1
|
Mukhopadhyay S, Kaufman DA, Saha S, Puopolo KM, Flannery DD, Weimer KED, Greenberg RG, Sanchez PJ, Eichenwald EC, Cotten CM, Stoll BJ, Laptook A. Late-Onset Sepsis Among Extremely Preterm Infants During the COVID-19 Pandemic. Pediatrics 2025; 155:e2024067675. [PMID: 39842471 PMCID: PMC11908436 DOI: 10.1542/peds.2024-067675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/21/2024] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVES To compare incidence of late-onset sepsis (LOS) among extremely preterm infants before and during the COVID-19 pandemic. METHODS Multicenter cohort study of infants with birthweight 401 to 1000 g or gestational age 22 to 28 weeks. LOS was defined as a bacterial or fungal pathogen isolated from blood or cerebrospinal fluid culture obtained after 72 hours of age. Primary outcome was LOS incidence calculated as incidence proportion (LOS cases among all admissions) and incidence rate (LOS events/1000 patient days). A multivariable Poisson regression model was used to compare the adjusted risk of LOS incidence proportion before (1/1/18-3/31/20) and during the pandemic (4/1/20-12/31/21). An interrupted time series analysis using a generalized linear mixed model with center as a random effect was used to compare LOS incidence rates during the 2 periods. RESULTS Among 6509 eligible infants, LOS incidence proportion was not different before (18.2%) and during the pandemic (16.9%; P = .18). The adjusted relative risk (95% CI) for LOS was 0.93 (0.82-1.05) and for LOS or mortality was 0.98 (0.88-1.08) during the pandemic compared to the period before the pandemic. In the interrupted time series analysis, there was no significant change in LOS incidence rates at the start of the pandemic (0.219, 95% CI, -0.453 to 0.891) or microbiology of LOS, and change in trends of LOS incidence rates before and during the pandemic was not significant (-0.005, 95% CI, -0.025 to 0.015). CONCLUSIONS In a large multicenter study of extremely preterm infants, rates of LOS remained unchanged during the pandemic.
Collapse
Affiliation(s)
- Sagori Mukhopadhyay
- Division of Neonatology, Children’s Hospital of
Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania
Perelman School of Medicine, Philadelphia, PA
- Clinical Futures, CHOP Research Institute,
Children’s Hospital of Philadelphia, Philadelphia, PA
| | | | - Shampa Saha
- Social, Statistical and Environmental Sciences Unit, RTI
International, Research Triangle Park, NC
| | - Karen M. Puopolo
- Division of Neonatology, Children’s Hospital of
Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania
Perelman School of Medicine, Philadelphia, PA
- Clinical Futures, CHOP Research Institute,
Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Dustin D. Flannery
- Division of Neonatology, Children’s Hospital of
Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania
Perelman School of Medicine, Philadelphia, PA
- Clinical Futures, CHOP Research Institute,
Children’s Hospital of Philadelphia, Philadelphia, PA
| | | | | | - Pablo J. Sanchez
- Department of Pediatrics, Nationwide Children’s
Hospital, The Ohio State University College of Medicine, Columbus, OH
| | - Eric C. Eichenwald
- Division of Neonatology, Children’s Hospital of
Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania
Perelman School of Medicine, Philadelphia, PA
| | | | | | - Abbot Laptook
- Department of Pediatrics, The Warren Alpert Medical School,
Brown University
| |
Collapse
|
2
|
Didembourg M, David C, Morimont L, Cransquint E, Favresse J, Douxfils J, Gillot C. Evaluation of neutralizing antibody titers against SARS-CoV-2 JN.1 omicron subvariant during pregnancy - A case series study. Heliyon 2025; 11:e41249. [PMID: 39811280 PMCID: PMC11729664 DOI: 10.1016/j.heliyon.2024.e41249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Background SARS-CoV-2 infection during pregnancy poses health risks to both mother and fetus. This study investigates neutralizing antibodies (NAbs) against the SARS-CoV-2 JN.1 Omicron subvariant in pregnant women, focusing on responses to natural infection, vaccination, and passive immunity. Methods A single-center, prospective study collected blood samples from 19 pregnant women at various pregnancy stages and postpartum. NAb titers were analyzed using a pseudovirus neutralization assay, with statistical analyses (p-value <0.05) conducted using unpaired t-test with Welch's correction. Results Among participants, 63.2 % had at least one positive NAb titer, with only one vaccinated case. No significant difference in NAb titers was found between symptomatic and asymptomatic women. NAbs were detected in cord blood, especially when infection or vaccination occurred close to delivery, indicating passive immunity transfer to the newborn. Conclusion NAb titers change dynamically during pregnancy, increasing then decreasing. Most pregnant women were asymptomatic and NAbs were effectively transferred to the fetus when infection or vaccination occurred near delivery. These findings highlight the importance of vaccination timing, suggesting late second or third trimester vaccination may provide better protection, emphasizing the need for adherence to vaccination guidelines to optimize maternal and neonatal immunity.
Collapse
Affiliation(s)
- Marie Didembourg
- Clinical Pharmacology and Toxicology Research Unit, Namur Research Institute for Life Sciences, University of Namur, 5000, Namur, Belgium
- Qualiblood s.a., Research and Development Department, Liège, Belgium
| | - Clara David
- Qualiblood s.a., Research and Development Department, Liège, Belgium
| | - Laure Morimont
- Clinical Pharmacology and Toxicology Research Unit, Namur Research Institute for Life Sciences, University of Namur, 5000, Namur, Belgium
- Qualiblood s.a., Research and Development Department, Liège, Belgium
| | - Eva Cransquint
- Clinical Pharmacology and Toxicology Research Unit, Namur Research Institute for Life Sciences, University of Namur, 5000, Namur, Belgium
| | - Julien Favresse
- Clinical Pharmacology and Toxicology Research Unit, Namur Research Institute for Life Sciences, University of Namur, 5000, Namur, Belgium
- Department of Laboratory Medicine, Clinique Saint Luc Bouge, Bouge, Belgium
| | - Jonathan Douxfils
- Clinical Pharmacology and Toxicology Research Unit, Namur Research Institute for Life Sciences, University of Namur, 5000, Namur, Belgium
- Qualiblood s.a., Research and Development Department, Liège, Belgium
- Department of Biological Hematology, Centre Hospitalier Universitaire Clermont-Ferrand, Hôpital Estaing, Clermont-Ferrand, France
| | - Constant Gillot
- Clinical Pharmacology and Toxicology Research Unit, Namur Research Institute for Life Sciences, University of Namur, 5000, Namur, Belgium
| |
Collapse
|
3
|
M. Sami M, Al Zuheiri S, Sabaneh NK, Amir Abdul Latif M, Al-Blooshi SY, Osman M. COVID-19 and Pregnancy Outcomes: A Descriptive Study From a Tertiary Hospital in Ras Al Khaimah, UAE. Obstet Gynecol Int 2024; 2024:5252919. [PMID: 39659767 PMCID: PMC11631290 DOI: 10.1155/ogi/5252919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/03/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Background: Over 768 million cases of COVID-19 infection have been reported worldwide, with pregnant women being the most vulnerable members of society during such an infectious disease outbreak. In the United Arab Emirates, there are limited studies explaining the effects of COVID-19 on pregnant women and their fetuses. In this study, the maternal and fetal outcomes in pregnant women with COVID-19 in a tertiary maternal hospital, United Arab Emirates, were examined. Materials and Methods: A descriptive study was conducted in a tertiary hospital for Obstetrics and Gynecology in Ras Al Khaimah, UAE. The study included all pregnant women who tested positive for COVID-19 infection from April 2020 to September 2021. Results: The study revealed that a higher number of COVID-19-infected pregnant patients presented in their third trimester (69.1%). The comorbidity of body mass index (BMI) had the most effect on the severity/hospitalization status of the patients (p=0.018). In the nonhospitalized group, fever was the most common symptom (26%), whereas in the hospitalized group, cough was the most common symptom (94%). Emergency cesarean delivery was found to be significant (p=0.0007) in hospitalized patients. COVID-19 pneumonia was the prevailing adverse maternal outcome. NICU admission and prematurity were the most frequent neonatal outcomes. Conclusions: In conclusion, our findings show that adverse maternal outcomes, obesity, and mode of delivery were related to COVID-19 severity in pregnant patients. However, there was no effect generally on the adverse fetal outcomes except for jaundice and birth weight.
Collapse
Affiliation(s)
- Manal M. Sami
- Department of Pathology, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Shatha Al Zuheiri
- Obstetrics and Gynecology Department, Abdullah Bin Omran Hospital for Obstetrics and Gynecology, Emirates Health Services, Ras Al Khaimah, UAE
| | - Nour K. Sabaneh
- Clinical Sciences Department, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Mustafa Amir Abdul Latif
- Clinical Sciences Department, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Shooq Yousef Al-Blooshi
- Clinical Sciences Department, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Mira Osman
- Clinical Sciences Department, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| |
Collapse
|
4
|
Druškovič M, Lučovnik M, Mesarič VA, Kavšek G, Vidmar Šimic M, Trojner Bregar A, Avšič Županc T, Ihan A, Premru Sršen T. Immune Response to SARS-CoV-2 in Vaccine-naive Pregnant Women: Assessment of IgG and IgA Antibody Profile at Delivery and 42 Days Postpartum. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1371-1379. [PMID: 39258926 DOI: 10.4049/jimmunol.2400055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024]
Abstract
This prospective cohort study assessed the SARS-CoV-2 IgG and IgA Ab profiles at delivery and 42 d postpartum in unvaccinated SARS-CoV-2-positive pregnant women and determined the association with the timing and the clinical course of the infection. A total of 387 vaccine-naive women with confirmed SARS-CoV-2 infection during pregnancy were included. IgG and IgA Abs were detected in maternal blood at delivery and 42 d postpartum using ELISA kits. The relationships between Ab detection and value and clinical features, including the timing of the infection, were analyzed using univariate and multivariate logistic and linear regression models. The mean gestational age at infection was 31 4/7 wk of pregnancy. Symptoms of SARS-CoV-2 infection were present in 88.1% of women. IgG and IgA Abs were detected in 45.7 and 58.9% at delivery, respectively, increasing to 72.7 and 76.8% at 42 d postpartum. Detection of IgG and IgA Abs in maternal blood at delivery was independently associated with symptomatic infection (adjusted odds ratio [OR] 3.13, 95% confidence interval (CI): 1.47-6.69 and adjusted OR 3.62, 95% CI: 1.8-7.26, respectively), but not with the time from positive swab to delivery or gestational age at positive swab. Detection of Abs at 42 d postpartum was also strongly associated with the detection of Abs at delivery (OR 29.97, 95% CI: 10.11-88.82 for IgG and OR 13.09, 95% CI: 6.37-26.9 for IgA). Vaccine-naive pregnant women exhibit a significant and durable immune response to SARS-CoV-2, which is more pronounced in symptomatic women but independent of gestational age at diagnosis or the diagnosis-to-delivery interval.
Collapse
Affiliation(s)
- Mirjam Druškovič
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Lučovnik
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vita Andreja Mesarič
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Gorazd Kavšek
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Marijana Vidmar Šimic
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Andreja Trojner Bregar
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tatjana Avšič Županc
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Cell Immunology, Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Cell Immunology, Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Premru Sršen
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
5
|
Anderson AR, Strouse JJ, Manwani D, Brandow AM, Vichinsky E, Campbell A, Leavey PJ, Nero A, Ibrahim IF, Field JJ, Baer A, Soto-Calderon H, Vincent L, Zhao Y, Santos JJS, Hensley SE, Mortier N, Lanzkron S, Neuberg D, Abrams CS. COVID-19 mRNA vaccination responses in individuals with sickle cell disease: an ASH RC Sickle Cell Research Network Study. Blood Adv 2024; 8:4549-4553. [PMID: 38991137 PMCID: PMC11399661 DOI: 10.1182/bloodadvances.2024013878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
ABSTRACT Children and adults with sickle cell disease (SCD) have increases in morbidity and mortality with COVID-19 infections. The American Society of Hematology Research Collaborative Sickle Cell Disease Research Network performed a prospective COVID-19 vaccine study to assess antibody responses and analyze whether messenger RNA (mRNA) vaccination precipitated any adverse effects unique to individuals with SCD. Forty-one participants received 2 doses of the Pfizer-BioNTech vaccine and provided baseline blood samples before vaccination and 2 months after the initial vaccination for analysis of immunoglobulin G (IgG) reactivity against the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 spike protein. Six-month IgG reactivity against the viral RBD was also available in 37 patients. Postvaccination reactogenicity was common and similar to the general population. There were no fevers that required inpatient admission. Vaso-occlusive pain within 2 to 3 days of first or second vaccination was reported by 5 participants (12%) including 4 (10%) who sought medical care. Twenty-seven participants (66%) were seropositive at baseline, and all 14 initially seronegative participants (34%) converted to seropositive after vaccination. Overall, mRNA vaccination had a good risk-benefit profile in individuals with SCD. This mRNA vaccine study also marks the first evaluation of vaccine safety and antibody response in very young children with SCD. This trial was registered at www.ClinicalTrials.gov as #NCT05139992.
Collapse
Affiliation(s)
- Alan R. Anderson
- PRISMA Health Comprehensive Sickle Cell Disease Program, Division of Pediatric Hematology-Oncology, University of South Carolina School of Medicine, Greenville, SC
| | - John J. Strouse
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Deepa Manwani
- Children's Hospital at Montefiore, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin and Children’s Research Institute of Children’s Wisconsin, Milwaukee, WI
| | - Elliott Vichinsky
- Benioff Children's Hospital Oakland, Department of Pediatrics, UCSF, Oakland, CA
| | - Andrew Campbell
- Department of Pediatrics, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Patrick J. Leavey
- Simmons Comprehensive Cancer Center, Children's Medical Center, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Alecia Nero
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Ibrahim F. Ibrahim
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Joshua J. Field
- Department of Medicine, Medical College of Wisconsin, Versiti Blood Research Institute, Milwaukee, WI
| | - Amanda Baer
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Lauren Vincent
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yan Zhao
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | | | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | | | - Sophie Lanzkron
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Charles S. Abrams
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
6
|
Chong EA, Kumashie KG, Chong ER, Fabrizio J, Gupta A, Svoboda J, Barta SK, Walsh KM, Napier EB, Lundberg RK, Nasta SD, Gerson JN, Landsburg DJ, Gonzalez J, Gaano A, Weirick ME, McAllister CM, Awofolaju M, John GN, Kammerman SC, Novacek J, Pajarillo R, Lundgreen KA, Tanenbaum N, Gouma S, Drapeau EM, Adamski S, D’Andrea K, Pattekar A, Hicks A, Korte S, Sharma H, Herring S, Williams JC, Hamilton JT, Bates P, Hensley SE, Prak ETL, Greenplate AR, Wherry EJ, Schuster SJ, Ruella M, Vella LA. Immunologic Predictors of Vaccine Responsiveness in Patients With Lymphoma and Chronic Lymphocytic Leukemia. J Infect Dis 2024; 230:15-27. [PMID: 39052709 PMCID: PMC11272091 DOI: 10.1093/infdis/jiae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/13/2024] [Accepted: 02/24/2024] [Indexed: 03/06/2024] Open
Abstract
Patients with B-cell lymphomas have altered cellular components of vaccine responses due to malignancy and therapy, and the optimal timing of vaccination relative to therapy remains unknown. Severe acute respiratory syndrome coronavirus 2 vaccines created an opportunity for new insights in vaccine timing because patients were challenged with a novel antigen across multiple phases of treatment. We studied serologic messenger RNA vaccine response in retrospective and prospective cohorts with lymphoma and chronic lymphocytic leukemia, paired with clinical and research immune parameters. Reduced serologic response was observed more frequently during active treatment, but nonresponse was also common within observation and posttreatment groups. Total immunoglobulin A and immunoglobulin M correlated with successful vaccine response. In individuals treated with anti-CD19-directed chimeric antigen receptor-modified T cells, nonresponse was associated with reduced B and T follicular helper cells. Predictors of vaccine response varied by disease and therapeutic group, and therefore further studies of immune health during and after cancer therapies are needed to individualize vaccine timing.
Collapse
Affiliation(s)
- Elise A Chong
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | | | - Emeline R Chong
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Joseph Fabrizio
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Aditi Gupta
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Jakub Svoboda
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Stefan K Barta
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Kristy M Walsh
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Ellen B Napier
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Rachel K Lundberg
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Sunita D Nasta
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - James N Gerson
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Daniel J Landsburg
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | | | | | | | | | | | - Gavin N John
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
| | - Shane C Kammerman
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
| | - Josef Novacek
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
| | | | | | | | | | | | - Sharon Adamski
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Kurt D’Andrea
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Ajinkya Pattekar
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine
| | - Amanda Hicks
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Scott Korte
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Harsh Sharma
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Sarah Herring
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | | | - Jacob T Hamilton
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | | | | | | | | | - E John Wherry
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Stephen J Schuster
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Marco Ruella
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
- Center for Cellular Immunotherapies
- Institute for Immunology
| | - Laura A Vella
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
- Department of Pathology and Laboratory Medicine
| |
Collapse
|
7
|
Figueroa-Romero A, Atchadé A, Yadouleton A, Fiogbe M, Bonnet E, Yovo E, Accrombessi M, Hounsa S, Paper T, Dupont R, Gaudart J, Le Hesran JY, Massougbodji A, Cottrell G, González R. SARS-CoV-2 seroprevalence among Beninese pregnant women in the third year of the pandemic. BMC Public Health 2024; 24:1762. [PMID: 38956517 PMCID: PMC11221113 DOI: 10.1186/s12889-024-19087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Pregnant women are a vulnerable population to COVID-19 given an increased susceptibility to severe SARS-CoV-2 infection and pregnancy complications. However, few SARS-CoV-2 serological surveys have been performed among this population to assess the extent of the infection in sub-Saharan countries. The objectives of this study were to determine SARS-CoV-2 seroprevalence among Beninese pregnant women, to identify spatial seropositivity clusters and to analyse factors associated with the infection. METHODS A cross-sectional study including women in their third trimester of pregnancy attending the antenatal care (ANC) clinics at Allada (south Benin) and Natitingou (north Benin) was conducted. Rapid diagnostic tests (RDT) for detection of IgG/IgM against the SARS-CoV-2 spike protein were performed using capillary blood. Seroprevalence of SARS-CoV-2 antibodies and associations between SARS-CoV-2 serostatus and maternal characteristics were analyzed by multivariate logistic regression. Spatial analyses were performed using the spatial scan statistics to identify spatial clusters of SARS-CoV-2 infection. RESULTS A total of 861 pregnant women were enrolled between May 4 and June 29, 2022. 58/861 (6.7%) participants reported having received COVID-19 vaccine. None of the participants had been diagnosed with COVID-19 during their pregnancy. SARS-CoV-2 antibodies were detected in 607/802 (75.7%; 95% CI 72.56%-78.62%) of unvaccinated participants. Several urban and rural spatial clusters of SARS-CoV-2 cases were identified in Allada and one urban spatial cluster was identified in Natitingou. Unvaccinated participants from Allada with at least one previous morbidity were at a three-times higher risk of presenting SARS-CoV-2 antibodies (OR = 2.89; 95%CI 1.19%-7.00%). CONCLUSION Three out of four pregnant women had SARS-CoV-2 antibodies, suggesting a high virus circulation among pregnant women in Benin, while COVID-19 vaccination coverage was low. Pregnant women with comorbidities may be at increased risk of SARS-CoV-2 infection. This population should be prioritized for COVID-19 diagnosis and vaccination in order to prevent its deleterious effects. TRIAL REGISTRATION NCT06170320 (retrospectively registered on December 21, 2023).
Collapse
Affiliation(s)
- Antía Figueroa-Romero
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic- Universitat de Barcelona, Barcelona, Spain.
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain.
| | - Aurore Atchadé
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Anges Yadouleton
- Laboratoire des fièvres hémorragiques virales du Bénin, Ministère de la Santé du Bénin, Cotonou, 01BP918, Bénin
| | - Marc Fiogbe
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Emmanuel Bonnet
- Institut de recherche pour le développement PRODIG UMR 215, CNRS Université Paris 1 Panthéon- Sorbonne, AgroParisTech 5, cours des Humanités, Aubervilliers, Île-de-France, F-93 322, France
| | - Emmanuel Yovo
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Manfred Accrombessi
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
- Faculty of Infectious and Tropical Diseases, Disease Control Department, London School of Hygiene & Tropical Medicine, London, UK
- Population Services International, Malaria Department, Country-Based Global Employee, Cotonou, Benin
| | - Sandrine Hounsa
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Thierry Paper
- Biosynex S.A, 22 boulevard Sebastien Brant, Illkirch Graffenstaden, Strasbourg, F-67400, France
| | - Raphael Dupont
- Biosynex S.A, 22 boulevard Sebastien Brant, Illkirch Graffenstaden, Strasbourg, F-67400, France
| | - Jean Gaudart
- Aix Marseille Univ, IRD, INSERM, SESSTIM, ISSPAM, AP-HM, Hop La Timone, BioSTIC, Biostatistic and ICT, Marseille, France
| | - Jean-Yves Le Hesran
- Institut de Recherche pour le Développement, MERIT UMR216, Université Paris-Cité, Faculté de pharmacie, laboratoire de parasitologie, Paris, France
| | | | - Gilles Cottrell
- Institut de Recherche pour le Développement, MERIT UMR216, Université Paris-Cité, Faculté de pharmacie, laboratoire de parasitologie, Paris, France
| | - Raquel González
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic- Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| |
Collapse
|
8
|
Flannery DD, Shah NC, Puopolo KM. Perinatal COVID-19: Implications for care of the newborn. Semin Perinatol 2024; 48:151921. [PMID: 38871489 DOI: 10.1016/j.semperi.2024.151921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The maternal/newborn dyad presents special challenges to infection management. Early in the COVID-19 pandemic, lack of information regarding SARS-CoV-2 transmission and virulence made it difficult to develop appropriate care guidance when pregnant persons had COVID-19 at the time of presentation for childbirth. We will review the considerations for the parturient, newborn, and care team, and describe the evolution of perinatal COVID management guidance.
Collapse
Affiliation(s)
- Dustin D Flannery
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Neil C Shah
- Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
| | - Karen M Puopolo
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
9
|
Lv A, BianBaZhuoMa, DeQiong, DaWaZhuoMa, PuBuZhuoMa, Yao D, LangJiQuZhen, Lu Y, Cai L, DaZhen, Tang C, BianBaZhuoMa, Zhang Y, Yin J, Ding T, DaWaCang, Wu M, Chen Y, Li Y. Effect of COVID-19 infection on pregnant women in plateau regions. Public Health 2024; 229:57-62. [PMID: 38401193 DOI: 10.1016/j.puhe.2023.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/04/2023] [Accepted: 12/28/2023] [Indexed: 02/26/2024]
Abstract
OBJECTIVE The present study aims to explore the effect of COVID-19 infection on pregnant women in plateau regions. STUDY DESIGN Data from 381 pregnant women infected with COVID-19 who underwent prenatal examination or treatment at Women and Children's Hospital of Tibet Autonomous Region between January 2020 and December 2022 and 314 pregnant women not infected with COVID-19 were retrospectively collected. METHODS The study participants were divided into an infected and non-infected group according to whether they were infected with COVID-19. Basic information (ethnicity, age, body mass index and gestational age [GA]), vaccination status, intensive care unit (ICU) admission and delivery outcomes were compared. Binary logistic regression was used to analyse the influencing factors of ICU admission. RESULTS The results revealed significant differences in the GA, vaccination rate, blood pressure, partial pressure of oxygen, white blood cell (WBC) count, ICU admission rate, preeclampsia rate, forearm presentation rate, thrombocytopenia rate, syphilis infection rate and placental abruption rate between the two groups (P < 0.05). A univariate analysis showed that COVID-19 infection, hepatitis B virus infection, the WBC count and hypoproteinaemia were risk factors for ICU admission. The results of the multivariate analysis of the ICU admission of pregnant women showed that COVID-19 infection (odds ratio [OR] = 4.271, 95 % confidence interval [CI]: 3.572-5.820, P < 0.05) was a risk factor for ICU admission and the WBC count (OR = 0.935, 95 % CI: 0.874-0.947, P < 0.05) was a protective factor for ICU admission. CONCLUSION Pregnant women are vulnerable to the adverse consequences of COVID-19 infection, and public health measures such as vaccination are needed to protect this population subgroup.
Collapse
Affiliation(s)
- A Lv
- Department of Obstetrics and Gynecology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO. 1 DaHua Road, Dong Dan, Beijing, 100730, PR China; Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - BianBaZhuoMa
- Lhasa People's Hospital, No. 1, Beijing Middle Road, Chengguan District, Lhasa, Tibet Autonomous Region, 850000, PR China
| | - DeQiong
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - DaWaZhuoMa
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - PuBuZhuoMa
- Lhasa People's Hospital, No. 1, Beijing Middle Road, Chengguan District, Lhasa, Tibet Autonomous Region, 850000, PR China
| | - D Yao
- Nyingchi People's Hospital, No. 11, Water Garden, Bayi Town, Bayi District, Nyingchi City, Tibet Autonomous Region, 860000, PR China
| | - LangJiQuZhen
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - Y Lu
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - L Cai
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - DaZhen
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - C Tang
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - BianBaZhuoMa
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - Y Zhang
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - J Yin
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - T Ding
- Women and Children's Hospital of Tibet Autonomous Region, NO. 10 Chagu Avenue, Doilungdêqên District, Liuwu New Area, Lhasa, Tibet Autonomous Region, 851414, PR China
| | - DaWaCang
- Tibet University Medical School, No. 10, Zangda East Road, Chengguan District, Lhasa, Tibet Autonomous Region, 850000, PR China
| | - M Wu
- Tibet University Medical School, No. 10, Zangda East Road, Chengguan District, Lhasa, Tibet Autonomous Region, 850000, PR China
| | - Y Chen
- Tibet University Medical School, No. 10, Zangda East Road, Chengguan District, Lhasa, Tibet Autonomous Region, 850000, PR China
| | - Y Li
- Department of Obstetrics and Gynecology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO. 1 DaHua Road, Dong Dan, Beijing, 100730, PR China.
| |
Collapse
|
10
|
Farrell R, Dahler C, Pope R, Divoky E, Collart C. COVID testing hesitancy among pregnant patients: Lessons learned from the COVID-19 pandemic about the unique needs and challenges of medically complex populations. RESEARCH SQUARE 2024:rs.3.rs-3892181. [PMID: 38352470 PMCID: PMC10862955 DOI: 10.21203/rs.3.rs-3892181/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Background Pregnant patients were a significant population to consider during the pandemic, given the impact of SARS-CoV-2 infection on obstetric outcomes. While COVID testing was a central pillar of infection control, it became apparent that a subset of the population declined to test. At the same time, data emerged about pregnant persons also declining to test. Yet, it was unknown why pregnant patients declined tests and if those reasons were similar or different from those of the general population. We conducted this study to explore pregnant patients' attitudes, access, and utilization of COVID-19 testing to support healthcare for infection prevention management for this unique and medically complex population. Methods We conducted a qualitative study of patients who were currently or recently pregnant during the early stages of the pandemic and received outpatient prenatal care at one of the participating study sites. An interview guide was used to conduct in-depth telephone interviews. Coding was performed using NVivo, and analysis was conducted using Grounded Theory. Results The average age of the participants (N = 37) was 32 (SD 4.21) years. Most were < 35 years of age (57%) and self-described as White (68%). Qualitative analysis identified themes related to barriers to COVID-19 testing access and use during pregnancy, including concerns about test accuracy, exposure to COVID-19 in testing facilities, isolation and separation during labor and delivery, and diminished healthcare quality and patient experience. Conclusions The implementation of widespread and universal COVID testing policies did not address the unique needs and challenges of pregnant patients as a medically complex population. It is important to understand the reasons and implications for pregnant patients who declined COVID testing during the current pandemic to inform strategies to prevent infection spread in future public health emergencies.
Collapse
|
11
|
Virtanen J, Korhonen EM, Salonen S, Vapalahti O, Sironen T, Jääskeläinen AJ. SARS-CoV-2 infections among pregnant women, 2020, Finland-Cross-testing of neutralization assays. J Med Virol 2024; 96:e29415. [PMID: 38293724 DOI: 10.1002/jmv.29415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
We studied the development of the severe acute respiratory syndrome-related coronavirus (SARS-CoV-2) pandemic in southern Finland in 2020 and evaluated the performance of two surrogate immunoassays for the detection of neutralizing antibodies (NAbs). The data set consisted of 12 000 retrospectively collected samples from pregnant women in their first trimester throughout 2020. All the samples were initially screened for immunoglobulin G (IgG) with SARS-CoV-2 spike antibody assay (EIM-S1, Euroimmun) followed by confirmation with nucleocapsid antibody assay (Architect SARS-CoV-2, Abbott). Samples that were reactive (positive or borderline) with both assays were subjected to testing with commercial surrogate immunoassays of NeutraLISA (EIM) and cPassTM (GenScript Biotech Corporation) by using pseudoneutralization assay (PNAbA) as a golden standard. No seropositive cases were detected between January and March. Between April and December, IgG (EIM-S1 and Abbott positive) and NAb (PNAbA positive) seroprevalences were between 0.4% and 1.4%. NeutraLISA showed 90% and cPass 55% concordant results with PNAbA among PNAbA negative samples and 49% and 92% among PNAbA positive samples giving NeutraLISA better specificity but lower sensitivity than cPass. To conclude, seroprevalence in pregnant women reflected that of the general population but the variability of the performance of serological protocols needs to be taken into account in inter-study comparison.
Collapse
Affiliation(s)
- Jenni Virtanen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Essi M Korhonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Sami Salonen
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Olli Vapalahti
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tarja Sironen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Anne J Jääskeläinen
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
12
|
Rayo MN, Aquise A, Fernandez-Buhigas I, Gonzalez-Gea L, Garcia-Gonzalez C, Sanchez-Tudela M, Rodriguez-Fernandez M, Tuñon-Le Poultel D, Santacruz B, Gil MM. Maternal COVID-19 Serological Changes-Comparison between Seroconversion Rate in First and Third Trimesters of Pregnancy and Subsequent Obstetric Complications: A Cohort Study. Viruses 2023; 15:2386. [PMID: 38140627 PMCID: PMC10747315 DOI: 10.3390/v15122386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Pregnant women are especially vulnerable to respiratory diseases. We aimed to study seroconversion rates during pregnancy in a cohort of consecutive pregnancies tested in the first and third trimesters and to compare the maternal and obstetric complications in the women who seroconverted in the first trimester and those who did so in the third. This was an observational cohort study carried out at the Hospital Universitario de Torrejón, in Madrid, Spain, during the first peak of the COVID-19 pandemic. All consecutive singleton pregnancies with a viable fetus attending their 11-13-week scan between 1 January and 15 May 2020 were included and seropositive women for SARS-CoV2 were monthly follow up until delivery. Antibodies against SARS-CoV-2 (IgA and IgG) were analyzed on stored serum samples obtained from first- and third-trimester routine antenatal bloods in 470 pregnant women. Antibodies against SARS-CoV-2 were detected in 31 (6.6%) women in the first trimester and in 66 (14.0%) in the third trimester, including 48 (10.2%) that were negative in the first trimester (seroconversion during pregnancy). Although the rate of infection was significantly higher in the third versus the first trimester (p = 0.003), no significant differences in maternal or obstetric complications were observed in women testing positive in the first versus the third trimester.
Collapse
Affiliation(s)
- Maria N. Rayo
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Adriana Aquise
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Irene Fernandez-Buhigas
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Lorena Gonzalez-Gea
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Coral Garcia-Gonzalez
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Mirian Sanchez-Tudela
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Miguel Rodriguez-Fernandez
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
| | | | - Belen Santacruz
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Maria M. Gil
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, 28850 Madrid, Spain; (M.N.R.); (I.F.-B.); (L.G.-G.); (C.G.-G.); (M.S.-T.); (M.R.-F.); (B.S.)
- School of Medicine, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, 28223 Madrid, Spain
| |
Collapse
|
13
|
Painter MM, Johnston TS, Lundgreen KA, Santos JJS, Qin JS, Goel RR, Apostolidis SA, Mathew D, Fulmer B, Williams JC, McKeague ML, Pattekar A, Goode A, Nasta S, Baxter AE, Giles JR, Skelly AN, Felley LE, McLaughlin M, Weaver J, Kuthuru O, Dougherty J, Adamski S, Long S, Kee M, Clendenin C, da Silva Antunes R, Grifoni A, Weiskopf D, Sette A, Huang AC, Rader DJ, Hensley SE, Bates P, Greenplate AR, Wherry EJ. Prior vaccination promotes early activation of memory T cells and enhances immune responses during SARS-CoV-2 breakthrough infection. Nat Immunol 2023; 24:1711-1724. [PMID: 37735592 DOI: 10.1038/s41590-023-01613-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 08/07/2023] [Indexed: 09/23/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of vaccinated individuals is increasingly common but rarely results in severe disease, likely due to the enhanced potency and accelerated kinetics of memory immune responses. However, there have been few opportunities to rigorously study early recall responses during human viral infection. To better understand human immune memory and identify potential mediators of lasting vaccine efficacy, we used high-dimensional flow cytometry and SARS-CoV-2 antigen probes to examine immune responses in longitudinal samples from vaccinated individuals infected during the Omicron wave. These studies revealed heightened spike-specific responses during infection of vaccinated compared to unvaccinated individuals. Spike-specific cluster of differentiation (CD)4 T cells and plasmablasts expanded and CD8 T cells were robustly activated during the first week. In contrast, memory B cell activation, neutralizing antibody production and primary responses to nonspike antigens occurred during the second week. Collectively, these data demonstrate the functionality of vaccine-primed immune memory and highlight memory T cells as rapid responders during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mark M Painter
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Timothy S Johnston
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendall A Lundgreen
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jefferson J S Santos
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Juliana S Qin
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Rishi R Goel
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Sokratis A Apostolidis
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Bria Fulmer
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Justine C Williams
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Michelle L McKeague
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ahmad Goode
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Sean Nasta
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Amy E Baxter
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Josephine R Giles
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ashwin N Skelly
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura E Felley
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Maura McLaughlin
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Joellen Weaver
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeanette Dougherty
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Sharon Adamski
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Sherea Long
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Macy Kee
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Cynthia Clendenin
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alexander C Huang
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn Medicine Biobank, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott E Hensley
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Paul Bates
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Immune Health, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Coggins SA, Mukhopadhyay S, Triebwasser J, Downes KJ, Christie JD, Puopolo KM. Association of delivery risk phenotype with early-onset sepsis in preterm infants. J Perinatol 2023; 43:1166-1172. [PMID: 37543652 PMCID: PMC10530160 DOI: 10.1038/s41372-023-01743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
OBJECTIVE To determine delivery risk phenotype-specific incidence of early-onset sepsis (EOS) among preterm infants. STUDY DESIGN Retrospective cohort study of infants born <35 weeks' gestation at four perinatal centers during 2017-2021. Infants were classified into one of six delivery risk phenotypes incorporating delivery mode, presence of labor, and duration of rupture of membranes (ROM). The primary outcome was EOS incidence within the overall cohort and each risk phenotype. RESULTS Among 2937 preterm infants, 21 had EOS (0.7%, or 7.1 cases/1000 preterm infants). The majority of EOS cases (13/21, 62%) occurred in the setting of prolonged ROM ≥ 18 h, with a phenotype incidence of 23.8 cases/1000 preterm infants. There were no EOS cases among infants born by cesarean section without ROM (with or without labor), nor via cesarean section with ROM < 18 h without labor. CONCLUSION Delivery risk phenotyping may inform EOS risk stratification in preterm infants.
Collapse
Affiliation(s)
- Sarah A Coggins
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA.
- Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
- Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jourdan Triebwasser
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Kevin J Downes
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
- Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jason D Christie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | - Karen M Puopolo
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
- Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
15
|
Colamatteo A, Fusco C, Micillo T, D'Hooghe T, de Candia P, Alviggi C, Longobardi S, Matarese G. Immunobiology of pregnancy: from basic science to translational medicine. Trends Mol Med 2023; 29:711-725. [PMID: 37331882 DOI: 10.1016/j.molmed.2023.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023]
Abstract
Embryo implantation failure and spontaneous abortions represent the main causes of infertility in developed countries. Unfortunately, incomplete knowledge of the multiple factors involved in implantation and fetal development keeps the success rate of medically assisted procreation techniques relatively low. According to recent literature, cellular and molecular mechanisms of 'immunogenic tolerance' towards the embryo are crucial to establish an 'anti-inflammatory' state permissive of a healthy pregnancy. In this review we dissect the role played by the immune system in the endometrial-embryo crosstalk, with a particular emphasis towards the fork-head-box-p3 (Foxp3+) CD4+CD25+ regulatory T (Treg) cells and discuss the most recent therapeutic advances in the context of early immune-mediated pregnancy loss.
Collapse
Affiliation(s)
- Alessandra Colamatteo
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Clorinda Fusco
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy; Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Teresa Micillo
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Thomas D'Hooghe
- Global Medical Affairs Fertility, Merck Healthcare KGaA, Darmstadt, Germany; Research Group Reproductive Medicine, Department of Development and Regeneration, Organ Systems, Group Biomedical Sciences, KU Leuven (University of Leuven), Leuven, Belgium; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Paola de Candia
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Carlo Alviggi
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | | | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy; Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Napoli, Italy.
| |
Collapse
|
16
|
Skarke C, Lordan R, Barekat K, Naik A, Mathew D, Ohtani T, Greenplate AR, Grant GR, Lahens NF, Gouma S, Troisi E, Sengupta A, Weljie AM, Meng W, Luning Prak ET, Lundgreen K, Bates P, Meng H, FitzGerald GA. Modulation of the Immune Response to Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination by Nonsteroidal Anti-Inflammatory Drugs. J Pharmacol Exp Ther 2023; 386:198-204. [PMID: 37105582 PMCID: PMC10353078 DOI: 10.1124/jpet.122.001415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/13/2023] [Accepted: 02/09/2023] [Indexed: 04/29/2023] Open
Abstract
Evidence is scarce to guide the use of nonsteroidal anti-inflammatory drugs (NSAIDs) to mitigate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine-related adverse effects, given the possibility of blunting the desired immune response. In this pilot study, we deeply phenotyped a small number of volunteers who did or did not take NSAIDs concomitant with SARS-CoV-2 immunizations to seek initial information on the immune response. A SARS-CoV-2 vaccine-specific receptor binding domain (RBD) IgG antibody response and efficacy in the evoked neutralization titers were evident irrespective of concomitant NSAID consumption. Given the sample size, only a large and consistent signal of immunomodulation would have been detectable, and this was not apparent. However, the information gathered may inform the design of a definitive clinical trial. Here we report a series of divergent omics signals that invites additional hypotheses testing. SIGNIFICANCE STATEMENT: The impact of nonsteroidal anti-inflammatory drugs (NSAIDs) on the immune response elicited by repeat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunizations was profiled by immunophenotypic, proteomic, and metabolomic approaches in a clinical pilot study of small sample size. A SARS-CoV-2 vaccine-specific immune response was evident irrespective of concomitant NSAID consumption. The information gathered may inform the design of a definitive clinical trial.
Collapse
Affiliation(s)
- Carsten Skarke
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kayla Barekat
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Amruta Naik
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Divij Mathew
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Takuya Ohtani
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Allison R Greenplate
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Gregory R Grant
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sigrid Gouma
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth Troisi
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Arjun Sengupta
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Aalim M Weljie
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Wenzhao Meng
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Eline T Luning Prak
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kendall Lundgreen
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Paul Bates
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Hu Meng
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Abhold J, Wozniak A, Mulcahy J, Walsh S, Zepeda E, Demmer R, Yendell S, Hedberg C, Ulrich A, Wurtz R, Beebe T. Demographic, social, and behavioral correlates of SARS-CoV-2 seropositivity in a representative, population-based study of Minnesota residents. PLoS One 2023; 18:e0279660. [PMID: 37319239 PMCID: PMC10270347 DOI: 10.1371/journal.pone.0279660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Monitoring COVID-19 infection risk in the general population is a public health priority. Few studies have measured seropositivity using representative, probability samples. The present study measured seropositivity in a representative population of Minnesota residents prior to vaccines and assess the characteristics, behaviors, and beliefs of the population at the outset of the pandemic and their association with subsequent infection. METHODS Participants in the Minnesota COVID-19 Antibody Study (MCAS) were recruited from residents of Minnesota who participated in the COVID-19 Household Impact Survey (CIS), a population-based survey that collected data on physical health, mental health, and economic security information between April 20 and June 8 of 2020. This was followed by collection of antibody test results between December 29, 2020 and February 26, 2021. Demographic, behavioral, and attitudinal exposures were assessed for association with the outcome of interest, SARS-CoV-2 seroprevalence, using univariate and multivariate logistic regression. RESULTS Of the 907 potential participants from the CIS, 585 respondents then consented to participate in the antibody testing (64.4% consent rate). Of these, results from 537 test kits were included in the final analytic sample, and 51 participants (9.5%) were seropositive. The overall weighted seroprevalence was calculated to be 11.81% (95% CI, 7.30%-16.32%) at of the time of test collection. In adjusted multivariate logistic regression models, significant associations between seroprevalence and the following were observed; being from 23-64 and 65+ age groups were both associated with higher odds of COVID-19 seropositivity compared to the 18-22 age group (17.8 [1.2-260.1] and 24.7 [1.5-404.4] respectively). When compared to a less than $30k annual income reference group, all higher income groups had significantly lower odds of seropositivity. Reporting practicing a number of 10 (median reported value in sample) or more of 19 potential COVID-19 mitigation factors (e.g. handwashing and mask wearing) was associated with lower odds of seropositivity (0.4 [0.1-0.99]) Finally, the presence of at least one household member in the age range of 6 to 17 years old was associated with higher odds of seropositivity (8.3 [1.2-57.0]). CONCLUSIONS The adjusted odds ratio of SARS-CoV-2 seroprevalence was significantly positively associated with increasing age and having household member(s) in the 6-17 year age group, while increasing income levels and a mitigation score at or above the median were shown to be significantly protective factors.
Collapse
Affiliation(s)
- Jordan Abhold
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| | - Abigail Wozniak
- Opportunity & Inclusive Growth Institute, Federal Reserve Bank of Minneapolis, Minneapolis, MN, United States of America
| | - John Mulcahy
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| | - Sara Walsh
- Health Sciences, NORC at the University of Chicago, Chicago, IL, United States of America
| | - Evelyn Zepeda
- Health Sciences, NORC at the University of Chicago, Chicago, IL, United States of America
| | - Ryan Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States of America
| | - Stephanie Yendell
- Health Risk Intervention Unit, Minnesota Department of Health, St. Paul, MN, United States of America
| | - Craig Hedberg
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| | - Angela Ulrich
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
- Center for Infectious Disease Research and Policy, Office of the Vice President for Research, University of Minnesota, Minneapolis, MN, United States of America
| | - Rebecca Wurtz
- School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| | - Timothy Beebe
- School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
18
|
Pecks U, Agel L, Doubek KJ, Hagenbeck C, Jennewein L, von Kaisenberg C, Kranke P, Leitner S, Mand N, Rüdiger M, Zöllkau J, Mingers N, Sitter M, Louwen F. SARS-CoV-2 in Pregnancy, Birth and Puerperium. Guideline of the DGGG and DGPM (S2k-Level, AWMF Registry Number 015/092, March 2022). Geburtshilfe Frauenheilkd 2023; 83:517-546. [PMID: 39258218 PMCID: PMC11384259 DOI: 10.1055/a-2003-5983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 03/11/2023] Open
Abstract
Objective This S2k guideline of the German Society for Gynecology and Obstetrics (DGGG) and the German Society of Perinatal Medicine (DGPM) contains consensus-based recommendations for the care and treatment of pregnant women, parturient women, women who have recently given birth, and breastfeeding women with SARS-CoV-2 infection and their newborn infants. The aim of the guideline is to provide recommendations for action in the time of the COVID-19 pandemic for professionals caring for the above-listed groups of people. Methods The PICO format was used to develop specific questions. A systematic targeted search of the literature was carried out using PubMed, and previously formulated statements and recommendations issued by the DGGG and the DGPM were used to summarize the evidence. This guideline also drew on research data from the CRONOS registry. As the data basis was insufficient for a purely evidence-based guideline, the guideline was compiled using an S2k-level consensus-based process. After summarizing and presenting the available data, the guideline authors drafted recommendations in response to the formulated PICO questions, which were then discussed and voted on. Recommendations Recommendations on hygiene measures, prevention measures and care during pregnancy, delivery, the puerperium and while breastfeeding were prepared. They also included aspects relating to the monitoring of mother and child during and after infection with COVID-19, indications for thrombosis prophylaxis, caring for women with COVID-19 while they are giving birth, the presence of birth companions, postnatal care, and testing and monitoring the neonate during rooming-in or on the pediatric ward.
Collapse
Affiliation(s)
- Ulrich Pecks
- Klinik für Gynäkologie und Geburtshilfe, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Lena Agel
- Technische Hochschule Aschaffenburg, Hebammenkunde, Aschaffenburg, Germany
| | | | - Carsten Hagenbeck
- Geburtshilfe und Perinatalmedizin, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Lukas Jennewein
- Geburtshilfe und Pränatalmedizin, Universitätsklinikum Frankfurt Goethe-Universität, Frankfurt am Main, Germany
| | - Constantin von Kaisenberg
- Pränatalmedizin und Geburtshilfe im Perinatalzentrum, Universitätsklinik der Medizinischen Hochschule Hannover, Hannover, Germany
| | - Peter Kranke
- Klinik und Poliklinik für Anästhesiologie, Intensivmedizin, Notfallmedizin und Schmerztherapie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Sabine Leitner
- Bundesverband "Das frühgeborene Kind" e. V., Frankfurt, Germany
| | - Nadine Mand
- Philipps-Universität Marburg, Zentrum für Kinder- und Jugendmedizin, Marburg, Germany
| | - Mario Rüdiger
- Klinik für Kinder- und Jugendmedizin, Fachbereich Neonatologie und pädiatrische Intensivmedizin, Medizinische Fakultät der TU Dresden, Dresden, Germany
| | - Janine Zöllkau
- Klinik für Geburtsmedizin, Universitätsklinikum Jena, Jena, Germany
| | - Nina Mingers
- Klinik für Gynäkologie und Geburtshilfe, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Magdalena Sitter
- Klinik und Poliklinik für Anästhesiologie, Intensivmedizin, Notfallmedizin und Schmerztherapie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Frank Louwen
- Geburtshilfe und Pränatalmedizin, Universitätsklinikum Frankfurt Goethe-Universität, Frankfurt am Main, Germany
| |
Collapse
|
19
|
Painter MM, Johnston TS, Lundgreen KA, Santos JJS, Qin JS, Goel RR, Apostolidis SA, Mathew D, Fulmer B, Williams JC, McKeague ML, Pattekar A, Goode A, Nasta S, Baxter AE, Giles JR, Skelly AN, Felley LE, McLaughlin M, Weaver J, Kuthuru O, Dougherty J, Adamski S, Long S, Kee M, Clendenin C, da Silva Antunes R, Grifoni A, Weiskopf D, Sette A, Huang AC, Rader DJ, Hensley SE, Bates P, Greenplate AR, Wherry EJ. Prior vaccination enhances immune responses during SARS-CoV-2 breakthrough infection with early activation of memory T cells followed by production of potent neutralizing antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.05.527215. [PMID: 36798171 PMCID: PMC9934532 DOI: 10.1101/2023.02.05.527215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
SARS-CoV-2 infection of vaccinated individuals is increasingly common but rarely results in severe disease, likely due to the enhanced potency and accelerated kinetics of memory immune responses. However, there have been few opportunities to rigorously study early recall responses during human viral infection. To better understand human immune memory and identify potential mediators of lasting vaccine efficacy, we used high-dimensional flow cytometry and SARS-CoV-2 antigen probes to examine immune responses in longitudinal samples from vaccinated individuals infected during the Omicron wave. These studies revealed heightened Spike-specific responses during infection of vaccinated compared to unvaccinated individuals. Spike-specific CD4 T cells and plasmablasts expanded and CD8 T cells were robustly activated during the first week. In contrast, memory B cell activation, neutralizing antibody production, and primary responses to non-Spike antigens occurred during the second week. Collectively, these data demonstrate the functionality of vaccine-primed immune memory and highlight memory T cells as rapid responders during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mark M Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Timothy S Johnston
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kendall A Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Jefferson J S Santos
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Juliana S Qin
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Rishi R Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Sokratis A Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Bria Fulmer
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Justine C Williams
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Michelle L McKeague
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Ahmad Goode
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Sean Nasta
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Amy E Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Josephine R Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Ashwin N Skelly
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura E Felley
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Maura McLaughlin
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Joellen Weaver
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Jeanette Dougherty
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Sharon Adamski
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Sherea Long
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Macy Kee
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Cynthia Clendenin
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alexander C Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott E Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| |
Collapse
|
20
|
Miyadahira MY, Brizot MDL, Alexander N, Cerdeira Sabino E, Campos de Oliveira da Silva L, Hoshida MS, da Silva Sousa Oliveira AM, Silva Farche AC, Pulcineli Vieira Francisco R, Mayaud P. Monitoring SARS-CoV-2 seroprevalence over time among pregnant women admitted to delivery units: Suitability for surveillance. PLoS One 2023; 18:e0280109. [PMID: 36603011 DOI: 10.1371/journal.pone.0280109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES To determine SARS-CoV-2 seroprevalence over time and risk factors among pregnant women at delivery in São Paulo, Brazil; and to evaluate the suitability of pregnant women as a sentinel population for SARS-CoV-2 serosurveillance. METHODS Unselected consecutive pregnant women presenting at the labor ward of a single large hospital between July 20th 2020 to February 21st 2021 were enrolled and tested for SARS-CoV-2 serology using two assays: the rapid chromatic Wondfo One Step (for total IgA and IgG detection) and Roche Elecsys assay (detecting anti-nucleoprotein [N] IgG). SARS-CoV-2 seroprevalence was computed as smooth spline function over time with 95% confidence intervals (CI). Risk factors were evaluated for positivity by each assay. We compared timepoint seroprevalence by the two assays with four concomitant community household surveys (HHS), in which the Roche assay was used, to determine the sensitivity and relevance of the pregnant women population as sentinel population. RESULTS Overall SARS-CoV-2 seroprevalence was 28.9% (221/763) by Roche and 17.9% (137/763) by Wondfo. Reported symptoms experienced during pregnancy were all significantly correlated with being SARS-CoV-2 seropositive at delivery with any assay (with odds-ratios ranging from 3.0 [95% CI: 2.1-4.3] for coryza to 22.8 [95% CI: 12.3-46.6] for ageusia). Seropositivity by either assay was high in women at delivery in the early period of the pandemic (June 2020), compared with seropositivity in women from the concomitant HHS: 44.1% (95% CI: 21.8-66.4) for Roche, 54.1% (30.9-78.5) for Wondfo, versus 11.4% (95% CI: 9.2-13.6) for HHS. For later periods (October 2020 and January 2021), the seropositivity in women at delivery measured by Roche corresponded well with the prevalence found among women in the HHS using the same assay, whilst prevalence measured by Wondfo dropped. CONCLUSIONS Women at delivery represent a highly exposed and readily accessible population for sentinel surveillance of emerging infections such as SARS-CoV-2.
Collapse
Affiliation(s)
- Mariana Yumi Miyadahira
- Disciplina de Obstetrícia, Departamento de Obstetrícia e Ginecologia da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Maria de Lourdes Brizot
- Disciplina de Obstetrícia, Departamento de Obstetrícia e Ginecologia da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Neal Alexander
- Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ester Cerdeira Sabino
- Departamento de Moléstias Infecciosas e Parasitárias, Instituto de Medicina Tropical da FMUSP, São Paulo, Brazil
| | | | - Mara Sandra Hoshida
- Disciplina de Obstetrícia, Departamento de Obstetrícia e Ginecologia da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Laboratório de Investigação Médica- LIM 57, Hospital das Clínicas da FMUSP, São Paulo, Brazil
| | - Ana Maria da Silva Sousa Oliveira
- Disciplina de Obstetrícia, Departamento de Obstetrícia e Ginecologia da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Ana Claudia Silva Farche
- Disciplina de Obstetrícia, Departamento de Obstetrícia e Ginecologia da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Rossana Pulcineli Vieira Francisco
- Disciplina de Obstetrícia, Departamento de Obstetrícia e Ginecologia da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Hospital Universitário da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Philippe Mayaud
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
21
|
Accurti V, Gambitta B, Iodice S, Manenti A, Boito S, Dapporto F, Leonardi M, Molesti E, Fabietti I, Montomoli E, Bollati V, Persico N. SARS-CoV-2 Seroconversion and Pregnancy Outcomes in a Population of Pregnant Women Recruited in Milan, Italy, between April 2020 and October 2020. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph192416720. [PMID: 36554602 PMCID: PMC9778651 DOI: 10.3390/ijerph192416720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 06/12/2023]
Abstract
The possible link between SARS-CoV-2 infection and adverse pregnancy outcomes has so far demonstrated heterogeneous results in terms of maternal, fetal, and neonatal complications. We aim to investigate the correlation between SARS-CoV-2 seroconversion and/or neutralization titer and pregnancy outcomes. We analyzed a population of 528 pregnant women followed up from the first trimester of gestation until delivery. For each woman, we collected a first blood sample between 11 and 13 weeks of gestation and a second sample in the perinatal period (between peripartum and puerperium) to assess the presence of SARS-CoV-2 antibodies and/or microneutralization titer (MN titer). Data on pregnancy outcomes (gestational age at delivery, preterm birth before 34 weeks, hypertensive disorders, gestational diabetes, and abnormal fetal growth) were collected. We observed that serologic status per se is not associated with major pregnancy complications. On the contrary, the MN titer was associated with increased odds of gestational diabetes. Although we mainly reported asymptomatic SARS-CoV-2 infections and the absence of severe maternal and neonatal adverse outcomes, SARS-CoV-2 infection might challenge the maternal immune system and explain the moderate increase in adverse outcome odds.
Collapse
Affiliation(s)
- Veronica Accurti
- Fetal Medicine and Surgery Service, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Bianca Gambitta
- Fetal Medicine and Surgery Service, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Simona Iodice
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | | | - Simona Boito
- Fetal Medicine and Surgery Service, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | | | | | - Isabella Fabietti
- Fetal Medicine and Surgery Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Emanuele Montomoli
- VisMederi Srl, 53100 Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Valentina Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Occupational Health Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Nicola Persico
- Fetal Medicine and Surgery Service, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
22
|
Abdolmaleki G, Taheri MA, Paridehpour S, Mohammadi NM, Tabatabaei YA, Mousavi T, Amin M. A comparison between SARS-CoV-1 and SARS-CoV2: an update on current COVID-19 vaccines. Daru 2022; 30:379-406. [PMID: 36050585 PMCID: PMC9436716 DOI: 10.1007/s40199-022-00446-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/05/2022] [Indexed: 10/31/2022] Open
Abstract
Since the outbreak of the novel coronavirus disease 2019 (COVID-19) in Wuhan, China, many health care systems have been heavily engaged in treating and preventing the disease, and the year 2020 may be called as "historic COVID-19 vaccine breakthrough". Due to the COVID-19 pandemic, many companies have initiated investigations on developing an efficient and safe vaccine against the virus. From Moderna and Pfizer in the United States to PastocoVac in Pasteur Institute of Iran and the University of Oxford in the United Kingdom, different candidates have been introduced to the market. COVID-19 vaccine research has been facilitated based on genome and structural information, bioinformatics predictions, epitope mapping, and data obtained from the previous developments of severe acute respiratory syndrome coronavirus (SARS-CoV or SARS-CoV-1) and middle east respiratory syndrome coronavirus (MERS-CoV) vaccine candidates. SARS-CoV genome sequence is highly homologous to the one in COVID-19 and both viruses use the same receptor, angiotensin-converting enzyme 2 (ACE2). Moreover, the immune system responds to these viruses, partially in the same way. Considering the on-going COVID-19 pandemic and previous attempts to manufacture SARS-CoV vaccines, this paper is going to discuss clinical cases as well as vaccine challenges, including those related to infrastructures, transportation, possible adverse reactions, utilized delivery systems (e.g., nanotechnology and electroporation) and probable vaccine-induced mutations.
Collapse
Affiliation(s)
- Gelareh Abdolmaleki
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Mina Azam Taheri
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Sarina Paridehpour
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Neshaut Mashreghi Mohammadi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Ahmadi Tabatabaei
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Taraneh Mousavi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran.
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Verma A, Damrauer SM, Naseer N, Weaver J, Kripke CM, Guare L, Sirugo G, Kember RL, Drivas TG, Dudek SM, Bradford Y, Lucas A, Judy R, Verma SS, Meagher E, Nathanson KL, Feldman M, Ritchie MD, Rader DJ, BioBank FTPM. The Penn Medicine BioBank: Towards a Genomics-Enabled Learning Healthcare System to Accelerate Precision Medicine in a Diverse Population. J Pers Med 2022; 12:jpm12121974. [PMID: 36556195 PMCID: PMC9785650 DOI: 10.3390/jpm12121974] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 12/02/2022] Open
Abstract
The Penn Medicine BioBank (PMBB) is an electronic health record (EHR)-linked biobank at the University of Pennsylvania (Penn Medicine). A large variety of health-related information, ranging from diagnosis codes to laboratory measurements, imaging data and lifestyle information, is integrated with genomic and biomarker data in the PMBB to facilitate discoveries and translational science. To date, 174,712 participants have been enrolled into the PMBB, including approximately 30% of participants of non-European ancestry, making it one of the most diverse medical biobanks. There is a median of seven years of longitudinal data in the EHR available on participants, who also consent to permission to recontact. Herein, we describe the operations and infrastructure of the PMBB, summarize the phenotypic architecture of the enrolled participants, and use body mass index (BMI) as a proof-of-concept quantitative phenotype for PheWAS, LabWAS, and GWAS. The major representation of African-American participants in the PMBB addresses the essential need to expand the diversity in genetic and translational research. There is a critical need for a "medical biobank consortium" to facilitate replication, increase power for rare phenotypes and variants, and promote harmonized collaboration to optimize the potential for biological discovery and precision medicine.
Collapse
Affiliation(s)
- Anurag Verma
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence: (A.V.); (D.J.R.)
| | - Scott M. Damrauer
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nawar Naseer
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - JoEllen Weaver
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Colleen M. Kripke
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lindsay Guare
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgio Sirugo
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachel L. Kember
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theodore G. Drivas
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott M. Dudek
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuki Bradford
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anastasia Lucas
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Renae Judy
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shefali S. Verma
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emma Meagher
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine L. Nathanson
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Feldman
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J. Rader
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence: (A.V.); (D.J.R.)
| | | |
Collapse
|
24
|
Flannery DD, Gouma S, Dhudasia MB, Mukhopadhyay S, Pfeifer MR, Woodford EC, Briker SM, Triebwasser JE, Gerber JS, Morris JS, Weirick ME, McAllister CM, Hensley SE, Puopolo KM. Comparison of Maternal and Neonatal Antibody Levels After COVID-19 Vaccination vs SARS-CoV-2 Infection. JAMA Netw Open 2022; 5:e2240993. [PMID: 36350652 PMCID: PMC9647482 DOI: 10.1001/jamanetworkopen.2022.40993] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
Importance Pregnant persons are at an increased risk of severe COVID-19 from SARS-CoV-2 infection, and COVID-19 vaccination is currently recommended during pregnancy. Objective To ascertain the association of vaccine type, time from vaccination, gestational age at delivery, and pregnancy complications with placental transfer of antibodies to SARS-CoV-2. Design, Setting, and Participants This cohort study was conducted in Pennsylvania Hospital in Philadelphia, Pennsylvania, and included births at the study site between August 9, 2020, and April 25, 2021. Maternal and cord blood serum samples were available for antibody level measurements for maternal-neonatal dyads. Exposures SARS-CoV-2 infection vs COVID-19 vaccination. Main Outcomes and Measures IgG antibodies to the receptor-binding domain of the SARS-CoV-2 spike protein were measured by quantitative enzyme-linked immunosorbent assay. Antibody concentrations and transplacental transfer ratios were measured after SARS-CoV-2 infection or receipt of COVID-19 vaccines. Results A total of 585 maternal-newborn dyads (median [IQR] maternal age, 31 [26-35] years; median [IQR] gestational age, 39 [38-40] weeks) with maternal IgG antibodies to SARS-CoV-2 detected at the time of delivery were included. IgG was detected in cord blood from 557 of 585 newborns (95.2%). Among 169 vaccinated persons without SARS-CoV-2 infection, the interval from first dose of vaccine to delivery ranged from 12 to 122 days. The geometric mean IgG level among 169 vaccine recipients was significantly higher than that measured in 408 persons after infection (33.88 [95% CI, 27.64-41.53] arbitrary U/mL vs 2.80 [95% CI, 2.50-3.13] arbitrary U/mL). Geometric mean IgG levels were higher after vaccination with the mRNA-1273 (Moderna) vaccine compared with the BNT162b2 (Pfizer/BioNTech) vaccine (53.74 [95% CI, 40.49-71.33] arbitrary U/mL vs 25.45 [95% CI, 19.17-33.79] arbitrary U/mL; P < .001). Placental transfer ratios were lower after vaccination compared with after infection (0.80 [95% CI, 0.68-0.93] vs 1.06 [95% CI, 0.98-1.14]; P < .001) but were similar between the mRNA vaccines (mRNA-1273: 0.70 [95% CI, 0.55-0.90]; BNT162b2: 0.85 [95% CI, 0.69-1.06]; P = .25). Time from infection or vaccination to delivery was associated with transfer ratio in models that included gestational age at delivery and maternal hypertensive disorders, diabetes, and obesity. Placental antibody transfer was detectable as early as 26 weeks' gestation. Transfer ratio that was higher than 1.0 was present for 48 of 51 (94.1%) births at 36 weeks' gestation or later by 8 weeks after vaccination. Conclusions and Relevance This study found that maternal and cord blood IgG antibody levels were higher after COVID-19 vaccination compared with after SARS-CoV-2 infection, with slightly lower placental transfer ratios after vaccination than after infection. The findings suggest that time from infection or vaccination to delivery was the most important factor in transfer efficiency.
Collapse
Affiliation(s)
- Dustin D. Flannery
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Miren B. Dhudasia
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Madeline R. Pfeifer
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Emily C. Woodford
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sara M. Briker
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | | | - Jeffrey S. Gerber
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Jeffrey S. Morris
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Madison E. Weirick
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | | | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Karen M. Puopolo
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Fisher BT, Sharova A, Boge CLK, Gouma S, Kamrin A, Blumenstock J, Shuster S, Gianchetti L, Collins D, Akaho E, Weirick ME, McAllister CM, Bolton MJ, Arevalo CP, Goodwin EC, Anderson EM, Christensen SR, Balamuth F, John ARO, Li Y, Coffin S, Gerber JS, Hensley SE. Evolution of severe acute respiratory coronavirus virus 2 (SARS-CoV-2) seroprevalence among employees of a US academic children's hospital during coronavirus disease 2019 (COVID-19) pandemic. Infect Control Hosp Epidemiol 2022; 43:1647-1655. [PMID: 34852866 PMCID: PMC8668398 DOI: 10.1017/ice.2021.487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/30/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To describe the cumulative seroprevalence of severe acute respiratory coronavirus virus 2 (SARS-CoV-2) antibodies during the coronavirus disease 2019 (COVID-19) pandemic among employees of a large pediatric healthcare system. DESIGN, SETTING, AND PARTICIPANTS Prospective observational cohort study open to adult employees at the Children's Hospital of Philadelphia, conducted April 20-December 17, 2020. METHODS Employees were recruited starting with high-risk exposure groups, utilizing e-mails, flyers, and announcements at virtual town hall meetings. At baseline, 1 month, 2 months, and 6 months, participants reported occupational and community exposures and gave a blood sample for SARS-CoV-2 antibody measurement by enzyme-linked immunosorbent assays (ELISAs). A post hoc Cox proportional hazards regression model was performed to identify factors associated with increased risk for seropositivity. RESULTS In total, 1,740 employees were enrolled. At 6 months, the cumulative seroprevalence was 5.3%, which was below estimated community point seroprevalence. Seroprevalence was 5.8% among employees who provided direct care and was 3.4% among employees who did not perform direct patient care. Most participants who were seropositive at baseline remained positive at follow-up assessments. In a post hoc analysis, direct patient care (hazard ratio [HR], 1.95; 95% confidence interval [CI], 1.03-3.68), Black race (HR, 2.70; 95% CI, 1.24-5.87), and exposure to a confirmed case in a nonhealthcare setting (HR, 4.32; 95% CI, 2.71-6.88) were associated with statistically significant increased risk for seropositivity. CONCLUSIONS Employee SARS-CoV-2 seroprevalence rates remained below the point-prevalence rates of the surrounding community. Provision of direct patient care, Black race, and exposure to a confirmed case in a nonhealthcare setting conferred increased risk. These data can inform occupational protection measures to maximize protection of employees within the workplace during future COVID-19 waves or other epidemics.
Collapse
Affiliation(s)
- Brian T. Fisher
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anna Sharova
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Craig L. K. Boge
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Audrey Kamrin
- Center for Human Phenomic Science, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jesse Blumenstock
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sydney Shuster
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Lauren Gianchetti
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Danielle Collins
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elikplim Akaho
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Madison E. Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher M. McAllister
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marcus J. Bolton
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Claudia P. Arevalo
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Eileen C. Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth M. Anderson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shannon R. Christensen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Fran Balamuth
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Audrey R. Odom John
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yun Li
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan Coffin
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey S. Gerber
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Anderson EM, Li SH, Awofolaju M, Eilola T, Goodwin E, Bolton MJ, Gouma S, Manzoni TB, Hicks P, Goel RR, Painter MM, Apostolidis SA, Mathew D, Dunbar D, Fiore D, Brock A, Weaver J, Millar JS, DerOhannessian S, Greenplate AR, Frank I, Rader DJ, Wherry EJ, Bates P, Hensley SE. SARS-CoV-2 infections elicit higher levels of original antigenic sin antibodies compared with SARS-CoV-2 mRNA vaccinations. Cell Rep 2022; 41:111496. [PMID: 36261003 PMCID: PMC9578169 DOI: 10.1016/j.celrep.2022.111496] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 07/19/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
It is important to determine if severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and SARS-CoV-2 mRNA vaccinations elicit different types of antibodies. Here, we characterize the magnitude and specificity of SARS-CoV-2 spike-reactive antibodies from 10 acutely infected health care workers with no prior SARS-CoV-2 exposure history and 23 participants who received SARS-CoV-2 mRNA vaccines. We found that infection and primary mRNA vaccination elicit S1- and S2-reactive antibodies, while secondary vaccination boosts mostly S1 antibodies. Using absorption assays, we found that SARS-CoV-2 infections elicit a large proportion of original antigenic sin-like antibodies that bind efficiently to the spike of common seasonal human coronaviruses but poorly to the spike of SARS-CoV-2. In converse, vaccination modestly boosts antibodies reactive to the spike of common seasonal human coronaviruses, and these antibodies cross-react more efficiently to the spike of SARS-CoV-2. Our data indicate that SARS-CoV-2 infections and mRNA vaccinations elicit fundamentally different antibody responses.
Collapse
Affiliation(s)
- Elizabeth M Anderson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuk Hang Li
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Moses Awofolaju
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theresa Eilola
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eileen Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcus J Bolton
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tomaz B Manzoni
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Philip Hicks
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rishi R Goel
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark M Painter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sokratis A Apostolidis
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Rheumatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Debora Dunbar
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Danielle Fiore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amanda Brock
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - JoEllen Weaver
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John S Millar
- Department of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephanie DerOhannessian
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Allison R Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J Rader
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Sharma KS, Sharma R, Nehra S, Rajpurohit NA, Bhakar K, Kumar D. COVID-19: Consequences on pregnant women and neonates. HEALTH SCIENCES REVIEW 2022; 4:100044. [PMID: 35875514 PMCID: PMC9295334 DOI: 10.1016/j.hsr.2022.100044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 11/27/2022]
Abstract
Introduction Human species is confronting with a gigantic global COVID-19 pandemic. Initially, it was observed in Wuhan, China, and the COVID-19 cases spread across the globe with lightning speed and resulted in the 21st century pandemic. If scientific reports are taken care of, it is noteworthy that this virus possesses more specific characteristics due to its structure. The distinctive structure has a higher binding affinity with angiotensin-converting enzyme 2 (ACE2) protein, and this is used as an access point to gain access to hosts. Methods A complete literature search was conducted using PubMed, Google Scholar, SciFinder, and deep-diving Google Search using keywords such as "Pregnancy, COVID-19, Newborn, Fetus, Coronavirus 2019, Neonate, Pregnant women, and vertical transmission". Result and discussion The SARS-CoV-2 virus is unlike its former analogs: SARS-CoV, and MERS-CoV in 2002 and 2012, respectively, or anything mankind has faced earlier concerning viciousness, global spread, and gravity of a causative agent. The current review has delved into articles published in various journals worldwide including the latest studies on the impact of COVID-19 on pregnant women and neonates and has discussed complications and challenges, psychological health, immunological response, vertical transmission, concurrent disorders, vaccine debate, management recommendations, recent news of the approval of COVID-19 vaccine for 6 months and older babies, and future perspectives.
Collapse
|
28
|
Triebwasser JE, Dhudasia MB, Mukhopadhyay S, Flannery DD, Gouma S, Hensley SE, Puopolo KM. Assessment of SARS-CoV-2 serostatus and hypertensive disorders of pregnancy. Am J Obstet Gynecol 2022; 227:664-666.e1. [PMID: 35718110 PMCID: PMC9212257 DOI: 10.1016/j.ajog.2022.06.012] [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: 05/22/2022] [Accepted: 06/12/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Jourdan E Triebwasser
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
| | - Miren B Dhudasia
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Dustin D Flannery
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott E Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Karen M Puopolo
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
29
|
Azami M, Moradi Y, Moradkhani A, Aghaei A. SARS-CoV-2 seroprevalence around the world: an updated systematic review and meta-analysis. Eur J Med Res 2022; 27:81. [PMID: 35655237 PMCID: PMC9160514 DOI: 10.1186/s40001-022-00710-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/16/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Covid-19 has been one of the major concerns around the world in the last 2 years. One of the challenges of this disease has been to determine its prevalence. Conflicting results of the serology test in Covid explored the need for an updated meta-analysis on this issue. Thus, this systematic review aimed to estimate the prevalence of global SARS-CoV-2 serology in different populations and geographical areas. METHODS To identify studies evaluating the seroprevalence of SARS-CoV-2, a comprehensive literature search was performed from international databases, including Medline (PubMed), Web of Sciences, Scopus, EMBASE, and CINHAL. RESULTS In this meta-analysis, the results showed that SARS-CoV-2 seroprevalence is between 3 and 15% worldwide. In Eastern Mediterranean, the pooled estimate of seroprevalence SARS-CoV-2 was 15% (CI 95% 5-29%), and in Africa, the pooled estimate was 6% (CI 95% 1-13%). In America, the pooled estimate was 8% (CI 95% 6-11%), and in Europe, the pooled estimate was 5% (CI 95% 4-6%). Also the last region, Western Pacific, the pooled estimate was 3% (CI 95% 2-4%). Besides, we analyzed three of these areas separately. This analysis estimated the prevalence in subgroups such as study population, diagnostic methods, sampling methods, time, perspective, and type of the study. CONCLUSION The present meta-analysis showed that the seroprevalence of SARS-CoV-2 has been between 3 and 15% worldwide. Even considering the low estimate of this rate and the increasing vaccination in the world, many people are still susceptible to SARS-CoV-2.
Collapse
Affiliation(s)
- Mobin Azami
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Yousef Moradi
- Department of Epidemiology and Biostatistics, Faculty of Medicine, Kurdistan University of Medical Science, Sanandaj, Iran
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Asra Moradkhani
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Abbas Aghaei
- Department of Epidemiology and Biostatistics, Faculty of Medicine, Kurdistan University of Medical Science, Sanandaj, Iran.
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
30
|
Burris HH, Mullin AM, Dhudasia MB, Flannery DD, Mukhopadhyay S, Pfeifer MR, Woodford EC, Briker SM, Triebwasser JE, Morris JS, Montoya-Williams D, Gouma S, Hensley SE, Puopolo KM. Neighborhood Characteristics and Racial Disparities in Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Seropositivity in Pregnancy. Obstet Gynecol 2022; 139:1018-1026. [PMID: 35675599 PMCID: PMC9180815 DOI: 10.1097/aog.0000000000004791] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/03/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To quantify the extent to which neighborhood characteristics contribute to racial and ethnic disparities in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) seropositivity in pregnancy. METHODS This cohort study included pregnant patients who presented for childbirth at two hospitals in Philadelphia, Pennsylvania from April 13 to December 31, 2020. Seropositivity for SARS-CoV-2 was determined by measuring immunoglobulin G and immunoglobulin M antibodies by enzyme-linked immunosorbent assay in discarded maternal serum samples obtained for clinical purposes. Race and ethnicity were self-reported and abstracted from medical records. Patients' residential addresses were geocoded to obtain three Census tract variables: community deprivation, racial segregation (Index of Concentration at the Extremes), and crowding. Multivariable mixed effects logistic regression models and causal mediation analyses were used to quantify the extent to which neighborhood variables may explain racial and ethnic disparities in seropositivity. RESULTS Among 5,991 pregnant patients, 562 (9.4%) were seropositive for SARS-CoV-2. Higher seropositivity rates were observed among Hispanic (19.3%, 104/538) and Black (14.0%, 373/2,658) patients, compared with Asian (3.2%, 13/406) patients, White (2.7%, 57/2,133) patients, and patients of another race or ethnicity (5.9%, 15/256) (P<.001). In adjusted models, per SD increase, deprivation (adjusted odds ratio [aOR] 1.16, 95% CI 1.02-1.32) and crowding (aOR 1.15, 95% CI 1.05-1.26) were associated with seropositivity, but segregation was not (aOR 0.90, 95% CI 0.78-1.04). Mediation analyses revealed that crowded housing may explain 6.7% (95% CI 2.0-14.7%) of the Hispanic-White disparity and that neighborhood deprivation may explain 10.2% (95% CI 0.5-21.1%) of the Black-White disparity. CONCLUSION Neighborhood deprivation and crowding were associated with SARS-CoV-2 seropositivity in pregnancy in the prevaccination era and may partially explain high rates of SARS-CoV-2 seropositivity among Black and Hispanic patients. Investing in structural neighborhood improvements may reduce inequities in viral transmission.
Collapse
Affiliation(s)
- Heather H. Burris
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, USA
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Miren B. Dhudasia
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dustin D. Flannery
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sagori Mukhopadhyay
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Madeline R. Pfeifer
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emily C. Woodford
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sara M. Briker
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Jeffrey S. Morris
- Department of Biostatistics Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Diana Montoya-Williams
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Karen M. Puopolo
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
31
|
Goel RR, Painter MM, Lundgreen KA, Apostolidis SA, Baxter AE, Giles JR, Mathew D, Pattekar A, Reynaldi A, Khoury DS, Gouma S, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Kc W, Oldridge DA, Erickson RI, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Dougherty J, Long S, D'Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, Drapeau EM, Davenport MP, Hensley SE, Bates P, Greenplate AR, Wherry EJ. Efficient recall of Omicron-reactive B cell memory after a third dose of SARS-CoV-2 mRNA vaccine. Cell 2022; 185:1875-1887.e8. [PMID: 35523182 PMCID: PMC8989683 DOI: 10.1016/j.cell.2022.04.009] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/20/2022] [Accepted: 04/06/2022] [Indexed: 01/07/2023]
Abstract
We examined antibody and memory B cell responses longitudinally for ∼9-10 months after primary 2-dose SARS-CoV-2 mRNA vaccination and 3 months after a 3rd dose. Antibody decay stabilized between 6 and 9 months, and antibody quality continued to improve for at least 9 months after 2-dose vaccination. Spike- and RBD-specific memory B cells remained durable over time, and 40%-50% of RBD-specific memory B cells simultaneously bound the Alpha, Beta, Delta, and Omicron variants. Omicron-binding memory B cells were efficiently reactivated by a 3rd dose of wild-type vaccine and correlated with the corresponding increase in neutralizing antibody titers. In contrast, pre-3rd dose antibody titers inversely correlated with the fold-change of antibody boosting, suggesting that high levels of circulating antibodies may limit the added protection afforded by repeat short interval boosting. These data provide insight into the quantity and quality of mRNA-vaccine-induced immunity over time through 3 or more antigen exposures.
Collapse
Affiliation(s)
- Rishi R Goel
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark M Painter
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kendall A Lundgreen
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sokratis A Apostolidis
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Rheumatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amy E Baxter
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Josephine R Giles
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ajinkya Pattekar
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Philip Hicks
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Dysinger
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amanda Hicks
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Harsh Sharma
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Herring
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott Korte
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wumesh Kc
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Derek A Oldridge
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachel I Erickson
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Madison E Weirick
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher M McAllister
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Moses Awofolaju
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Tanenbaum
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeanette Dougherty
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sherea Long
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kurt D'Andrea
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacob T Hamilton
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maura McLaughlin
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justine C Williams
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sharon Adamski
- Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Oliva Kuthuru
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth M Drapeau
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Allison R Greenplate
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Qaseem A, Yost J, Etxeandia-Ikobaltzeta I, Forciea MA, Abraham GM, Miller MC, Obley AJ, Humphrey LL, Akl EA, Andrews R, Dunn A, Haeme R, Kansagara DL, Tschanz MP. What Is the Antibody Response and Role in Conferring Natural Immunity After SARS-CoV-2 Infection? Rapid, Living Practice Points From the American College of Physicians (Version 2). Ann Intern Med 2022; 175:556-565. [PMID: 35073153 PMCID: PMC8803138 DOI: 10.7326/m21-3272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
DESCRIPTION The Scientific Medical Policy Committee (SMPC) of the American College of Physicians (ACP) developed these living, rapid practice points to summarize the current best available evidence on the antibody response to SARS-CoV-2 infection and protection against reinfection with SARS-CoV-2. This is version 2 of the ACP practice points, which serves to update version 1, published on 16 March 2021. These practice points do not evaluate vaccine-acquired immunity or cellular immunity. METHODS The SMPC developed this version of the living, rapid practice points based on an updated living, rapid, systematic review conducted by the Portland VA Research Foundation and funded by the Agency for Healthcare Research and Quality. PRACTICE POINT 1 Do not use SARS-CoV-2 antibody tests for the diagnosis of SARS-CoV-2 infection. PRACTICE POINT 2 Do not use SARS-CoV-2 antibody tests to predict the degree or duration of natural immunity conferred by antibodies against reinfection, including natural immunity against different variants. RETIREMENT FROM LIVING STATUS Although natural immunity remains a topic of scientific interest, this topic is being retired from living status given the availability of effective vaccines for SARS-CoV-2 and widespread recommendations for and prevalence of their use. Currently, vaccination is the best clinical recommendation for preventing infection, reinfection, and serious illness from SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Amir Qaseem
- American College of Physicians, Philadelphia, Pennsylvania (A.Q., I.E.)
| | - Jennifer Yost
- American College of Physicians, Philadelphia, and Villanova University, Villanova, Pennsylvania (J.Y.)
| | | | | | - George M Abraham
- University of Massachusetts Medical School/Saint Vincent Hospital, Worcester, Massachusetts (G.M.A.)
| | | | - Adam J Obley
- Portland Veterans Affairs Medical Center and Oregon Health & Science University, Portland, Oregon (A.J.O., L.L.H.)
| | - Linda L Humphrey
- Portland Veterans Affairs Medical Center and Oregon Health & Science University, Portland, Oregon (A.J.O., L.L.H.)
| | | | | | | | | | | | | |
Collapse
|
33
|
Lederer K, Bettini E, Parvathaneni K, Painter MM, Agarwal D, Lundgreen KA, Weirick M, Muralidharan K, Castaño D, Goel RR, Xu X, Drapeau EM, Gouma S, Ort JT, Awofolaju M, Greenplate AR, Le Coz C, Romberg N, Trofe-Clark J, Malat G, Jones L, Rosen M, Weiskopf D, Sette A, Besharatian B, Kaminiski M, Hensley SE, Bates P, Wherry EJ, Naji A, Bhoj V, Locci M. Germinal center responses to SARS-CoV-2 mRNA vaccines in healthy and immunocompromised individuals. Cell 2022; 185:1008-1024.e15. [PMID: 35202565 PMCID: PMC8808747 DOI: 10.1016/j.cell.2022.01.027] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/13/2021] [Accepted: 01/28/2022] [Indexed: 12/21/2022]
Abstract
Vaccine-mediated immunity often relies on the generation of protective antibodies and memory B cells, which commonly stem from germinal center (GC) reactions. An in-depth comparison of the GC responses elicited by SARS-CoV-2 mRNA vaccines in healthy and immunocompromised individuals has not yet been performed due to the challenge of directly probing human lymph nodes. Herein, through a fine-needle aspiration-based approach, we profiled the immune responses to SARS-CoV-2 mRNA vaccines in lymph nodes of healthy individuals and kidney transplant recipients (KTXs). We found that, unlike healthy subjects, KTXs presented deeply blunted SARS-CoV-2-specific GC B cell responses coupled with severely hindered T follicular helper cell, SARS-CoV-2 receptor binding domain-specific memory B cell, and neutralizing antibody responses. KTXs also displayed reduced SARS-CoV-2-specific CD4 and CD8 T cell frequencies. Broadly, these data indicate impaired GC-derived immunity in immunocompromised individuals and suggest a GC origin for certain humoral and memory B cell responses following mRNA vaccination.
Collapse
Affiliation(s)
- Katlyn Lederer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Bettini
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kalpana Parvathaneni
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark M Painter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Divyansh Agarwal
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kendall A Lundgreen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Madison Weirick
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kavitha Muralidharan
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Diana Castaño
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia, Medellín, Antioquia 050010, Colombia
| | - Rishi R Goel
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Xiaoming Xu
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth M Drapeau
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sigrid Gouma
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jordan T Ort
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Moses Awofolaju
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Allison R Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Carole Le Coz
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Neil Romberg
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jennifer Trofe-Clark
- Department of Medicine, Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory Malat
- Department of Medicine, Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lisa Jones
- Department of Radiology, Division of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark Rosen
- Department of Radiology, Division of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, La Jolla, CA 92093, USA
| | - Behdad Besharatian
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mary Kaminiski
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Ali Naji
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Vijay Bhoj
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Michela Locci
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
34
|
Emeruwa UN, Gyamfi-Bannerman C, Miller RS. Health Care Disparities in the COVID-19 Pandemic in the United States: A Focus on Obstetrics. Clin Obstet Gynecol 2022; 65:123-133. [PMID: 35045035 PMCID: PMC8767922 DOI: 10.1097/grf.0000000000000665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The influence of social determinants of health on disease dynamics and outcomes has become increasingly clear, making them a prime target of investigation and mitigation efforts. The obstetric population is uniquely positioned to provide insight into the health inequities exacerbated by the coronavirus disease 2019 pandemic given their susceptibility to infectious disease morbidity and frequent interactions with the health care system, which provide opportunities for ascertainment of disease incidence and severity. This review summarizes the data on disparities identified in the US obstetric population during the coronavirus disease 2019 pandemic as they relate to race and ethnicity, built environment, insurance status, language, and immigration status.
Collapse
Affiliation(s)
- Ukachi N. Emeruwa
- Division of Maternal- Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital, New York, New York
| | - Cynthia Gyamfi-Bannerman
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego School of Medicine, UC San Diego Health, La Jolla, California
| | - Russell S. Miller
- Division of Maternal- Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital, New York, New York
| |
Collapse
|
35
|
Goel RR, Painter MM, Lundgreen KA, Apostolidis SA, Baxter AE, Giles JR, Mathew D, Pattekar A, Reynaldi A, Khoury DS, Gouma S, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, KC W, Oldridge DA, Erickson RI, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Dougherty J, Long S, D’Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, Drapeau EM, Davenport MP, Hensley SE, Bates P, Greenplate AR, Wherry EJ. Efficient recall of Omicron-reactive B cell memory after a third dose of SARS-CoV-2 mRNA vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.02.20.481163. [PMID: 35233575 PMCID: PMC8887077 DOI: 10.1101/2022.02.20.481163] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Despite a clear role in protective immunity, the durability and quality of antibody and memory B cell responses induced by mRNA vaccination, particularly by a 3 rd dose of vaccine, remains unclear. Here, we examined antibody and memory B cell responses in a cohort of individuals sampled longitudinally for ∼9-10 months after the primary 2-dose mRNA vaccine series, as well as for ∼3 months after a 3 rd mRNA vaccine dose. Notably, antibody decay slowed significantly between 6- and 9-months post-primary vaccination, essentially stabilizing at the time of the 3 rd dose. Antibody quality also continued to improve for at least 9 months after primary 2-dose vaccination. Spike- and RBD-specific memory B cells were stable through 9 months post-vaccination with no evidence of decline over time, and ∼40-50% of RBD-specific memory B cells were capable of simultaneously recognizing the Alpha, Beta, Delta, and Omicron variants. Omicron-binding memory B cells induced by the first 2 doses of mRNA vaccine were boosted significantly by a 3rd dose and the magnitude of this boosting was similar to memory B cells specific for other variants. Pre-3 rd dose memory B cell frequencies correlated with the increase in neutralizing antibody titers after the 3 rd dose. In contrast, pre-3 rd dose antibody titers inversely correlated with the fold-change of antibody boosting, suggesting that high levels of circulating antibodies may limit reactivation of immunological memory and constrain further antibody boosting by mRNA vaccines. These data provide a deeper understanding of how the quantity and quality of antibody and memory B cell responses change over time and number of antigen exposures. These data also provide insight into potential immune dynamics following recall responses to additional vaccine doses or post-vaccination infections. GRAPHICAL SUMMARY
Collapse
Affiliation(s)
- Rishi R. Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Mark M. Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Kendall A. Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Sokratis A. Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Amy E. Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Josephine R. Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales; Sydney, Australia
| | - David S. Khoury
- Kirby Institute, University of New South Wales; Sydney, Australia
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Philip Hicks
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Sarah Dysinger
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Amanda Hicks
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Harsh Sharma
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Sarah Herring
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Scott Korte
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Wumesh KC
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Derek A. Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Rachel I. Erickson
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Madison E. Weirick
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Christopher M. McAllister
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Moses Awofolaju
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Nicole Tanenbaum
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Sherea Long
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Kurt D’Andrea
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Jacob T. Hamilton
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Maura McLaughlin
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Justine C. Williams
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Sharon Adamski
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Elizabeth M. Drapeau
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | | | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Allison R. Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
| | - E. John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USAs
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| |
Collapse
|
36
|
Experiences of New Mothers During the Coronavirus Disease 2019 (COVID-19) Pandemic. Obstet Gynecol 2022; 139:244-253. [DOI: 10.1097/aog.0000000000004660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/05/2021] [Indexed: 11/25/2022]
|
37
|
Prevalence of antibodies against SARS-CoV-2 among pregnant women in Norway during the period December 2019 through December 2020. Epidemiol Infect 2022; 150:e28. [PMID: 35022102 PMCID: PMC8886077 DOI: 10.1017/s0950268822000073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
We studied severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) seroprevalence among pregnant women in Norway by including all women who were first trimester pregnant (n = 6520), each month from December 2019 through December 2020, in the catchment region of Norway's second-largest hospital. We used sera that had been frozen stored after compulsory testing for syphilis antibodies in antenatal care. The sera were analysed with the Elecsys® Anti-SARS-CoV-2 immunoassay (Roche Diagnostics, Cobas e801). This immunoassay detects IgG/IgM against SARS-CoV-2 nucleocapsid antigen. Sera with equivocal or positive test results were retested with the Liaison® SARS-CoV-2 S1/S2 IgG (DiaSorin), which detects IgG against the spike (S)1 and S2 protein on the SARS-CoV-2 virus. In total, 98 women (adjusted prevalence 1.7%) had SARS CoV-2 antibodies. The adjusted seroprevalence increased from 0.3% (1/445) in December 2019 to 5.7% (21/418) in December 2020. Out of the 98 seropositive women, 36 (36.7%) had serological signs of current SARS-CoV-2 infection at the time of serum sampling, and the incidence remained low during the study period. This study suggests that SARS CoV-2 was present in the first half of December 2019, 6 weeks before the first case was recognised in Norway. The low occurrence of SARS-CoV-2 infection during 2020, may be explained by high compliance to extensive preventive measures implemented early in the epidemic.
Collapse
|
38
|
Upchurch DM, Wong MS, Yuan AH, Haderlein TP, McClendon J, Christy A, Washington DL. COVID-19 Infection in the Veterans Health Administration: Gender-specific Racial and Ethnic Differences. Womens Health Issues 2022; 32:41-50. [PMID: 34702652 PMCID: PMC8486675 DOI: 10.1016/j.whi.2021.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/10/2021] [Accepted: 09/27/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Racial/ethnic minoritized groups, women, and economically disadvantaged groups are disproportionately affected by the COVID-19 pandemic. We investigated racial/ethnic differences by gender in correlates of COVID-19 infection among veterans seeking health care services at the Veterans Health Administration. Little is known about gender-specific factors associated with infection among veterans. This study seeks to fill this gap. METHODS The sample was veterans with results from a COVID-19 test (polymerase chain reaction) conducted at Veterans Health Administration facilities between March 1, 2020, and August 5, 2020, and linked to the Centers for Disease Control and Prevention Social Vulnerability Index data (39,223 women and 316,380 men). Bivariate, multivariate logistic, and predicted probability analyses were conducted. All analyses were stratified by gender. RESULTS Similar percentages of women and men tested positive for COVID-19 (9.6% vs. 10.0%). In multivariate analysis, compared with non-Hispanic White women, American Indian/Alaska Native, Black, and Hispanic women all had significantly higher odds of infection. Similar racial/ethnic differences were found for men. Both older men and women (>40 years) had lower odds of infection, but the age cut points differed (40 for women, 55 for men). Men 80 years and older had a higher odds than those aged less than 40 years of age. For men, but not for women, being employed (vs. unemployed) was associated with an increased odds of infection, and having comorbidities was associated with decreased odds. There were significant differences within and across gender-by-race/ethnicity in infection, after adjusting for covariates. CONCLUSIONS American Indian/Alaska Native, Hispanic, and Black women and men veterans are disproportionately impacted by COVID-19 infection. Widespread testing and tracking, education, and outreach regarding COVID-19 mitigation and vaccination efforts are recommended.
Collapse
Affiliation(s)
- Dawn M Upchurch
- Department of Community Health Sciences, UCLA Fielding School of Public Health, Los Angeles, California.
| | - Michelle S Wong
- VA HSR&D Center for the Study of Healthcare Innovation, Implementation & Policy (CSHIIP), VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Anita H Yuan
- VA HSR&D Center for the Study of Healthcare Innovation, Implementation & Policy (CSHIIP), VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Taona P Haderlein
- VA HSR&D Center for the Study of Healthcare Innovation, Implementation & Policy (CSHIIP), VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Juliette McClendon
- National Center for PTSD, Women's Health Science Division, VA Boston Health Care System, Boston, Massachusetts; Department of Psychiatry, Boston University School of Medicine, Boston, Massachusetts
| | - Alicia Christy
- Women's Health Services, Veterans Health Administration, Washington, District of Columbia
| | - Donna L Washington
- VA HSR&D Center for the Study of Healthcare Innovation, Implementation & Policy (CSHIIP), VA Greater Los Angeles Healthcare System, Los Angeles, California; Division of General Internal Medicine and Health Services Research, Department of Medicine, University of California Los Angeles Geffen School of Medicine, Los Angeles, California
| |
Collapse
|
39
|
Lucinde RK, Mugo D, Bottomley C, Karani A, Gardiner E, Aziza R, Gitonga JN, Karanja H, Nyagwange J, Tuju J, Wanjiku P, Nzomo E, Kamuri E, Thuranira K, Agunda S, Nyutu G, Etyang AO, Adetifa IMO, Kagucia E, Uyoga S, Otiende M, Otieno E, Ndwiga L, Agoti CN, Aman RA, Mwangangi M, Amoth P, Kasera K, Nyaguara A, Ng’ang’a W, Ochola LB, Namdala E, Gaunya O, Okuku R, Barasa E, Bejon P, Tsofa B, Ochola-Oyier LI, Warimwe GM, Agweyu A, Scott JAG, Gallagher KE. Sero-surveillance for IgG to SARS-CoV-2 at antenatal care clinics in three Kenyan referral hospitals: Repeated cross-sectional surveys 2020-21. PLoS One 2022; 17:e0265478. [PMID: 36240176 PMCID: PMC9565697 DOI: 10.1371/journal.pone.0265478] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION The high proportion of SARS-CoV-2 infections that have remained undetected presents a challenge to tracking the progress of the pandemic and estimating the extent of population immunity. METHODS We used residual blood samples from women attending antenatal care services at three hospitals in Kenya between August 2020 and October 2021and a validated IgG ELISA for SARS-Cov-2 spike protein and adjusted the results for assay sensitivity and specificity. We fitted a two-component mixture model as an alternative to the threshold analysis to estimate of the proportion of individuals with past SARS-CoV-2 infection. RESULTS We estimated seroprevalence in 2,981 women; 706 in Nairobi, 567 in Busia and 1,708 in Kilifi. By October 2021, 13% of participants were vaccinated (at least one dose) in Nairobi, 2% in Busia. Adjusted seroprevalence rose in all sites; from 50% (95%CI 42-58) in August 2020, to 85% (95%CI 78-92) in October 2021 in Nairobi; from 31% (95%CI 25-37) in May 2021 to 71% (95%CI 64-77) in October 2021 in Busia; and from 1% (95% CI 0-3) in September 2020 to 63% (95% CI 56-69) in October 2021 in Kilifi. Mixture modelling, suggests adjusted cross-sectional prevalence estimates are underestimates; seroprevalence in October 2021 could be 74% in Busia and 72% in Kilifi. CONCLUSIONS There has been substantial, unobserved transmission of SARS-CoV-2 in Nairobi, Busia and Kilifi Counties. Due to the length of time since the beginning of the pandemic, repeated cross-sectional surveys are now difficult to interpret without the use of models to account for antibody waning.
Collapse
Affiliation(s)
- Ruth K. Lucinde
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- * E-mail:
| | - Daisy Mugo
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Christian Bottomley
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Angela Karani
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Rabia Aziza
- School of Life Sciences and the Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research (SBIDER), University of Warwick, Coventry, United Kingdom
| | | | - Henry Karanja
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - James Tuju
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Edward Nzomo
- Kilifi County Hospital, Ministry of Health, Government of Kenya, Nairobi, Kenya
| | - Evans Kamuri
- Kenyatta National Hospital, Ministry of Health, Government of Kenya, Nairobi, Kenya
| | - Kaugiria Thuranira
- Kenyatta National Hospital, Ministry of Health, Government of Kenya, Nairobi, Kenya
| | - Sarah Agunda
- Kenyatta National Hospital, Ministry of Health, Government of Kenya, Nairobi, Kenya
| | - Gideon Nyutu
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Ifedayo M. O. Adetifa
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Sophie Uyoga
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Mark Otiende
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Edward Otieno
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | | | | | - Patrick Amoth
- Ministry of Health, Government of Kenya, Nairobi, Kenya
| | | | - Amek Nyaguara
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Wangari Ng’ang’a
- Presidential Policy and Strategy Unit, The Presidency, Government of Kenya, Nairobi, Kenya
| | | | | | - Oscar Gaunya
- Busia Country Teaching & Referral Hospital, Busia, Kenya
| | - Rosemary Okuku
- Busia Country Teaching & Referral Hospital, Busia, Kenya
| | - Edwine Barasa
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | | | | | - George M. Warimwe
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | | | - J. Anthony G. Scott
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Katherine E. Gallagher
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
40
|
Bar KJ, Shaw PA, Choi GH, Aqui N, Fesnak A, Yang JB, Soto-Calderon H, Grajales L, Starr J, Andronov M, Mastellone M, Amonu C, Feret G, DeMarshall M, Buchanan M, Caturla M, Gordon J, Wanicur A, Monroy MA, Mampe F, Lindemuth E, Gouma S, Mullin AM, Barilla H, Pronina A, Irwin L, Thomas R, Eichinger RA, Demuth F, Luning Prak ET, Pascual JL, Short WR, Elovitz MA, Baron J, Meyer NJ, Degnan KO, Frank I, Hensley SE, Siegel DL, Tebas P. A randomized controlled study of convalescent plasma for individuals hospitalized with COVID-19 pneumonia. J Clin Invest 2021; 131:e155114. [PMID: 34788233 PMCID: PMC8670841 DOI: 10.1172/jci155114] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/01/2021] [Indexed: 11/17/2022] Open
Abstract
BackgroundAntibody-based strategies for COVID-19 have shown promise in prevention and treatment of early disease. COVID-19 convalescent plasma (CCP) has been widely used but results from randomized trials supporting its benefit in hospitalized patients with pneumonia are limited. Here, we assess the efficacy of CCP in severely ill, hospitalized adults with COVID-19 pneumonia.MethodsWe performed a randomized control trial (PennCCP2), with 80 adults hospitalized with COVID-19 pneumonia, comparing up to 2 units of locally sourced CCP plus standard care versus standard care alone. The primary efficacy endpoint was comparison of a clinical severity score. Key secondary outcomes include 14- and 28-day mortality, 14- and 28-day maximum 8-point WHO ordinal score (WHO8) score, duration of supplemental oxygenation or mechanical ventilation, respiratory SARS-CoV-2 RNA, and anti-SARS-CoV-2 antibodies.ResultsEighty hospitalized adults with confirmed COVID-19 pneumonia were enrolled at median day 6 of symptoms and day 1 of hospitalization; 60% were anti-SARS-CoV-2 antibody seronegative. Participants had a median of 3 comorbidities, including risk factors for severe COVID-19 and immunosuppression. CCP treatment was safe and conferred significant benefit by clinical severity score (median [MED] and interquartile range [IQR] 10 [5.5-30] vs. 7 [2.75-12.25], P = 0.037) and 28-day mortality (n = 10, 26% vs. n = 2, 5%; P = 0.013). All other prespecified outcome measures showed weak evidence toward benefit of CCP.ConclusionTwo units of locally sourced CCP administered early in hospitalization to majority seronegative participants conferred a significant benefit in clinical severity score and 28-day mortality. Results suggest CCP may benefit select populations, especially those with comorbidities who are treated early.Trial RegistrationClinicalTrials.gov NCT04397757.FundingUniversity of Pennsylvania.
Collapse
Affiliation(s)
- Katharine J. Bar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pamela A. Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Kaiser Permanente Washington Health Research Group, Seattle, Washington, USA
| | - Grace H. Choi
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicole Aqui
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrew Fesnak
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jasper B. Yang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Kaiser Permanente Washington Health Research Group, Seattle, Washington, USA
| | | | - Lizette Grajales
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Julie Starr
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle Andronov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Miranda Mastellone
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chigozie Amonu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Geoff Feret
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maureen DeMarshall
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marie Buchanan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria Caturla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Gordon
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alan Wanicur
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - M. Alexandra Monroy
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Felicity Mampe
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emily Lindemuth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sigrid Gouma
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anne M. Mullin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Holly Barilla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anastasiya Pronina
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leah Irwin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Raeann Thomas
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Risa A. Eichinger
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Faye Demuth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eline T. Luning Prak
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jose L. Pascual
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William R. Short
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michal A. Elovitz
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jillian Baron
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nuala J. Meyer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathleen O. Degnan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian Frank
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Scott E. Hensley
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donald L. Siegel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pablo Tebas
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Boelig RC, Chaudhury S, Aghai ZH, Oliver E, Manusco F, Berghella V, Bergmann-Leitner E. Comprehensive Serological Profile and Specificity of Maternal and Neonatal Cord Blood SARS CoV-2 Antibodies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.12.06.21267328. [PMID: 34909795 PMCID: PMC8669862 DOI: 10.1101/2021.12.06.21267328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To describe the profile and specificity of maternal and neonatal cord-blood antibody profile in response SARS-CoV-2 virus exposure. METHODS This is a Prospective cohort study of delivering patients at Thomas Jefferson University Hospital from April 2020-February 2021. Primary objective was to describe unique maternal and fetal antibody epitope titers and specificity in those patients with COVID-19 history. Serologic profile assessed with a multiplex platform. Antigens used were: HA-trimer Influenza A (Hong Kong H3), spike trimers for SARS-CoV-2, SARS-CoV-1, MERS-CoV, and betacoronaviruses HKU-1 and OC43, as well as the spike N-terminal domain (NTD), spike receptor binding domain (RBD), and nucleocapsid protein (N; full length) for SARS-CoV-2. RESULTS 112 maternal samples and 101 maternal and cord blood pairs were analyzed. Thirty-seven had a known history of COVID-19 (positive PCR test) in the pregnancy and of those, 17 (47%) were diagnosed with COVID-19 within 30 days of delivery. Fifteen of remaining seventy-six (20%) without a known diagnosis had positive maternal serology. For those with history of COVID-19 we identified robust IgG response in maternal blood to CoV2 nucleocapsid (N), spike (S) full-length and S (RBD) antigens with more modest responses to the S (NTD) antigen. By contrast, the maternal blood IgM response appeared more specific to S (full-length), than N, S (RBD) or S (NTD) epitopes. There were significantly higher maternal and cord blood IgG response not just to CoV2 spike (p < 10 -18 ), but also CoV1 spike (p < 10 -9 ) and MERS spike (p < 10 -8 ). By contrast, maternal IgM responses were more specific to CoV2 (p < 10 -19 ), but to a lesser degree for CoV1 (p < 10 -5 ), and no significant differences for MERS. Maternal and cord-blood IgG were highly correlated for both S and N (R 2 = 0.96 and 0.94). CONCLUSIONS Placental transfer is efficient, with robust N and S responses. Both nucleocapsid and spike antibody responses should be studied for a better understanding of COVID-19 immunity. IgG antibodies are cross reactive with related CoV-1 and MERS spike epitopes while IgM, which cannot cross placenta to provide neonatal passive immunity, is more SARS CoV-2 specific. Neonatal cord blood may have significantly different fine-specificity than maternal blood, despite the high efficiency of IgG transfer.
Collapse
|
42
|
Goel RR, Painter MM, Apostolidis SA, Mathew D, Meng W, Rosenfeld AM, Lundgreen KA, Reynaldi A, Khoury DS, Pattekar A, Gouma S, Kuri-Cervantes L, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Baxter AE, Oldridge DA, Giles JR, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Drapeau EM, Dougherty J, Long S, D’Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, The UPenn COVID Processing Unit ‡, Frank I, Betts MR, Vella LA, Grifoni A, Weiskopf D, Sette A, Hensley SE, Davenport MP, Bates P, Luning Prak ET, Greenplate AR, Wherry EJ. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 2021; 374:abm0829. [PMID: 34648302 PMCID: PMC9284784 DOI: 10.1126/science.abm0829] [Citation(s) in RCA: 621] [Impact Index Per Article: 155.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022]
Abstract
The durability of immune memory after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) messenger RNA (mRNA) vaccination remains unclear. In this study, we longitudinally profiled vaccine responses in SARS-CoV-2–naïve and –recovered individuals for 6 months after vaccination. Antibodies declined from peak levels but remained detectable in most subjects at 6 months. By contrast, mRNA vaccines generated functional memory B cells that increased from 3 to 6 months postvaccination, with the majority of these cells cross-binding the Alpha, Beta, and Delta variants. mRNA vaccination further induced antigen-specific CD4+ and CD8+ T cells, and early CD4+ T cell responses correlated with long-term humoral immunity. Recall responses to vaccination in individuals with preexisting immunity primarily increased antibody levels without substantially altering antibody decay rates. Together, these findings demonstrate robust cellular immune memory to SARS-CoV-2 and its variants for at least 6 months after mRNA vaccination.
Collapse
Affiliation(s)
- Rishi R. Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark M. Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sokratis A. Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Wenzhao Meng
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aaron M. Rosenfeld
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kendall A. Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip Hicks
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Dysinger
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amanda Hicks
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Harsh Sharma
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Herring
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott Korte
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amy E. Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A. Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Josephine R. Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Madison E. Weirick
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christopher M. McAllister
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Moses Awofolaju
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicole Tanenbaum
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth M. Drapeau
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sherea Long
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kurt D’Andrea
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jacob T. Hamilton
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maura McLaughlin
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Justine C. Williams
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sharon Adamski
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - The UPenn COVID Processing Unit‡
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Disease, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Ian Frank
- Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael R. Betts
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura A. Vella
- Division of Infectious Disease, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eline T. Luning Prak
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R. Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E. John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
43
|
Comprehensive Serological Profile and Specificity of Maternal and Neonatal Cord Blood SARS CoV-2 Antibodies. AJOG GLOBAL REPORTS 2021; 2:100046. [PMID: 34961853 PMCID: PMC8697419 DOI: 10.1016/j.xagr.2021.100046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Initial studies on COVID-19 in pregnancy have demonstrated a range of neutralizing activity, but little has been published on the full profile of SARS CoV-2 related antibodies in maternal and cordblood. OBJECTIVE This study aimed to describe the profile and specificity of maternal and neonatal cord blood antibody profiles in response to SARS-CoV-2 virus exposure. STUDY DESIGN This was a prospective cohort study of delivering patients at Thomas Jefferson University Hospital from April 2020 to February 2021. The primary objective was to describe unique maternal and fetal antibody epitope titers and specificity in patients with COVID-19 history. Serologic profile was assessed with a multiplex platform. Antigens used were hemagglutinin trimer influenza A (Hong Kong H3); spike trimers for SARS-CoV-2, SARS-CoV-1, Middle East respiratory syndrome coronavirus, and betacoronaviruses HKU-1 and OC43; and spike N-terminal domain, spike receptor-binding domain, and nucleocapsid protein (full length) for SARS-CoV-2. RESULTS Here, 112 maternal samples and 101 maternal and cord blood pairs were analyzed. Of note, 37 patients had a known history of COVID-19 (positive polymerase chain reaction test) during pregnancy. Of 36 patients, 16 (44%) were diagnosed with COVID-19 within 7 days of delivery. Moreover, 15 of the remaining 76 patients (20%) without a known diagnosis had positive maternal serology. For those with a history of COVID-19, we identified robust immunoglobulin G response in maternal blood to CoV-2 nucleocapsid, spike (full length), and spike (receptor-binding domain) antigens with more modest responses to the spike (N-terminal domain) antigen. In contrast, the maternal blood immunoglobulin M response seemed more specific to spike (full length) epitopes than nucleocapsid, spike (receptor-binding domain), or spike (N-terminal domain) epitopes. There were significantly higher maternal and cord blood immunoglobulin G responses not only to CoV-2 spike (127.1-fold; standard deviation, 2.0; P<.00001) but also to CoV-1 spike (21.1-fold higher; standard deviation, 1.8; P<.00001) and Middle East respiratory syndrome spike (6.9-fold higher; standard deviation, 2.5; P<.00001). In contrast, maternal immunoglobulin M responses were more specific to CoV-2 spike (15.8-fold; standard deviation, 2.1; P<.00001) but less specific to CoV-1 (2.5-fold higher; standard deviation, 0.71; P<.00001) and no significant difference for Middle East respiratory syndrome. Maternal and cord blood immunoglobulin G antibodies were highly correlated for both spike and nucleocapsid (R2=0.96 and 0.94, respectively). CONCLUSION Placental transfer was efficient, with robust nucleocapsid and spike responses. Both nucleocapsid and spike antibody responses should be studied for a better understanding of COVID-19 immunity. Immunoglobulin G antibodies were cross-reactive with related CoV-1 and Middle East respiratory syndrome spike epitopes, whereas immunoglobulin M antibodies, which cannot cross the placenta to provide neonatal passive immunity, were more SARS-CoV-2 specific. Neonatal cord blood may have significantly different fine specificity than maternal blood, despite the high efficiency of immunoglobulin G transfer.
Collapse
|
44
|
Lieberman-Cribbin W, Galanti M, Shaman J. Socioeconomic Disparities in Severe Acute Respiratory Syndrome Coronavirus 2 Serological Testing and Positivity in New York City. Open Forum Infect Dis 2021; 8:ofab534. [PMID: 34877365 PMCID: PMC8643621 DOI: 10.1093/ofid/ofab534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/15/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND We characterized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody test prevalence and positive test prevalence across New York City (NYC) in order to investigate disparities in testing outcomes by race and socioeconomic status (SES). METHODS Serologic data were downloaded from the NYC Coronavirus data repository (August 2020-December 2020). Area-level characteristics for NYC neighborhoods were downloaded from United States census data and a socioeconomic vulnerability index was created. Spatial generalized linear mixed models were performed to examine the association between SES and antibody testing and positivity. RESULTS The proportion of Hispanic population (posterior median, 0.001 [95% credible interval, 0.0003-0.002]), healthcare workers (0.003 [0.0001-0.006]), essential workers (0.003 [0.001-0.005]), age ≥65 years (0.003 [0.00002-0.006]), and high SES (SES quartile 3 vs 1: 0.034 [0.003-0.062]) were positively associated with antibody tests per 100000 residents. The White proportion (-0.002 [-0.003 to -0.001]), SES index (quartile 3 vs 1, -0.068 [-0.115 to -0.017]; quartile 4 vs 1, -0.077 [-0.134 to -0.018]) and age ≥65 years (-0.005 [-0.009 to -0.002]) were inversely associated with positive test prevalence (%), whereas the Hispanic (0.004 [0.002-0.006]) and essential worker (0.008 [0.003-0.012]) proportions had positive coefficients. CONCLUSIONS Disparities in serologic testing and seropositivity exist on SES and race/ethnicity across NYC, indicative of excess coronavirus disease burden in vulnerable and marginalized populations.
Collapse
Affiliation(s)
- Wil Lieberman-Cribbin
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Marta Galanti
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Jeffrey Shaman
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| |
Collapse
|
45
|
Sun L, Surya S, Goodman NG, Le AN, Kelly G, Owoyemi O, Desai H, Zheng C, DeLuca S, Good ML, Hussain J, Jeffries SD, Kry YR, Kugler EM, Mansour M, Ndicu J, Osei-Akoto A, Prior T, Pundock SL, Varughese LA, Weaver J, Doucette A, Dudek S, Verma SS, Gouma S, Weirick ME, McAllister CM, Bange E, Gabriel P, Ritchie M, Rader DJ, Vonderheide RH, Schuchter LM, Verma A, Maillard I, Mamtani R, Hensley SE, Gross R, Wileyto EP, Huang AC, Maxwell KN, DeMichele A. SARS-CoV-2 Seropositivity and Seroconversion in Patients Undergoing Active Cancer-Directed Therapy. JCO Oncol Pract 2021; 17:e1879-e1886. [PMID: 34133219 PMCID: PMC8677966 DOI: 10.1200/op.21.00113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Multiple studies have demonstrated the negative impact of cancer care delays during the COVID-19 pandemic, and transmission mitigation techniques are imperative for continued cancer care delivery. We aimed to gauge the effectiveness of these measures at the University of Pennsylvania. METHODS We conducted a longitudinal study of SARS-CoV-2 antibody seropositivity and seroconversion in patients presenting to infusion centers for cancer-directed therapy between May 21, 2020, and October 8, 2020. Participants completed questionnaires and had up to five serial blood collections. RESULTS Of 124 enrolled patients, only two (1.6%) had detectable SARS-CoV-2 antibodies on initial blood draw, and no initially seronegative patients developed newly detectable antibodies on subsequent blood draw(s), corresponding to a seroconversion rate of 0% (95% CI, 0.0 TO 4.1%) over 14.8 person-years of follow up, with a median of 13 health care visits per patient. CONCLUSION These results suggest that patients with cancer receiving in-person care at a facility with aggressive mitigation efforts have an extremely low likelihood of COVID-19 infection.
Collapse
Affiliation(s)
- Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Sanjna Surya
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Noah G. Goodman
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Anh N. Le
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Gregory Kelly
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Olutosin Owoyemi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Heena Desai
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Cathy Zheng
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Shannon DeLuca
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Madeline L. Good
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Jasmin Hussain
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Seth D. Jeffries
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Yolanda R. Kry
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Emily M. Kugler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Maikel Mansour
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - John Ndicu
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - AnnaClaire Osei-Akoto
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Timothy Prior
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Stacy L. Pundock
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Lisa A. Varughese
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - JoEllen Weaver
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Abigail Doucette
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Scott Dudek
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Shefali Setia Verma
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | - Madison E. Weirick
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | | | - Erin Bange
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Peter Gabriel
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Marylyn Ritchie
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Daniel J. Rader
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Robert H. Vonderheide
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Lynn M. Schuchter
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Anurag Verma
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Ronac Mamtani
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | - Robert Gross
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - E. Paul Wileyto
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Alexander C. Huang
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Kara N. Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Angela DeMichele
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA,Angela DeMichele, MD, MSCE, Division of Hematology/Oncology, Department of Medicine, 3400 Civic Center Blvd, PCAM 10-South, Philadelphia, PA 19104; e-mail:
| |
Collapse
|
46
|
Assefa N, Regassa LD, Teklemariam Z, Oundo J, Madrid L, Dessie Y, Scott J. Seroprevalence of anti-SARS-CoV-2 antibodies in women attending antenatal care in eastern Ethiopia: a facility-based surveillance. BMJ Open 2021; 11:e055834. [PMID: 34819290 PMCID: PMC8613670 DOI: 10.1136/bmjopen-2021-055834] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/01/2021] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE We conducted serosurveillance of anti-SARS-CoV-2 antibodies among pregnant women attending their first antenatal care. SETTING The surveillance was set in one referral hospital in Harar, one district hospital and one health centre located in Haramaya district in rural eastern Ethiopia. PARTICIPANTS We collected questionnaire data and a blood sample from 3312 pregnant women between 1 April 2020 and 31 March 2021. We selected 1447 blood samples at random and assayed these for anti-SARS-CoV-2 antibodies at Hararghe Health Research laboratory using WANTAI SARS-CoV-2 Rapid Test for total immunoglobulin. OUTCOME We assayed for anti-SARS-CoV-2 antibodies and temporal trends in seroprevalence were analysed with a χ2 test for trend and multivariable binomial regression. RESULTS Among 1447 sera tested, 83 were positive for anti-SARS-CoV-2 antibodies giving a crude seroprevalence of 5.7% (95% CI 4.6% to 7.0%). Of 160 samples tested in April-May 2020, none was seropositive; the first seropositive sample was identified in June and seroprevalence rose steadily thereafter (χ2 test for trend, p=0.003) reaching a peak of 11.8% in February 2021. In the multivariable model, seroprevalence was approximately 3% higher in first-trimester mothers compared with later presentations, and rose by 0.75% (95% CI 0.31% to 1.20%) per month of calendar time. CONCLUSIONS This clinical convenience sample illustrates the dynamic of the SARS-CoV-2 epidemic in pregnant women in eastern Ethiopia; infection was rare before June 2020 but it spread in a linear fashion thereafter, rather than following intermittent waves, and reached 10% by the beginning of 2021. After 1 year of surveillance, most pregnant mothers remained susceptible.
Collapse
Affiliation(s)
- Nega Assefa
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
- London School of Hygiene & Tropical Medicine, London, UK
| | | | - Zelalem Teklemariam
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Joseph Oundo
- London School of Hygiene & Tropical Medicine, London, UK
| | - Lola Madrid
- London School of Hygiene & Tropical Medicine, London, UK
| | - Yadeta Dessie
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Jag Scott
- London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
47
|
Gonik CO, Alonso AM, Gonik B. SARS-CoV-2 Seroprevalence in Florida Department of Health in Palm Beach County Obstetric Clinics: A Cross-Sectional Study during the First Pandemic Surge. Am J Perinatol 2021; 40:912-916. [PMID: 34758496 DOI: 10.1055/s-0041-1739353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Estimating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) seroprevalence is an important part of the public health approach to coronavirus disease 2019 (COVID-19) understanding and containment. This is particularly relevant to an obstetric population because of implications in the management of the pregnant host, care of the newborn, and disease progression within the community. STUDY DESIGN A cross-sectional seroprevalence study was performed in four Department of Health Palm Beach County clinics from June 29, 2020, to August 5, 2020. Samples were collected from asymptomatic antepartum and postpartum participants. A web-based surveillance system was used to identify subsequent antibody or polymerase chain reaction (PCR) testing encounters. RESULTS A total of 163 of 618 subjects were seropositive (26.4%). Racial makeup was white 2.5%, black 19.0%, and Hispanic 78.5%. Positive serology was seen in 16.0, 35.6, and 30.1% of first, second, and third trimesters, respectively; 18.4% were positive postpartum. Only four patients voluntarily reported PCR positivity prior to antibody testing. Six home zip codes accounted for the majority (68.1%) of positive results. Thirty-two patients had repeat serology (65.6% positive and 34.4% negative). Of the 163 subjects, 65 underwent later PCR testing with 92% negative for SAR-CoV-2. CONCLUSION Almost one in four subjects had serologic evidence of previous SARS-CoV-2 infection. These very high seroprevalence rates have not been previously reported and highlight the concern for health disparities in the United States. Most were asymptomatic and without a history for SARS-CoV-2 exposure. There was a loss of seropositivity in a significant number of subjects, raising concern for risk of reinfection, inadequate transplacental antibody transfer, and subsequent limited passive protection to the newborn. These seroprevalence data will also allow for better newborn follow-up of unanticipated consequences of COVID-19 infection in pregnancy. KEY POINTS · SAR-CoV-2 seroprevalence is disproportionately high in this obstetric population. · The majority of subjects who tested positive for SAR-CoV-2 were asymptomatic for COVID-19 infection. · Estimating seroprevalence is an important public health approach to disease surveillance and understanding.
Collapse
Affiliation(s)
- Charles O Gonik
- Florida Department of Health, Palm Beach County, Delray Beach, Florida
| | - Alina M Alonso
- Florida Department of Health, Palm Beach County, Delray Beach, Florida
| | - Bernard Gonik
- Department of Obstetrics and Gynecology, Maternal-Fetal Medicine, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
48
|
Flamand C, Alves Sarmento C, Enfissi A, Bailly S, Beillard E, Gaillet M, Michaud C, Servas V, Clement N, Perilhou A, Carage T, Musso D, Carod JF, Eustache S, Tourbillon C, Boizon E, James S, Djossou F, Salje H, Cauchemez S, Rousset D. Seroprevalence of anti-SARS-CoV-2 IgG at the first epidemic peak in French Guiana, July 2020. PLoS Negl Trop Dis 2021; 15:e0009945. [PMID: 34767549 PMCID: PMC8639096 DOI: 10.1371/journal.pntd.0009945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/02/2021] [Accepted: 10/22/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND While Latin America has been heavily affected by the pandemic, only a few seroprevalence studies have been conducted there during the first epidemic wave in the first half of 2020. METHODOLOGY/PRINCIPAL FINDINGS A cross-sectional survey was performed between 15 July 2020 and 23 July 2020 among individuals who visited 4 medical laboratories or 5 health centers for routine screening or clinical management, with the exception of symptomatic suggestive cases of covid-19. Samples were screened for the presence of anti-SARS-CoV-2 IgG directed against domain S1 of the SARS-CoV-2 spike protein using the anti-SARS-CoV-2 enzyme-linked immunosorbent assay (ELISA) from Euroimmun. CONCLUSIONS/SIGNIFICANCE The overall seroprevalence was 15.4% [9.3%-24.4%] among 480 participants, ranging from 4.0% to 25.5% across the different municipalities. The seroprevalence did not differ according to gender (p = 0.19) or age (p = 0.51). Among SARS-CoV-2 positive individuals, we found that 24.6% [11.5%-45.2%] reported symptoms consistent with COVID-19. Our findings revealed high levels of infection across the territory but a low number of resulting deaths, which can be explained by French Guiana's young population structure.
Collapse
Affiliation(s)
- Claude Flamand
- Epidemiology unit, Institut Pasteur in French Guiana, Cayenne, French Guiana
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, UMR2000, CNRS, Paris, France
| | | | - Antoine Enfissi
- Laboratory of Virology, Institut Pasteur in French Guiana, Cayenne, French Guiana
| | - Sarah Bailly
- Epidemiology unit, Institut Pasteur in French Guiana, Cayenne, French Guiana
| | - Emmanuel Beillard
- Medical Biology Laboratory, Institut Pasteur in French Guiana, Cayenne, French Guiana
| | - Mélanie Gaillet
- Health Centers Department, Cayenne Hospital Center, Cayenne, French Guiana
| | - Céline Michaud
- Health Centers Department, Cayenne Hospital Center, Cayenne, French Guiana
| | - Véronique Servas
- Health Centers Department, Cayenne Hospital Center, Cayenne, French Guiana
| | - Nathalie Clement
- Clinical Core of the Center for Translational Sciences, Institut Pasteur, Paris, France
| | - Anaïs Perilhou
- Clinical Core of the Center for Translational Sciences, Institut Pasteur, Paris, France
| | - Thierry Carage
- Carage Medical Biology Laboratory, Kourou, French Guiana
| | - Didier Musso
- Laboratoires Eurofins Labazur Guyane, Remire, French Guiana
- Aix Marseille University, IRD, AP-HM, SSA, VITROME, IHU-Méditerranée Infection, Marseille, France
| | - Jean-françois Carod
- Medical Biology laboratory, Centre Hospitalier de l’Ouest Guyanais, Saint-Laurent du Maroni, French Guiana
| | - Stéphanie Eustache
- Epidemiology unit, Institut Pasteur in French Guiana, Cayenne, French Guiana
| | - Céline Tourbillon
- Epidemiology unit, Institut Pasteur in French Guiana, Cayenne, French Guiana
| | - Elodie Boizon
- Epidemiology unit, Institut Pasteur in French Guiana, Cayenne, French Guiana
| | - Samantha James
- Epidemiology unit, Institut Pasteur in French Guiana, Cayenne, French Guiana
| | - Félix Djossou
- Infectious and Tropical Diseases Unit, Cayenne Hospital Center, Cayenne, French Guiana
| | - Henrik Salje
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, UMR2000, CNRS, Paris, France
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Simon Cauchemez
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, UMR2000, CNRS, Paris, France
| | - Dominique Rousset
- Laboratory of Virology, Institut Pasteur in French Guiana, Cayenne, French Guiana
| |
Collapse
|
49
|
Apostolidis SA, Kakara M, Painter MM, Goel RR, Mathew D, Lenzi K, Rezk A, Patterson KR, Espinoza DA, Kadri JC, Markowitz DM, E Markowitz C, Mexhitaj I, Jacobs D, Babb A, Betts MR, Prak ETL, Weiskopf D, Grifoni A, Lundgreen KA, Gouma S, Sette A, Bates P, Hensley SE, Greenplate AR, Wherry EJ, Li R, Bar-Or A. Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. Nat Med 2021; 27:1990-2001. [PMID: 34522051 PMCID: PMC8604727 DOI: 10.1038/s41591-021-01507-2] [Citation(s) in RCA: 386] [Impact Index Per Article: 96.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023]
Abstract
SARS-CoV-2 messenger RNA vaccination in healthy individuals generates immune protection against COVID-19. However, little is known about SARS-CoV-2 mRNA vaccine-induced responses in immunosuppressed patients. We investigated induction of antigen-specific antibody, B cell and T cell responses longitudinally in patients with multiple sclerosis (MS) on anti-CD20 antibody monotherapy (n = 20) compared with healthy controls (n = 10) after BNT162b2 or mRNA-1273 mRNA vaccination. Treatment with anti-CD20 monoclonal antibody (aCD20) significantly reduced spike-specific and receptor-binding domain (RBD)-specific antibody and memory B cell responses in most patients, an effect ameliorated with longer duration from last aCD20 treatment and extent of B cell reconstitution. By contrast, all patients with MS treated with aCD20 generated antigen-specific CD4 and CD8 T cell responses after vaccination. Treatment with aCD20 skewed responses, compromising circulating follicular helper T (TFH) cell responses and augmenting CD8 T cell induction, while preserving type 1 helper T (TH1) cell priming. Patients with MS treated with aCD20 lacking anti-RBD IgG had the most severe defect in circulating TFH responses and more robust CD8 T cell responses. These data define the nature of the SARS-CoV-2 vaccine-induced immune landscape in aCD20-treated patients and provide insights into coordinated mRNA vaccine-induced immune responses in humans. Our findings have implications for clinical decision-making and public health policy for immunosuppressed patients including those treated with aCD20.
Collapse
Grants
- U19AI082630 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- T32 AR076951 NIAMS NIH HHS
- AI082630 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- R21 AI142638 NIAID NIH HHS
- AI108545 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- R01 AI152236 NIAID NIH HHS
- 75N9301900065 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- AI149680 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- T32 CA009140 NCI NIH HHS
- R01 AI118694 NIAID NIH HHS
- U19 AI082630 NIAID NIH HHS
- AI152236 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- P30-AI0450080 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- T32 AR076951-01 U.S. Department of Health & Human Services | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
- R01 AI105343 NIAID NIH HHS
- AI105343 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- R01 AI155577 NIAID NIH HHS
- UM1 AI144288 NIAID NIH HHS
- U19 AI149680 NIAID NIH HHS
- AI155577 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- SI-2011-37160 National Multiple Sclerosis Society (National MS Society)
- UC4 DK112217 NIDDK NIH HHS
- P01 AI108545 NIAID NIH HHS
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (Division of Intramural Research of the NIAID)
- Penn | Perelman School of Medicine, University of Pennsylvania (Perelman School of Medicine at the University of Pennsylvania)
Collapse
Affiliation(s)
- Sokratis A Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mihir Kakara
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark M Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rishi R Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kerry Lenzi
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ayman Rezk
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kristina R Patterson
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Diego A Espinoza
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessy C Kadri
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel M Markowitz
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Clyde E Markowitz
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ina Mexhitaj
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Dina Jacobs
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison Babb
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael R Betts
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kendall A Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Penn Center for Research on Coronavirus and Other Emerging Pathogens, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sigrid Gouma
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA, USA
| | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Penn Center for Research on Coronavirus and Other Emerging Pathogens, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott E Hensley
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Rui Li
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Samore MH, Looney A, Orleans B, Greene T, Seegert N, Delgado JC, Presson A, Zhang C, Ying J, Zhang Y, Shen J, Slev P, Gaulin M, Yang MJ, Pavia AT, Alder SC. Probability-Based Estimates of Severe Acute Respiratory Syndrome Coronavirus 2 Seroprevalence and Detection Fraction, Utah, USA. Emerg Infect Dis 2021; 27:2786-2794. [PMID: 34469285 PMCID: PMC8544980 DOI: 10.3201/eid2711.204435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We aimed to generate an unbiased estimate of the incidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in 4 urban counties in Utah, USA. We used a multistage sampling design to randomly select community-representative participants >12 years of age. During May 4-June 30, 2020, we collected serum samples and survey responses from 8,108 persons belonging to 5,125 households. We used a qualitative chemiluminescent microparticle immunoassay to detect SARS-CoV-2 IgG in serum samples. We estimated the overall seroprevalence to be 0.8%. The estimated seroprevalence-to-case count ratio was 2.5, corresponding to a detection fraction of 40%. Only 0.2% of participants from whom we collected nasopharyngeal swab samples had SARS-CoV-2-positive reverse transcription PCR results. SARS-CoV-2 antibody prevalence during the study was low, and prevalence of PCR-positive cases was even lower. The comparatively high SARS-CoV-2 detection rate (40%) demonstrates the effectiveness of Utah's testing strategy and public health response.
Collapse
|