author Aditya Sharma MD US Department of Veterans Affairs Public Health Program Palo Alto, CA 94304 USA
| | - Gina Oda
- US Department of Veterans Affairs, Public Health Program, Washington, DC and Palo Alto, CA, USA
| | - Mark Holodniy
- US Department of Veterans Affairs, Public Health Program, Washington, DC and Palo Alto, CA, USA,Stanford University, Stanford, CA, USA,Alternate Corresponding Author Mark Holodniy MD US Department of Veterans Affairs Public Health Program Palo Alto, CA 94304 USA
| |
Collapse
|
433
|
Soraci L, Lattanzio F, Soraci G, Gambuzza ME, Pulvirenti C, Cozza A, Corsonello A, Luciani F, Rezza G. COVID-19 Vaccines: Current and Future Perspectives. Vaccines (Basel) 2022; 10:608. [PMID: 35455357 PMCID: PMC9025326 DOI: 10.3390/vaccines10040608] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022] Open
Abstract
Currently available vaccines against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) are highly effective but not able to keep the coronavirus disease 2019 (COVID-19) pandemic completely under control. Alternative R&D strategies are required to induce a long-lasting immunological response and to reduce adverse events as well as to favor rapid development and large-scale production. Several technological platforms have been used to develop COVID-19 vaccines, including inactivated viruses, recombinant proteins, DNA- and RNA-based vaccines, virus-vectored vaccines, and virus-like particles. In general, mRNA vaccines, protein-based vaccines, and vectored vaccines have shown a high level of protection against COVID-19. However, the mutation-prone nature of the spike (S) protein affects long-lasting vaccine protection and its effectiveness, and vaccinated people can become infected with new variants, also showing high virus levels. In addition, adverse effects may occur, some of them related to the interaction of the S protein with the angiotensin-converting enzyme 2 (ACE-2). Thus, there are some concerns that need to be addressed and challenges regarding logistic problems, such as strict storage at low temperatures for some vaccines. In this review, we discuss the limits of vaccines developed against COVID-19 and possible innovative approaches.
Collapse
Affiliation(s)
- Luca Soraci
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), 87100 Cosenza, Italy; (L.S.); (A.C.)
| | - Fabrizia Lattanzio
- Scientific Direction, Italian National Research Center on Aging (IRCCS INRCA), 60121 Ancona, Italy;
| | - Giulia Soraci
- Department of Obstetrics and Gynecology, University of Ferrara, 44121 Ferrara, Italy;
| | - Maria Elsa Gambuzza
- Territorial Office of Messina, Italian Ministry of Health, 98122 Messina, Italy
| | | | - Annalisa Cozza
- Laboratory of Pharmacoepidemiology and Biostatistics, Italian National Research Center on Aging (IRCCS INRCA), 87100 Cosenza, Italy;
| | - Andrea Corsonello
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), 87100 Cosenza, Italy; (L.S.); (A.C.)
- Laboratory of Pharmacoepidemiology and Biostatistics, Italian National Research Center on Aging (IRCCS INRCA), 87100 Cosenza, Italy;
| | - Filippo Luciani
- Infectious Diseases Unit of Annunziata Hospital, 87100 Cosenza, Italy;
| | - Giovanni Rezza
- Health Prevention Directorate, Italian Ministry of Health, 00144 Rome, Italy;
| |
Collapse
|
434
|
Rzymski P, Pazgan-Simon M, Kamerys J, Moniuszko-Malinowska A, Sikorska K, Wernik J, Zarębska-Michaluk D, Supronowicz Ł, Sobala-Szczygieł B, Skrzat-Klapaczyńska A, Simon K, Piekarska A, Czupryna P, Pawłowska M, Brzdęk M, Jaroszewicz J, Kowalska J, Renke M, Flisiak R. Severe Breakthrough COVID-19 Cases during Six Months of Delta Variant (B.1.617.2) Domination in Poland. Vaccines (Basel) 2022; 10:557. [PMID: 35455306 PMCID: PMC9025315 DOI: 10.3390/vaccines10040557] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 02/04/2023] Open
Abstract
The emergence of a highly transmissible and a more pathogenic B.1.617.2 (delta) variant of SARS-CoV-2 has brought concern over COVID-19 vaccine efficacy and the increased risk of severe breakthrough infections. The objective of this study was to assess the frequency and the clinical characteristics of severe breakthrough COVID-19 cases recorded in 10 Polish healthcare units between 1 June and 31 December 2021, a period during which a rapid surge in the share of B.1.617.2 infections was seen, while a significant number of populations were already fully vaccinated. Overall, 723 individuals who completed the initial vaccination regime (fully vaccinated group) and an additional 18 who received a booster dose were identified—together, they represented 20.8% of all the COVID-19 patients hospitalized during the same period in the same healthcare institutions (0.5% in the case of a group that received a booster dose). Although laboratory and clinical parameters did not differ between both groups, patients who received a booster tended to have lower CRP, IL-6, PCT, and d-dimer levels and they required oxygen therapy less frequently. The most common early COVID-19 symptoms in the studied group were fatigue, cough, fever (>38 °C), and dyspnea. Individuals with no detectable anti-spike IgG antibodies constituted 13%; the odds of being a humoral non-responder to the vaccine were increased in patients aged >70 years. Fully vaccinated patients hospitalized after more than 180 days from the last vaccine dose were significantly older and they were predominantly represented by individuals over 70 years and with comorbidities, particularly cardiovascular disease. Contrary to mRNA vaccines, most patients vaccinated with adenoviral vector vaccines were infected within six months. A total of 102 fatal cases (14% of all deaths among vaccinated individuals; 0.7% in the case of a group that received a booster dose) were recorded, representing 17.6% of all the COVID-19 fatalities recorded in June−December 2021 in the considered healthcare units. The odds of death were significantly increased in men, individuals aged >70 years, patients with comorbidities, and those identified as humoral non-responders to vaccination; in fully vaccinated patients the odds were also increased when the second vaccine dose was given >180 days before the first COVID-19 symptoms. The mortality rate in immunocompromised subjects was 19%. The results indicate that compared to vaccinated individuals, severe COVID-19 and deaths in the unvaccinated group were significantly more prevalent during the B.1.617.2-dominated wave in Poland; and, it highlight the protective role of a booster dose, particularly for more vulnerable individuals.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), 60-806 Poznań, Poland
| | - Monika Pazgan-Simon
- 1st Infectious Diseases Ward, Gromkowski Regional Specialist Hospital, 50-149 Wroclaw, Poland;
- Department of Infectious Diseases and Hepatology, Wrocław Medical University, 51-149 Wrocław, Poland;
| | - Juliusz Kamerys
- Department of Infectious Diseases and Hepatology, Medical University of Łódź, 90-549 Łódź, Poland; (J.K.); (A.P.)
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Białystok, 15-089 Białystok, Poland; (A.M.-M.); (P.C.)
| | - Katarzyna Sikorska
- Department of Tropical Medicine and Epidemiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Joanna Wernik
- Department of Infectious Diseases and Hepatology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Toruń, Poland; (J.W.); (M.P.)
| | - Dorota Zarębska-Michaluk
- Department of Infectious Diseases, Jan Kochanowski University, 25-369 Kielce, Poland; (D.Z.-M.); (M.B.)
| | - Łukasz Supronowicz
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, 15-089 Białystok, Poland; (Ł.S.); (R.F.)
| | - Barbara Sobala-Szczygieł
- Department of Infectious Diseases and Hepatology, Medical University of Silesia, 40-055 Katowice, Poland; (B.S.-S.); (J.J.)
| | - Agata Skrzat-Klapaczyńska
- Department of Adults’ Infectious Diseases, Hospital for Infectious Diseases, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.-K.); (J.K.)
| | - Krzysztof Simon
- Department of Infectious Diseases and Hepatology, Wrocław Medical University, 51-149 Wrocław, Poland;
| | - Anna Piekarska
- Department of Infectious Diseases and Hepatology, Medical University of Łódź, 90-549 Łódź, Poland; (J.K.); (A.P.)
| | - Piotr Czupryna
- Department of Infectious Diseases and Neuroinfections, Medical University of Białystok, 15-089 Białystok, Poland; (A.M.-M.); (P.C.)
| | - Małgorzata Pawłowska
- Department of Infectious Diseases and Hepatology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Toruń, Poland; (J.W.); (M.P.)
| | - Michał Brzdęk
- Department of Infectious Diseases, Jan Kochanowski University, 25-369 Kielce, Poland; (D.Z.-M.); (M.B.)
| | - Jerzy Jaroszewicz
- Department of Infectious Diseases and Hepatology, Medical University of Silesia, 40-055 Katowice, Poland; (B.S.-S.); (J.J.)
| | - Justyna Kowalska
- Department of Adults’ Infectious Diseases, Hospital for Infectious Diseases, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.-K.); (J.K.)
| | - Marcin Renke
- Division of Occupational, Metabolic and Internal Diseases, Institute of Maritime and Tropical Medicine, Faculty of Health Sciences, Medical University of Gdansk, 81-519 Gdynia, Poland;
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, 15-089 Białystok, Poland; (Ł.S.); (R.F.)
| |
Collapse
|
435
|
Hoffman TW, Meek B, Rijkers GT, van Kessel DA. Serologic response to a third dose of an mRNA-based SARS-CoV-2 vaccine in lung transplant recipients. Transpl Immunol 2022; 72:101599. [PMID: 35390480 PMCID: PMC8978449 DOI: 10.1016/j.trim.2022.101599] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/12/2022]
Abstract
Lung transplant recipients have an increased risk for severe coronavirus disease 2019 (COVID-19) due to infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A third dose of a SARS-CoV-2 vaccine has been recommended for all solid organ transplant recipients, but data from lung transplant recipients specifically are scarce. In this study, the serologic response to a third dose of an mRNA-based SARS-CoV-2 vaccine was measured in 78 lung transplant recipients. Sixty-two percent (n = 48) had a serological response to vaccination, which was significantly higher than after the second vaccine dose (27 patients (35%); p = 0.0013). A positive serologic response was associated with having had COVID-19 (p = 0.01), and higher serum IgG level and complement mannose binding lectin pathway activity prior to vaccination (p = 0.04 and p = 0.03, respectively). Serologic response was not associated with the dose of mycophenolate mofetil or prednisone or other immune status parameters. Eleven patients (14%) developed COVID-19 after the second or third vaccine dose, but this did not associate with serologic response after the second vaccine dose (9% in patients who developed COVID-19 versus 39% in patients who did not develop COVID-19 (p = 0.09)), or with serologic response above cut-off values associated with clinical protection in previous studies. In conclusion, the response to mRNA-based SARS-CoV-2 vaccines in lung transplant recipients improves significantly after a third vaccine dose. Factors associated with a positive serologic response are having had COVID-19 prior to vaccination, and serum IgG and complement mannose binding lectin pathway activity prior to vaccination. Serologic response did not associate with clinical protection against COVID-19 in this study.
Collapse
Affiliation(s)
- T W Hoffman
- Department of Pulmonology, St. Antonius Hospital, Nieuwegein/Utrecht, the Netherlands.
| | - B Meek
- Department of Medical Microbiology and Immunology, St. Antonius Hospital, Nieuwegein/Utrecht, the Netherlands
| | - G T Rijkers
- Science Department, University College Roosevelt, Middelburg, the Netherlands
| | - D A van Kessel
- Department of Pulmonology, St. Antonius Hospital, Nieuwegein/Utrecht, the Netherlands; Department of Pulmonology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
436
|
High vaccination coverage slows down genetic diversity of SARS-CoV-2. J Infect 2022; 85:90-122. [PMID: 35398408 PMCID: PMC8988574 DOI: 10.1016/j.jinf.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/02/2022] [Indexed: 11/20/2022]
|
437
|
Gener AR, Kottilil S. Toward demystifying HIV as a risk factor for coronavirus disease 2019 complications. AIDS 2022; 36:749-750. [PMID: 35323159 DOI: 10.1097/qad.0000000000003200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | - Shyam Kottilil
- Division of Infectious Diseases, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
438
|
Seybold U. [Vaccination against COVID-19: general recommendations and special populations]. MMW Fortschr Med 2022; 164:42-51. [PMID: 35449280 PMCID: PMC9023104 DOI: 10.1007/s15006-022-0924-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ulrich Seybold
- Medizinische Klinik u. Poliklinik IV / Sektion Klinische Infektiologie, Klinikum Innenstadt der LMU München, Pettenkoferstraße 8 a, 80336, München, Germany.
| |
Collapse
|
439
|
Hertanto DM, Sutanto H, Lusida MI, Kuntaman K, Santoso D. The genomic and clinical features of the COVID-19 Omicron variant: a narrative review. F1000Res 2022. [DOI: 10.12688/f1000research.110647.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a major cause of morbidity and mortality worldwide. Since late November 2021, the Omicron variant has emerged as the primary cause of COVID-19 and caused a huge increase in the reported incidence around the world. To date, 32-34 spike mutations have been reported to be present in the Omicron variant, 15 of which were located in the receptor-binding domain that interacts with the cell surface of the host cells, while the rest were located in the N-terminal domain and around the furin cleavage site. Recent studies have suggested that those mutations could have a major role in the transmissibility and pathogenicity of the Omicron variant. Additionally, some mutations might contribute to the change of viral tropism of this novel variant. Here, we aim to discuss the recent reports on the transmissibility and severity of the Omicron variant from both the genetic and clinical perspectives. Afterward, we also take the chance to deliver our personal view on the topic.
Collapse
|
440
|
Burns MD, Boribong BP, Bartsch YC, Loiselle M, St. Denis KJ, Sheehan ML, Chen JW, Davis JP, Lima R, Edlow AG, Fasano A, Balazs AB, Alter G, Yonker LM. Durability and Cross-Reactivity of SARS-CoV-2 mRNA Vaccine in Adolescent Children. Vaccines (Basel) 2022; 10:492. [PMID: 35455241 PMCID: PMC9032590 DOI: 10.3390/vaccines10040492] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 01/11/2023] Open
Abstract
Emergent SARS-CoV-2 variants and waning humoral immunity in vaccinated individuals have resulted in increased infections and hospitalizations. Children are not spared from infection nor complications of COVID-19, and the recent recommendation for boosters in individuals ages 12 years or older calls for broader understanding of the adolescent immune profile after mRNA vaccination. We tested the durability and cross-reactivity of anti-SARS-CoV-2 serologic responses over a six-month time course in vaccinated adolescents against the SARS-CoV-2 D614G ("wild type") and Omicron antigens. Serum from 77 adolescents showed that anti-Spike antibodies wane significantly over six months. After completion of a two-vaccine series, cross-reactivity against Omicron-specific receptor-binding domain (RBD) was seen. Functional humoral activation against wild type and Omicron SARS-CoV-2 also declines over time in vaccinated adolescent children. Evidence of waning mRNA-induced vaccine immunity underscores vulnerabilities in long-term pediatric protection against SARS-CoV-2 infection, while cross-reactivity highlights the additional benefits of vaccination. Characterization of adolescent immune signatures post-vaccination will inform guidance on vaccine platforms and timelines, and ultimately optimize immunoprotection of children.
Collapse
Affiliation(s)
- Madeleine D. Burns
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA; (M.D.B.); (B.P.B.); (M.L.); (J.P.D.); (R.L.); (A.F.)
| | - Brittany P. Boribong
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA; (M.D.B.); (B.P.B.); (M.L.); (J.P.D.); (R.L.); (A.F.)
| | - Yannic C. Bartsch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; (Y.C.B.); (K.J.S.D.); (M.L.S.); (J.W.C.); (A.B.B.); (G.A.)
| | - Maggie Loiselle
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA; (M.D.B.); (B.P.B.); (M.L.); (J.P.D.); (R.L.); (A.F.)
| | - Kerri J. St. Denis
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; (Y.C.B.); (K.J.S.D.); (M.L.S.); (J.W.C.); (A.B.B.); (G.A.)
| | - Maegan L. Sheehan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; (Y.C.B.); (K.J.S.D.); (M.L.S.); (J.W.C.); (A.B.B.); (G.A.)
| | - Jessica W. Chen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; (Y.C.B.); (K.J.S.D.); (M.L.S.); (J.W.C.); (A.B.B.); (G.A.)
| | - Jameson P. Davis
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA; (M.D.B.); (B.P.B.); (M.L.); (J.P.D.); (R.L.); (A.F.)
| | - Rosiane Lima
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA; (M.D.B.); (B.P.B.); (M.L.); (J.P.D.); (R.L.); (A.F.)
| | - Andrea G. Edlow
- Massachusetts General Hospital Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Vincent Center for Reproductive Biology, Boston, MA 02114, USA;
| | - Alessio Fasano
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA; (M.D.B.); (B.P.B.); (M.L.); (J.P.D.); (R.L.); (A.F.)
| | - Alejandro B. Balazs
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; (Y.C.B.); (K.J.S.D.); (M.L.S.); (J.W.C.); (A.B.B.); (G.A.)
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; (Y.C.B.); (K.J.S.D.); (M.L.S.); (J.W.C.); (A.B.B.); (G.A.)
| | - Lael M. Yonker
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA; (M.D.B.); (B.P.B.); (M.L.); (J.P.D.); (R.L.); (A.F.)
| |
Collapse
|
441
|
Bowen JE, Sprouse KR, Walls AC, Mazzitelli IG, Logue JK, Franko NM, Ahmed K, Shariq A, Cameroni E, Gori A, Bandera A, Posavad CM, Dan JM, Zhang Z, Weiskopf D, Sette A, Crotty S, Iqbal NT, Corti D, Geffner J, Grifantini R, Chu HY, Veesler D. Omicron BA.1 and BA.2 neutralizing activity elicited by a comprehensive panel of human vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.15.484542. [PMID: 35313570 PMCID: PMC8936098 DOI: 10.1101/2022.03.15.484542] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The SARS-CoV-2 Omicron variant of concern comprises three sublineages designated BA.1, BA.2, and BA.3, with BA.2 steadily replacing the globally dominant BA.1. We show that the large number of BA.1 and BA.2 spike mutations severely dampen plasma neutralizing activity elicited by infection or seven clinical vaccines, with cross-neutralization of BA.2 being consistently more potent than that of BA.1, independent of the vaccine platform and number of doses. Although mRNA vaccines induced the greatest magnitude of Omicron BA.1 and BA.2 plasma neutralizing activity, administration of a booster based on the Wuhan-Hu-1 spike sequence markedly increased neutralizing antibody titers and breadth against BA.1 and BA.2 across all vaccines evaluated. Our data suggest that although BA.1 and BA.2 evade polyclonal neutralizing antibody responses, current vaccine boosting regimens may provide sufficient protection against Omicron-induced disease.
Collapse
Affiliation(s)
- John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Kaitlin R. Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Ignacio G. Mazzitelli
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - Jennifer K. Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Nicholas M. Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Kumail Ahmed
- Department of Paediatrics and Child Health, and Biological & Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Asefa Shariq
- Department of Paediatrics and Child Health, and Biological & Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Elisabetta Cameroni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Andrea Gori
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation,, University of Milan, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation,, University of Milan, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy
| | - Christine M. Posavad
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jennifer M. Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA UC92037, USA
| | - Zeli Zhang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA UC92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA UC92037, USA
| | - Najeeha Talat Iqbal
- Department of Paediatrics and Child Health, and Biological & Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, 20122 Milan, Italy
| | - Helen Y. Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
442
|
Britton A, Fleming-Dutra KE, Shang N, Smith ZR, Dorji T, Derado G, Accorsi EK, Ajani UA, Miller J, Schrag SJ, Verani JR. Association of COVID-19 Vaccination With Symptomatic SARS-CoV-2 Infection by Time Since Vaccination and Delta Variant Predominance. JAMA 2022; 327:1032-1041. [PMID: 35157002 PMCID: PMC8845038 DOI: 10.1001/jama.2022.2068] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Monitoring COVID-19 vaccine performance over time since vaccination and against emerging variants informs control measures and vaccine policies. OBJECTIVE To estimate the associations between symptomatic SARS-CoV-2 infection and receipt of BNT162b2, mRNA-1273, and Ad26.COV2.S by day since vaccination before and during Delta variant predominance (pre-Delta period: March 13-May 29, 2021; Delta period: July 18-October 17, 2021). DESIGN, SETTING, AND PARTICIPANTS Test-negative, case-control design with data from 6884 US COVID-19 testing sites in the pharmacy-based Increasing Community Access to Testing platform. This study included 1 634 271 laboratory-based SARS-CoV-2 nucleic acid amplification tests (NAATs) from adults 20 years and older and 180 112 NAATs from adolescents 12 to 19 years old with COVID-19-like illness from March 13 to October 17, 2021. EXPOSURES COVID-19 vaccination (1 Ad26.COV2.S dose or 2 mRNA doses) 14 or more days prior. MAIN OUTCOMES AND MEASURES Association between symptomatic infection and prior vaccination measured using the odds ratio (OR) from spline-based multivariable logistic regression. RESULTS The analysis included 390 762 test-positive cases (21.5%) and 1 423 621 test-negative controls (78.5%) (59.9% were 20-44 years old; 9.9% were 12-19 years old; 58.9% were female; 71.8% were White). Among adults 20 years and older, the BNT162b2 mean OR for days 14 to 60 after a second dose (initial OR) was lower during the pre-Delta period (0.10 [95% CI, 0.09-0.11]) than during the Delta period (0.16 [95% CI, 0.16-0.17]) and increased with time since vaccination (per-month change in OR, pre-Delta: 0.04 [95% CI, 0.02-0.05]; Delta: 0.03 [95% CI, 0.02-0.03]). The initial mRNA-1273 OR was 0.05 (95% CI, 0.04-0.05) during the pre-Delta period, 0.10 (95% CI, 0.10-0.11) during the Delta period, and increased with time (per-month change in OR, pre-Delta: 0.02 [95% CI, 0.005-0.03]; Delta: 0.03 [95% CI, 0.03-0.04]). The Ad26.COV2.S initial OR was 0.42 (95% CI, 0.37-0.47) during the pre-Delta period and 0.62 (95% CI, 0.58-0.65) during the Delta period and did not significantly increase with time since vaccination. Among adolescents, the BNT162b2 initial OR during the Delta period was 0.06 (95% CI, 0.05-0.06) among 12- to 15-year-olds, increasing by 0.02 (95% CI, 0.01-0.03) per month, and 0.10 (95% CI, 0.09-0.11) among 16- to 19-year-olds, increasing by 0.04 (95% CI, 0.03-0.06) per month. CONCLUSIONS AND RELEVANCE Among adults, the OR for the association between symptomatic SARS-CoV-2 infection and COVID-19 vaccination (as an estimate of vaccine effectiveness) was higher during Delta variant predominance, suggesting lower protection. For mRNA vaccination, the steady increase in OR by month since vaccination was consistent with attenuation of estimated effectiveness over time; attenuation related to time was greater than that related to variant.
Collapse
Affiliation(s)
- Amadea Britton
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Nong Shang
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Zachary R. Smith
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Tandin Dorji
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Gordana Derado
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Emma K. Accorsi
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umed A. Ajani
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Joseph Miller
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Stephanie J. Schrag
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| | - Jennifer R. Verani
- Centers for Disease Control and Prevention COVID-19 Response, Atlanta, Georgia
| |
Collapse
|
443
|
Marcelin JR, Pettifor A, Janes H, Brown ER, Kublin JG, Stephenson KE. COVID-19 Vaccines and SARS-CoV-2 Transmission in the Era of New Variants: A Review and Perspective. Open Forum Infect Dis 2022; 9:ofac124. [PMID: 35493113 PMCID: PMC8992234 DOI: 10.1093/ofid/ofac124] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 11/22/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines have yielded definitive prevention and major reductions in morbidity and mortality from severe acute respiratory syndrome coronavirus 2 infection, even in the context of emerging and persistent variants of concern. Newer variants have revealed less vaccine protection against infection and attenuation of vaccine effects on transmission. COVID-19 vaccines still likely reduce transmission compared with not being vaccinated at all, even with variants of concern; however, determining the magnitude of transmission reduction is constrained by the challenges of performing these studies, requiring accurate linkage of infections to vaccine status and timing thereof, particularly within households. In this review, we synthesize the currently available data on the impact of COVID-19 vaccines on infection, serious illness, and transmission; we also identify the challenges and opportunities associated with policy development based on this data.
Collapse
Affiliation(s)
- Jasmine R Marcelin
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha Nebraska, USA
| | | | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth R Brown
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
444
|
Fontanarosa PB, Golub RM, Flanagin A. With Sincere Thanks and Appreciation to JAMA Authors and Reviewers. JAMA 2022; 327:931-933. [PMID: 35258543 DOI: 10.1001/jama.2022.2791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Phil B Fontanarosa
- Dr Fontanarosa is Interim Editor in Chief, Dr Golub is Executive Deputy Editor, and Ms Flanagin is Executive Managing Editor, JAMA and the JAMA Network
| | - Robert M Golub
- Dr Fontanarosa is Interim Editor in Chief, Dr Golub is Executive Deputy Editor, and Ms Flanagin is Executive Managing Editor, JAMA and the JAMA Network
| | - Annette Flanagin
- Dr Fontanarosa is Interim Editor in Chief, Dr Golub is Executive Deputy Editor, and Ms Flanagin is Executive Managing Editor, JAMA and the JAMA Network
| |
Collapse
|
445
|
Chenchula S, Karunakaran P. Current Evidence on Efficacy of COVID-19 Booster Dose Vaccination Against the Omicron Variant. A Systematic Review. J Med Virol 2022; 94:2969-2976. [PMID: 35246846 PMCID: PMC9088621 DOI: 10.1002/jmv.27697] [Citation(s) in RCA: 188] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/02/2022] [Indexed: 12/16/2022]
Abstract
Coronavirus disease 2019 (COVID‐19) is an ongoing pandemic, which affected around 45 million confirmed cases of COVID‐19, including more than 6 million deaths. However, on November 24, 2021, the World Health Organization announced a new severe acute respiratory syndrome coronavirus 2 variant designated as the B.1.1.529, a variant of concern (VOC), and the variant has been named as “Omicron.” Available preliminary evidence suggests that, as compared with previous VOCs, it has an increased risk of infectivity. Studies have shown that protection from various vaccines effectiveness against hospitalization and death from severe COVID‐19 disease is decreasing slowly after a two‐dose schedule of COVID‐19 vaccines. In response to experiencing a new COVID‐19 variant and ongoing resurgence of cases, the importance of COVID‐19 vaccine booster dose and durability of the effect of the third dose of vaccine against COVID‐19 Omicron variant is controversial yet. To address this, we conducted a systematic literature survey on effectiveness of the third or booster dose of COVID‐19 vaccine against the Omicron variant. We have performed a systematic search in PubMed (Medline), Google Scholar, and MedRXiv database, from inception to January 2022 using the MeSH terms and keywords “Corona Virus Disease‐2019 OR COVID‐19 AND Omicron AND COVID‐19 Booster Vaccine.” We have identified a total of 27 published studies. We have reviewed all the eligible available studies on the effectiveness of the COVID‐19 vaccine booster shots against the Omicron variant. This review may be helpful in accelerating the COVID‐19 booster dose vaccination.
Collapse
Affiliation(s)
- Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Mangalagiri, Andhra Pradesh, India
| | | |
Collapse
|
446
|
Klein NP, Stockwell MS, Demarco M, Gaglani M, Kharbanda AB, Irving SA, Rao S, Grannis SJ, Dascomb K, Murthy K, Rowley EA, Dalton AF, DeSilva MB, Dixon BE, Natarajan K, Stenehjem E, Naleway AL, Lewis N, Ong TC, Patel P, Konatham D, Embi PJ, Reese SE, Han J, Grisel N, Goddard K, Barron MA, Dickerson M, Liao IC, Fadel WF, Yang DH, Arndorfer J, Fireman B, Griggs EP, Valvi NR, Hallowell C, Zerbo O, Reynolds S, Ferdinands J, Wondimu MH, Williams J, Bozio CH, Link-Gelles R, Azziz-Baumgartner E, Schrag SJ, Thompson MG, Verani JR. Effectiveness of COVID-19 Pfizer-BioNTech BNT162b2 mRNA Vaccination in Preventing COVID-19-Associated Emergency Department and Urgent Care Encounters and Hospitalizations Among Nonimmunocompromised Children and Adolescents Aged 5-17 Years - VISION Network, 10 States, April 2021-January 2022. MMWR. MORBIDITY AND MORTALITY WEEKLY REPORT 2022; 71:352-358. [PMID: 35239634 PMCID: PMC8893336 DOI: 10.15585/mmwr.mm7109e3] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The efficacy of the BNT162b2 (Pfizer-BioNTech) vaccine against laboratory-confirmed COVID-19 exceeded 90% in clinical trials that included children and adolescents aged 5-11, 12-15, and 16-17 years (1-3). Limited real-world data on 2-dose mRNA vaccine effectiveness (VE) in persons aged 12-17 years (referred to as adolescents in this report) have also indicated high levels of protection against SARS-CoV-2 (the virus that causes COVID-19) infection and COVID-19-associated hospitalization (4-6); however, data on VE against the SARS-CoV-2 B.1.1.529 (Omicron) variant and duration of protection are limited. Pfizer-BioNTech VE data are not available for children aged 5-11 years. In partnership with CDC, the VISION Network* examined 39,217 emergency department (ED) and urgent care (UC) encounters and 1,699 hospitalizations† among persons aged 5-17 years with COVID-19-like illness across 10 states during April 9, 2021-January 29, 2022,§ to estimate VE using a case-control test-negative design. Among children aged 5-11 years, VE against laboratory-confirmed COVID-19-associated ED and UC encounters 14-67 days after dose 2 (the longest interval after dose 2 in this age group) was 46%. Among adolescents aged 12-15 and 16-17 years, VE 14-149 days after dose 2 was 83% and 76%, respectively; VE ≥150 days after dose 2 was 38% and 46%, respectively. Among adolescents aged 16-17 years, VE increased to 86% ≥7 days after dose 3 (booster dose). VE against COVID-19-associated ED and UC encounters was substantially lower during the Omicron predominant period than the B.1.617.2 (Delta) predominant period among adolescents aged 12-17 years, with no significant protection ≥150 days after dose 2 during Omicron predominance. However, in adolescents aged 16-17 years, VE during the Omicron predominant period increased to 81% ≥7 days after a third booster dose. During the full study period, including pre-Delta, Delta, and Omicron predominant periods, VE against laboratory-confirmed COVID-19-associated hospitalization among children aged 5-11 years was 74% 14-67 days after dose 2, with wide CIs that included zero. Among adolescents aged 12-15 and 16-17 years, VE 14-149 days after dose 2 was 92% and 94%, respectively; VE ≥150 days after dose 2 was 73% and 88%, respectively. All eligible children and adolescents should remain up to date with recommended COVID-19 vaccinations, including a booster dose for those aged 12-17 years.
Collapse
|
447
|
Immune response to SARS-CoV-2 after a booster of mRNA-1273: an open-label phase 2 trial. Nat Med 2022; 28:1042-1049. [PMID: 35241844 PMCID: PMC9117133 DOI: 10.1038/s41591-022-01739-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/09/2022] [Indexed: 01/09/2023]
Abstract
Rising breakthrough infections of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in previously immunized individuals have raised concerns for the need for a booster vaccine dose to combat waning antibody levels and new variants. Here we report the results of the open-label, non-randomized part B of a phase 2 trial in which we evaluated the safety and immunogenicity of a booster injection of 50 µg of the coronavirus disease 2019 (COVID-19) vaccine mRNA-1273 in 344 adult participants immunized 6-8 months earlier with a primary series of two doses of 50 µg or 100 µg of mRNA-1273 ( NCT04405076 ). Neutralizing antibody (nAb) titers against wild-type SARS-CoV-2 at 1 month after the booster were 1.7-fold (95% confidence interval (CI): 1.5, 1.9) higher than those at 28 days after the second injection of the primary series, which met the pre-specified non-inferiority criterion (primary immunogenicity objective) and might indicate a memory B cell response. The nAb titers against the Delta variant (B.1.617.2) (exploratory objective) at 1 month after the booster were 2.1-fold (95% CI: 1.8, 2.4) higher than those at 28 days after the second injection of the primary series. The seroresponse rate (95% CI (four-fold rise from baseline)) was 100% (98.7, 100.0) at 28 days after the booster compared to 98.3% (96.0, 99.4) after the primary series. The higher antibody titers at 28 days after the booster dose compared to 28 days after the second dose in the phase 3 COVE study were also observed in two assays for anti-spike IgG antibody measured by ELISA and by Meso Scale Discovery (MSD) Multiplex. The frequency of solicited local and systemic adverse reactions after the booster dose was similar to that after the second dose in the primary two-dose series of mRNA-1273 (50 µg or 100 µg); no new signals were observed in the unsolicited adverse events; and no serious adverse events were reported in the 1-month follow-up period. These results show that a booster injection of mRNA-1273 more than 6 months after completing the primary two-dose series is safe and elicited nAb titers that were statistically significantly higher than the peak titers detected after the primary vaccination series, suggesting that a booster dose of mRNA-1273 might result in increased vaccine effectiveness against infection and disease caused by SARS-CoV-2.
Collapse
|
448
|
Oh Omikron! HAUTNAH 2022. [PMCID: PMC8908757 DOI: 10.1007/s12326-022-00501-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
449
|
Ferré VM, Lebourgeois S, Chenane HR, Menidjel R, Masson C, Collin G, Visseaux B, Descamps D, Fidouh N, Charpentier C. Vaccine Ab neutralization against Omicron and SARS-CoV-2 variants using neutralization and specific ELISA assays. J Infect 2022; 84:834-872. [PMID: 35278483 PMCID: PMC8906917 DOI: 10.1016/j.jinf.2022.02.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 02/27/2022] [Indexed: 12/26/2022]
Affiliation(s)
- Valentine Marie Ferré
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Samuel Lebourgeois
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Houssem Redha Chenane
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Reyene Menidjel
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Christelle Masson
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Gilles Collin
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Benoit Visseaux
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Diane Descamps
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Nadhira Fidouh
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France
| | - Charlotte Charpentier
- Service de Virologie, Université de Paris, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, 46 Rue Henri Huchard, Paris F-75018, France.
| |
Collapse
|
450
|
Ao D, Lan T, He X, Liu J, Chen L, Baptista‐Hon DT, Zhang K, Wei X. SARS-CoV-2 Omicron variant: Immune escape and vaccine development. MedComm (Beijing) 2022; 3:e126. [PMID: 35317190 PMCID: PMC8925644 DOI: 10.1002/mco2.126] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
New genetic variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) constantly emerge through unmitigated spread of the virus in the ongoing Coronavirus disease 2019 pandemic. Omicron (B.1.1.529), the latest variant of concern (VOC), has so far shown exceptional spread and infectivity and has established itself as the dominant variant in recent months. The SARS-CoV-2 spike glycoprotein is a key component for the recognition and binding to host cell angiotensin-converting enzyme 2 receptors. The Omicron variant harbors a cluster of substitutions/deletions/insertions, and more than 30 mutations are located in spike. Some noticeable mutations, including K417N, T478K, N501Y, and P681H, are shared with the previous VOCs Alpha, Beta, Gamma, or Delta variants and have been proven to be associated with higher transmissibility, viral infectivity, and immune evasion potential. Studies have revealed that the Omicron variant is partially resistant to the neutralizing activity of therapeutic antibodies and convalescent sera, which poses significant challenges for the clinical effectiveness of the current vaccines and therapeutic antibodies. We provide a comprehensive analysis and summary of the epidemiology and immune escape mechanisms of the Omicron variant. We also suggest some therapeutic strategies against the Omicron variant. This review, therefore, aims to provide information for further research efforts to prevent and contain the impact of new VOCs during the ongoing pandemic.
Collapse
Affiliation(s)
- Danyi Ao
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jian Liu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Daniel T. Baptista‐Hon
- Center for Biomedicine and InnovationsFaculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Kang Zhang
- Center for Biomedicine and InnovationsFaculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|