51
|
Health Disparities and Their Effects on Children and Their Caregivers During the Coronavirus Disease 2019 Pandemic. Pediatr Clin North Am 2021; 68:1133-1145. [PMID: 34538304 PMCID: PMC8445754 DOI: 10.1016/j.pcl.2021.05.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has uncovered long-standing health disparities in marginalized communities, including racial and ethnic minorities and children with underlying medical and social problems. African Americans, Hispanics, and Native Americans have higher rates of COVID-19 infections and deaths than their population percentages in the United States. Unique populations of children, including children with developmental disabilities, children in the foster care system, children with chronic medical problems, and children who are homeless are particularly vulnerable to COVID-19 infection. This article explores how the COVID-19 pandemic superimposed on health disparities directly and indirectly affects children, adolescents, and their caregivers.
Collapse
|
52
|
Lederer K, Parvathaneni K, Painter MM, Bettini E, Agarwal D, Lundgreen KA, Weirick M, Goel RR, Xu X, Drapeau EM, Gouma S, Greenplate AR, Coz CL, Romberg N, Jones L, Rosen M, Besharatian B, Kaminiski M, Weiskopf D, Sette A, 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. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34580676 DOI: 10.1101/2021.09.16.21263686] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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. In this study, 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 (KTX) recipients. We found that, unlike healthy subjects, KTX recipients presented deeply blunted SARS-CoV-2-specific GC B cell responses coupled with severely hindered T follicular helper cells, SARS-CoV-2 receptor-binding-domain-specific memory B cells and neutralizing antibodies. KTX recipients 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
|
53
|
Glennon K, Donnelly J, Knowles S, McAuliffe FM, O’Reilly A, Corcoran S, Walsh J, McMorrow R, Higgins T, Bolger L, Clinton S, O’Riordan S, Start A, Roche D, Bartels H, Malone C, McAuley K, McDermott A, Inzitari R, O’Donnell CPF, Malone F, Higgins S, De Gascun C, Doran P, Brennan DJ. Immunological assessment of SARS-CoV-2 infection in pregnancy from diagnosis to delivery: A multicentre prospective study. PLoS One 2021; 16:e0253090. [PMID: 34543278 PMCID: PMC8451988 DOI: 10.1371/journal.pone.0253090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Background Population-based data on SARS-CoV-2 infection in pregnancy and assessment of passive immunity to the neonate, is lacking. We profiled the maternal and fetal response using a combination of viral RNA from naso-pharyngeal swabs and serological assessment of antibodies against SARS-CoV-2. METHODS This multicentre prospective observational study was conducted between March 24th and August 31st 2020. Two independent cohorts were established, a symptomatic SARS-CoV-2 cohort and a cohort of asymptomatic pregnant women attending two of the largest maternity hospitals in Europe. Symptomatic women were invited to provide a serum sample to assess antibody responses. Asymptomatic pregnant women provided a nasopharyngeal swab and serum sample. RT-PCR for viral RNA was performed using the Cobas SARS-CoV-2 6800 platform (Roche). Umbilical cord bloods were obtained at delivery. Maternal and fetal serological response was measured using both the Elecsys® Anti-SARS-CoV-2 immunoassay (Roche), Abbott SARS-CoV-2 IgG Assay and the IgM Architect assay. Informed written consent was obtained from all participants. RESULTS Ten of twenty three symptomatic women had SARS-CoV-2 RNA detected on nasopharyngeal swabs. Five (5/23, 21.7%) demonstrated serological evidence of anti-SARS-CoV-2 IgG antibodies and seven (30.4%, 7/23) were positive for IgM antibodies. In the asymptomatic cohort, the prevalence of SARS-CoV-2 infection in RNA was 0.16% (1/608). IgG SARS-CoV-2 antibodies were detected in 1·67% (10/598, 95% CI 0·8%-3·1%) and IgM in 3·51% (21/598, 95% CI 2·3-5·5%). Nine women had repeat testing post the baseline test. Four (4/9, 44%) remained IgM positive and one remained IgG positive. 3 IgG anti-SARS-CoV-2 antibodies were detectable in cord bloods from babies born to five seropositive women who delivered during the study. The mean gestation at serological test was 34 weeks. The mean time between maternal serologic positivity and detection in umbilical cord samples was 28 days. CONCLUSION Using two independent serological assays, we present a comprehensive illustration of the antibody response to SARS-CoV-2 in pregnancy, and show a low prevalence of asymptomatic SARS-CoV2. Transplacental migration of anti-SARS-CoV-2 antibodies was identified in cord blood of women who demonstrated antenatal anti-SARS-CoV-2 antibodies, raising the possibility of passive immunity.
Collapse
Affiliation(s)
- Kate Glennon
- UCD School of Medicine, National Maternity Hospital, Dublin, Ireland
| | | | - Susan Knowles
- Department of Microbiology, National Maternity Hospital, Dublin, Ireland
| | - Fionnuala M. McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Alma O’Reilly
- RCSI School of Medicine, Rotunda Hospital, Dublin, Ireland
| | | | | | | | | | - Lucy Bolger
- National Maternity Hospital, Dublin, Ireland
| | | | | | - Alexander Start
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | | | | | | | - Karl McAuley
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Anthony McDermott
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Rosanna Inzitari
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Colm P. F. O’Donnell
- Neonatal Unit, UCD School of Medicine National Maternity Hospital, Dublin, Ireland
| | - Fergal Malone
- RCSI School of Medicine, Rotunda Hospital, Dublin, Ireland
| | - Shane Higgins
- UCD School of Medicine, National Maternity Hospital, Dublin, Ireland
- National Maternity Hospital, Dublin, Ireland
| | - Cillian De Gascun
- National Virus Reference Laboratory, University College Dublin, Dublin, Ireland
| | - Peter Doran
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Donal J. Brennan
- UCD School of Medicine, National Maternity Hospital, Dublin, Ireland
- Systems Biology Ireland, UCD School of Medicine, Dublin, Ireland
| |
Collapse
|
54
|
Sherer ML, Lei J, Creisher PS, Jang M, Reddy R, Voegtline K, Olson S, Littlefield K, Park HS, Ursin RL, Ganesan A, Boyer T, Elsayed N, Brown DM, Walch SN, Antar AAR, Manabe YC, Jones-Beatty K, Golden WC, Satin AJ, Sheffield JS, Pekosz A, Klein SL, Burd I. Pregnancy alters interleukin-1 beta expression and antiviral antibody responses during severe acute respiratory syndrome coronavirus 2 infection. Am J Obstet Gynecol 2021; 225:301.e1-301.e14. [PMID: 33798476 PMCID: PMC8008823 DOI: 10.1016/j.ajog.2021.03.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2, the disease-causing pathogen of the coronavirus disease 2019 pandemic, has resulted in morbidity and mortality worldwide. Pregnant women are more susceptible to severe coronavirus disease 2019 and are at higher risk of preterm birth than uninfected pregnant women. Despite this evidence, the immunologic effects of severe acute respiratory syndrome coronavirus 2 infection during pregnancy remain understudied. OBJECTIVE This study aimed to assess the impact of severe acute respiratory syndrome coronavirus 2 infection during pregnancy on inflammatory and humoral responses in maternal and fetal samples and compare antibody responses to severe acute respiratory syndrome coronavirus 2 among pregnant and nonpregnant women. STUDY DESIGN Immune responses to severe acute respiratory syndrome coronavirus 2 were analyzed using samples from pregnant (n=33) and nonpregnant (n=17) women who tested either positive (pregnant, 22; nonpregnant, 17) or negative for severe acute respiratory syndrome coronavirus 2 (pregnant, 11) at Johns Hopkins Hospital. We measured proinflammatory and placental cytokine messenger RNAs, neonatal Fc receptor expression, and tetanus antibody transfer in maternal and cord blood samples. In addition, we evaluated antispike immunoglobulin G, antispike receptor-binding domain immunoglobulin G, and neutralizing antibody responses to severe acute respiratory syndrome coronavirus 2 in serum or plasma collected from nonpregnant women, pregnant women, and cord blood. RESULTS Pregnant women with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 infection expressed more interleukin-1 beta, but not interleukin 6, in blood samples collected within 14 days vs >14 days after performing severe acute respiratory syndrome coronavirus 2 test. Pregnant women with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 infection also had reduced antispike receptor-binding domain immunoglobulin G titers and were less likely to have detectable neutralizing antibody than nonpregnant women. Although severe acute respiratory syndrome coronavirus 2 infection did not disrupt neonatal Fc receptor expression in the placenta, maternal transfer of severe acute respiratory syndrome coronavirus 2 neutralizing antibody was inhibited by infection during pregnancy. CONCLUSION Severe acute respiratory syndrome coronavirus 2 infection during pregnancy was characterized by placental inflammation and reduced antiviral antibody responses, which may impact the efficacy of coronavirus disease 2019 treatment in pregnancy. In addition, the long-term implications of placental inflammation for neonatal health require greater consideration.
Collapse
Affiliation(s)
- Morgan L Sherer
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jun Lei
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Patrick S Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Minyoung Jang
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ramya Reddy
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kristin Voegtline
- Division of General Pediatrics, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD; Biostatistics, Data Management and Epidemiology Core, Johns Hopkins School of Medicine, Baltimore, MD
| | - Sarah Olson
- Biostatistics, Data Management and Epidemiology Core, Johns Hopkins School of Medicine, Baltimore, MD
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Han-Sol Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Rebecca L Ursin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore MD
| | - Abhinaya Ganesan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Theresa Boyer
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nada Elsayed
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Diane M Brown
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Samantha N Walch
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Annukka A R Antar
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yukari C Manabe
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kimberly Jones-Beatty
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - William Christopher Golden
- Eudowood Neonatal Pulmonary Division, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew J Satin
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jeanne S Sheffield
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Irina Burd
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
55
|
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, 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 Vaccination Induces Durable Immune Memory to SARS-CoV-2 with Continued Evolution to Variants of Concern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.08.23.457229. [PMID: 34462751 PMCID: PMC8404899 DOI: 10.1101/2021.08.23.457229] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SARS-CoV-2 mRNA vaccines have shown remarkable efficacy, especially in preventing severe illness and hospitalization. However, the emergence of several variants of concern and reports of declining antibody levels have raised uncertainty about the durability of immune memory following vaccination. In this study, we longitudinally profiled both antibody and cellular immune responses in SARS-CoV-2 naïve and recovered individuals from pre-vaccine baseline to 6 months post-mRNA vaccination. Antibody and neutralizing titers decayed from peak levels but remained detectable in all subjects at 6 months post-vaccination. Functional memory B cell responses, including those specific for the receptor binding domain (RBD) of the Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2) variants, were also efficiently generated by mRNA vaccination and continued to increase in frequency between 3 and 6 months post-vaccination. Notably, most memory B cells induced by mRNA vaccines were capable of cross-binding variants of concern, and B cell receptor sequencing revealed significantly more hypermutation in these RBD variant-binding clones compared to clones that exclusively bound wild-type RBD. Moreover, the percent of variant cross-binding memory B cells was higher in vaccinees than individuals who recovered from mild COVID-19. mRNA vaccination also generated antigen-specific CD8+ T cells and durable memory CD4+ T cells in most individuals, with early CD4+ T cell responses correlating with humoral immunity at later timepoints. These findings demonstrate robust, multi-component humoral and cellular immune memory to SARS-CoV-2 and current variants of concern for at least 6 months after mRNA vaccination. Finally, we observed that boosting of pre-existing immunity with mRNA vaccination in SARS-CoV-2 recovered individuals primarily increased antibody responses in the short-term without significantly altering antibody decay rates or long-term B and T cell memory. Together, this study provides insights into the generation and evolution of vaccine-induced immunity to SARS-CoV-2, including variants of concern, and has implications for future booster strategies.
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
| | - 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
| | - 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
| | - 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, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Ajinkya Pattekar
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USAs
| | - 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, 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
| | - 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, 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
| | - 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
| | - 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
| | - 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, 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
|
56
|
Gouma S, Weirick ME, Bolton MJ, Arevalo CP, Goodwin EC, Anderson EM, McAllister CM, Christensen SR, Dunbar D, Fiore D, Brock A, Weaver J, Millar J, DerOhannessian S, Unit TUPCOVIDP, Frank I, Rader DJ, Wherry EJ, Hensley SE. Health care worker seromonitoring reveals complex relationships between common coronavirus antibodies and COVID-19 symptom duration. JCI Insight 2021; 6:150449. [PMID: 34237028 PMCID: PMC8410018 DOI: 10.1172/jci.insight.150449] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Some studies suggest that recent common coronavirus (CCV) infections are associated with reduced COVID-19 severity upon SARS-CoV-2 infection. We completed serological assays using samples collected from health care workers to identify antibody types associated with SARS-CoV-2 protection and COVID-19 symptom duration. Rare SARS-CoV-2 cross-reactive antibodies elicited by past CCV infections were not associated with protection; however, the duration of symptoms following SARS-CoV-2 infections was significantly reduced in individuals with higher common betacoronavirus (βCoV) antibody titers. Since antibody titers decline over time after CCV infections, individuals in our cohort with higher βCoV antibody titers were more likely recently infected with common βCoVs compared with individuals with lower antibody titers. Therefore, our data suggest that recent βCoV infections potentially limit the duration of symptoms following SARS-CoV-2 infections through mechanisms that do not involve cross-reactive antibodies. Our data are consistent with the emerging hypothesis that cellular immune responses elicited by recent common βCoV infections transiently reduce symptom duration following SARS-CoV-2 infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Amanda Brock
- Institute for Translational Medicine and Therapeutics, and
| | - JoEllen Weaver
- Institute for Translational Medicine and Therapeutics, and
| | - John Millar
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephanie DerOhannessian
- Institute for Translational Medicine and Therapeutics, and
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Daniel J. Rader
- Institute for Translational Medicine and Therapeutics, and
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
57
|
Laajaj R, De Los Rios C, Sarmiento-Barbieri I, Aristizabal D, Behrentz E, Bernal R, Buitrago G, Cucunubá Z, de la Hoz F, Gaviria A, Hernández LJ, León L, Moyano D, Osorio E, Varela AR, Restrepo S, Rodriguez R, Schady N, Vives M, Webb D. COVID-19 spread, detection, and dynamics in Bogota, Colombia. Nat Commun 2021; 12:4726. [PMID: 34354078 PMCID: PMC8342514 DOI: 10.1038/s41467-021-25038-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Latin America has been severely affected by the COVID-19 pandemic but estimations of rates of infections are very limited and lack the level of detail required to guide policy decisions. We implemented a COVID-19 sentinel surveillance study with 59,770 RT-PCR tests on mostly asymptomatic individuals and combine this data with administrative records on all detected cases to capture the spread and dynamics of the COVID-19 pandemic in Bogota from June 2020 to early March 2021. We describe various features of the pandemic that appear to be specific to a middle income countries. We find that, by March 2021, slightly more than half of the population in Bogota has been infected, despite only a small fraction of this population being detected. The initial buildup of immunity contributed to the containment of the pandemic in the first and second waves. We also show that the share of the population infected by March 2021 varies widely by occupation, socio-economic stratum, and location. This, in turn, has affected the dynamics of the spread with different groups being infected in the two waves.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Giancarlo Buitrago
- Universidad Nacional de Colombia, Bogota, Colombia
- Hospital Universitario Nacional de Colombia, Bogota, Colombia
| | - Zulma Cucunubá
- Imperial College London, London, UK
- Universidad Pontificia Javeriana, Bogota, Colombia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Huayanay Bernabé ME, Calderón Lozano ML, Moreno Gonzáles ÁR, Vásquez Rojas JG. Anesthesia for cesarean section and SARS Cov-2: Observational study in Peru. COLOMBIAN JOURNAL OF ANESTHESIOLOGY 2021. [DOI: 10.5554/22562087.e998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction: Information regarding the clinical behavior and the anesthetic and perioperative management in pregnant patients with SARS-CoV-2 is starting to appear in the literature in the form of case reports or case series. However, strong evidence and recommendations are still limited.
Objective: To describe the clinical characteristics, the results of anesthetic and perioperative management, and complications in seroprevalent pregnant women for SARS-CoV-2 infection, delivered by cesarean section.
Methodology: Observational study in which 107 clinical records of pregnant women who were seroprevalent for SARS-CoV-2 infection were reviewed and analyzed between April and June, 2020. Demographic, clinical and serological data were collected, as well as data on the anesthetic technique and intraoperative and postoperative complications.
Results: Of the 107 pregnant women with SARS-CoV-2 infection, 99 (92.52%) were asymptomatic and 8 (7.48%) had mild symptoms. The most frequent reasons for cesarean section were cephalo-pelvic disproportion in 20 (18.68%), previous cesarean section in 20 (18.68%) and non-reassuring fetal status in 14 (13.08%). Anesthesia technique was neuraxial in all cases, with spinal used in 100 (93.5%), combined spinal-epidural in 4 (3.7%) and epidural catheter in 3 (2.8%) patients. No deaths had occurred until the third postoperative day of follow-up.
Conclusions: The majority of pregnant women with SARS-CoV-2 infection are asymptomatic. In this work, spinal, combined spinal-epidural and epidural neuroxial anesthesia techniques were shown to be effective and safe for these patients and their newborn babies.
Collapse
|
59
|
Song D, Prahl M, Gaw SL, Narasimhan SR, Rai DS, Huang A, Flores CV, Lin CY, Jigmeddagva U, Wu A, Warrier L, Levan J, Nguyen CBT, Callaway P, Farrington L, Acevedo GR, Gonzalez VJ, Vaaben A, Nguyen P, Atmosfera E, Marleau C, Anderson C, Misra S, Stemmle M, Cortes M, McAuley J, Metz N, Patel R, Nudelman M, Abraham S, Byrne J, Jegatheesan P. Passive and active immunity in infants born to mothers with SARS-CoV-2 infection during pregnancy: prospective cohort study. BMJ Open 2021; 11:e053036. [PMID: 34234001 PMCID: PMC8264915 DOI: 10.1136/bmjopen-2021-053036] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To investigate maternal immunoglobulins' (IgM, IgG) response to SARS-CoV-2 infection during pregnancy and IgG transplacental transfer, to characterise neonatal antibody response to SARS-CoV-2 infection, and to longitudinally follow actively and passively acquired antibodies in infants. DESIGN A prospective observational study. SETTING Public healthcare system in Santa Clara County (California, USA). PARTICIPANTS Women with symptomatic or asymptomatic SARS-CoV-2 infection during pregnancy and their infants were enrolled between 15 April 2020 and 31 March 2021. OUTCOMES SARS-CoV-2 serology analyses in the cord and maternal blood at delivery and longitudinally in infant blood between birth and 28 weeks of life. RESULTS Of 145 mothers who tested positive for SARS-CoV-2 during pregnancy, 86 had symptomatic infections: 78 with mild-moderate symptoms, and 8 with severe-critical symptoms. The seropositivity rates of the mothers at delivery was 65% (95% CI 0.56% to 0.73%) and the cord blood was 58% (95% CI 0.49% to 0.66%). IgG levels significantly correlated between the maternal and cord blood (Rs=0.93, p<0.0001). IgG transplacental transfer ratio was significantly higher when the first maternal positive PCR was 60-180 days before delivery compared with <60 days (1.2 vs 0.6, p<0.0001). Infant IgG seroreversion rates over follow-up periods of 1-4, 5-12, and 13-28 weeks were 8% (4 of 48), 12% (3 of 25), and 38% (5 of 13), respectively. The IgG seropositivity in the infants was positively related to IgG levels in the cord blood and persisted up to 6 months of age. Two newborns showed seroconversion at 2 weeks of age with high levels of IgM and IgG, including one premature infant with confirmed intrapartum infection. CONCLUSIONS Maternal SARS-CoV-2 IgG is efficiently transferred across the placenta when infections occur more than 2 months before delivery. Maternally derived passive immunity may persist in infants up to 6 months of life. Neonates are capable of mounting a strong antibody response to perinatal SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Dongli Song
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Mary Prahl
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Global Health, University of California San Francisco, San Francisco, California, USA
| | - Stephanie L Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Sudha Rani Narasimhan
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Daljeet S Rai
- Department of Family Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Angela Huang
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Claudia V Flores
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Christine Y Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Unurzul Jigmeddagva
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Global Health, University of California San Francisco, San Francisco, California, USA
| | - Alan Wu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lakshmi Warrier
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Justine Levan
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Catherine B T Nguyen
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Perri Callaway
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lila Farrington
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Gonzalo R Acevedo
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Veronica J Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Anna Vaaben
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Phuong Nguyen
- Department of Pathology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Elda Atmosfera
- Department of Pathology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Constance Marleau
- Department of Pathology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Christina Anderson
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Sonya Misra
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Monica Stemmle
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
- Department of Pediatrics, Division of Pediatric Hospital Medicine, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Maria Cortes
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Jennifer McAuley
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Nicole Metz
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Rupalee Patel
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Matthew Nudelman
- Department of Pediatrics, Marshall University, Huntington, West Virginia, USA
| | - Susan Abraham
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
- Department of Pediatrics, Division of Pediatric Hospital Medicine, Santa Clara Valley Medical Center, San Jose, California, USA
| | - James Byrne
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Santa Clara Valley Medical Center, San Jose, California, USA
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California, USA
| | - Priya Jegatheesan
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, California, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
60
|
Pramanick A, Kanneganti A, Wong JLJ, Li SW, Dimri PS, Mahyuddin AP, Kumar S, Illanes SE, Chan JKY, Su LL, Biswas A, Tambyah PA, Huang RY, Mattar CNZ, Choolani M. A reasoned approach towards administering COVID-19 vaccines to pregnant women. Prenat Diagn 2021; 41:1018-1035. [PMID: 34191294 PMCID: PMC8362094 DOI: 10.1002/pd.5985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/20/2021] [Accepted: 05/28/2021] [Indexed: 12/24/2022]
Abstract
There are over 50 SARS-CoV-2 candidate vaccines undergoing Phase II and III clinical trials. Several vaccines have been approved by regulatory authorities and rolled out for use in different countries. Due to concerns of potential teratogenicity or adverse effect on maternal physiology, pregnancy has been a specific exclusion criterion for most vaccine trials with only two trials not excluding pregnant women. Thus, other than limited animal studies, gradually emerging development and reproductive toxicity data, and observational data from vaccine registries, there is a paucity of reliable information to guide recommendations for the safe vaccination of pregnant women. Pregnancy is a risk factor for severe COVID-19, especially in women with comorbidities, resulting in increased rates of preterm birth and maternal morbidity. We discuss the major SARS-CoV-2 vaccines, their mechanisms of action, efficacy, safety profile and possible benefits to the maternal-fetal dyad to create a rational approach towards maternal vaccination while anticipating and mitigating vaccine-related complications. Pregnant women with high exposure risks or co-morbidities predisposing to severe COVID-19 infection should be prioritised for vaccination. Those with risk factors for adverse effects should be counselled accordingly. It is essential to support patient autonomy by shared decision-making involving a risk-benefit discussion with the pregnant woman.
Collapse
Affiliation(s)
- Angsumita Pramanick
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Abhiram Kanneganti
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
| | - Jing Lin Jeslyn Wong
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Sarah Weiling Li
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Pooja Sharma Dimri
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
| | - Aniza Puteri Mahyuddin
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Sailesh Kumar
- Mater Research Institute‐University of QueenslandSouth BrisbaneQueenslandAustralia
- Faculty of MedicineThe University of QueenslandHerstonQueenslandAustralia
| | | | - Jerry Kok Yen Chan
- Department of Reproductive MedicineKK Women's and Children's HospitalSingapore
- Academic Clinical Program in Obstetrics and GynaecologyDuke‐NUS Medical SchoolSingapore
| | - Lin Lin Su
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Arijit Biswas
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Paul Anantharajah Tambyah
- Infectious Diseases Translational Research Programme, Department of Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Ruby Yun‐Ju Huang
- School of MedicineCollege of Medicine, National Taiwan UniversityTaipeiTaiwan
| | - Citra Nurfarah Zaini Mattar
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Mahesh Choolani
- Department of Obstetrics and GynaecologyNational University Hospital SingaporeSingapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| |
Collapse
|
61
|
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Hwee MA, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CAG, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella LA, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer NJ, DeMichele AM, Vardhana SA, Mamtani R, Huang AC. CD8 + T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat Med 2021; 27:1280-1289. [PMID: 34017137 PMCID: PMC8291091 DOI: 10.1038/s41591-021-01386-7] [Citation(s) in RCA: 362] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Patients with cancer have high mortality from coronavirus disease 2019 (COVID-19), and the immune parameters that dictate clinical outcomes remain unknown. In a cohort of 100 patients with cancer who were hospitalized for COVID-19, patients with hematologic cancer had higher mortality relative to patients with solid cancer. In two additional cohorts, flow cytometric and serologic analyses demonstrated that patients with solid cancer and patients without cancer had a similar immune phenotype during acute COVID-19, whereas patients with hematologic cancer had impairment of B cells and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific antibody responses. Despite the impaired humoral immunity and high mortality in patients with hematologic cancer who also have COVID-19, those with a greater number of CD8 T cells had improved survival, including those treated with anti-CD20 therapy. Furthermore, 77% of patients with hematologic cancer had detectable SARS-CoV-2-specific T cell responses. Thus, CD8 T cells might influence recovery from COVID-19 when humoral immunity is deficient. These observations suggest that CD8 T cell responses to vaccination might provide protection in patients with hematologic cancer even in the setting of limited humoral responses.
Collapse
Affiliation(s)
- Erin M Bange
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas A Han
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Wileyto
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Justin Y Kim
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James Robinson
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madeline A Hwee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florence Porterfield
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olutosin Owoyemi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karan Naik
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cathy Zheng
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Galantino
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ariel R Weisman
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline A G Ittner
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily M Kugler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy E Baxter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olutwatosin Oniyide
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Roseline S Agyekum
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Thomas G Dunn
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Tiffanie K Jones
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Heather M Giannini
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madison E Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher M McAllister
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - N Esther Babady
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adam J Widman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sawsan R Boutemine
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte Roberts
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Krista R Budzik
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Tollett
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Carla Wright
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tara Perloff
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital, Philadelphia, NY, USA
| | - Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Josephine R Giles
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Derek A Oldridge
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer E Wu
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Cécile Alanio
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Sharon Adamski
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfred L Garfall
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura A Vella
- Department of Pediatrics, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Samuel J Kerr
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital, Philadelphia, PA, USA
| | - Justine V Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital, Philadelphia, NY, USA
| | - Randall A Oyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital, Philadelphia, PA, USA
| | - Ryan Massa
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Ivan P Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Kara N Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - John P Reilly
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter G Maslak
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert H Vonderheide
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Jedd D Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Scott E Hensley
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Nuala J Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela M DeMichele
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Santosha A Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA.
| | - Ronac Mamtani
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Alexander C Huang
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA.
| |
Collapse
|
62
|
Kubiak JM, Murphy EA, Yee J, Cagino KA, Friedlander RL, Glynn SM, Matthews KC, Jurkiewicz M, Sukhu AC, Zhao Z, Prabhu M, Riley LE, Yang YJ. Severe acute respiratory syndrome coronavirus 2 serology levels in pregnant women and their neonates. Am J Obstet Gynecol 2021; 225:73.e1-73.e7. [PMID: 33497654 PMCID: PMC7825873 DOI: 10.1016/j.ajog.2021.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 01/10/2023]
Abstract
Background Pregnant women and their neonates represent 2 vulnerable populations with an interdependent immune system that are highly susceptible to viral infections. The immune response of pregnant women to severe acute respiratory syndrome coronavirus 2 and the interplay of how the maternal immune response affects the neonatal passive immunity have not been studied systematically. Objective We characterized the serologic response in pregnant women and studied how this serologic response correlates with the maternal clinical presentation and with the rate and level of passive immunity that the neonate received from the mother. Study Design Women who gave birth and who tested positive for immunoglobulin M or immunoglobulin G against severe acute respiratory syndrome coronavirus 2 using semiquantitative detection in a New York City hospital between March 22, 2020, and May 31, 2020, were included in this study. A retrospective chart review of the cases that met the inclusion criteria was conducted to determine the presence of coronavirus disease 2019 symptoms and the use of oxygen support. Serology levels were compared between the symptomatic and asymptomatic patients using a Welch 2 sample t test. Further chart review of the same patient cohort was conducted to identify the dates of self-reported onset of coronavirus disease 2019 symptoms and the timing of the peak immunoglobulin M and immunoglobulin G antibody levels after symptom onset was visualized using local polynomial regression smoothing on log2-scaled serologic values. To study the neonatal serology response, umbilical cord blood samples of the neonates born to the subset of serology positive pregnant women were tested for serologic antibody responses. The maternal antibody levels of serology positive vs the maternal antibody levels of serology negative neonates were compared using the Welch 2 sample t test. The relationship between the quantitative maternal and quantitative neonatal serologic data was studied using a Pearson correlation and linear regression. A multiple linear regression analysis was conducted using maternal symptoms, maternal serology levels, and maternal use of oxygen support to determine the predictors of neonatal immunoglobulin G levels. Results A total of 88 serology positive pregnant women were included in this study. The antibody levels were higher in symptomatic pregnant women than in asymptomatic pregnant women. Serology studies in 34 women with symptom onset data revealed that the maternal immunoglobulin M and immunoglobulin G levels peak around 15 and 30 days after the onset of coronavirus disease 2019 symptoms, respectively. Furthermore, studies of 50 neonates born to this subset of serology positive women showed that passive immunity in the form of immunoglobulin G is conferred in 78% of all neonates. The presence of passive immunity is dependent on the maternal antibody levels, and the levels of neonatal immunoglobulin G correlate with maternal immunoglobulin G levels. The maternal immunoglobulin G levels and maternal use of oxygen support were predictive of the neonatal immunoglobulin G levels. Conclusion We demonstrated that maternal serologies correlate with symptomatic maternal infection, and higher levels of maternal antibodies are associated with passive neonatal immunity. The maternal immunoglobulin G levels and maternal use of oxygen support, a marker of disease severity, predicted the neonatal immunoglobulin G levels. These data will further guide the screening for this uniquely linked population of mothers and their neonates and can aid in developing maternal vaccination strategies.
Collapse
|
63
|
Rothe EM, Fortuna LR, Tobon AL, Postlethwaite A, Sanchez-Lacay JA, Anglero-Diaz YL. Structural Inequities and the Impact of COVID-19 on Latinx Children: Implications for Child and Adolescent Mental Health Practice. J Am Acad Child Adolesc Psychiatry 2021; 60:669-671. [PMID: 33662497 PMCID: PMC8709562 DOI: 10.1016/j.jaac.2021.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/22/2021] [Accepted: 02/23/2021] [Indexed: 11/26/2022]
Abstract
In the United States, the Latinx community (Latinx is a gender-neutral term to describe any person of Latin American descent or heritage) is a heterogeneous population with diverse cultural origins, different migratory experiences, and different socioeconomic and educational realities. The disruptions to daily life and the associated stresses of the 2019 novel coronavirus disease (COVID-19) pandemic have been perhaps most acutely felt by Black and Latinx children from low-income families, including first-generation and undocumented immigrants.1 Structural inequities, such as the lack of employer-sponsored insurance in the service and retail industries; barriers to applying for public benefits, even for those who qualify; chronic poverty; and the lack of linguistically and culturally effective services have contributed to the disproportionate impact. In this article, the authors consider how structural inequities have rendered Latinx children particularly vulnerable to the devastating physical and psychological effects of the pandemic, identify risk and protective factors that are related to mental health outcomes, and recommend ways in which child and adolescent psychiatrists can respond to the escalating needs.
Collapse
Affiliation(s)
- Eugenio M. Rothe
- Herbert Wertheim College of Medicine, Florida International University, Miami
| | - Lisa R. Fortuna
- University of California San Francisco,Correspondence to Lisa R. Fortuna, MD, Department of Psychiatry, Zuckerberg San Francisco General Hospital and Trauma Center, 1001 Potrero Avenue 7M8, San Francisco, CA 94110
| | | | | | - J. Arturo Sanchez-Lacay
- Bronx Care Hospital, New York, and the New York State Psychiatric Institute, Columbia University, Icahn School of Medicine, New York
| | | |
Collapse
|
64
|
Qaseem A, Yost J, Etxeandia-Ikobaltzeta I, Forciea MA, Abraham GM, Miller MC, Obley AJ, Humphrey LL, Centor RM, Akl EA, Andrews R, Bledsoe TA, Haeme R, Kansagara DL. 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 1). Ann Intern Med 2021; 174:828-835. [PMID: 33721518 PMCID: PMC8017476 DOI: 10.7326/m20-7569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [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 widespread availability of SARS-CoV-2 antibody tests raises important questions for clinicians, patients, and public health professionals related to the appropriate use and interpretation of these tests. The Scientific Medical Policy Committee (SMPC) of the American College of Physicians developed these rapid, living practice points to summarize the current and best available evidence on the antibody response to SARS-CoV-2 infection, antibody durability after initial infection with SARS-CoV-2, and antibody protection against reinfection with SARS-CoV-2. METHODS The SMPC developed these rapid, living practice points based on a rapid and living systematic evidence review done by the Portland VA Research Foundation and funded by the Agency for Healthcare Research and Quality. Ongoing literature surveillance is planned through December 2021. When new studies are identified and a full update of the evidence review is published, the SMPC will assess the new evidence and any effect on the practice points. PRACTICE POINT 1 Do not use SARS-CoV-2 antibody tests for the diagnosis of SARS-CoV-2 infection. PRACTICE POINT 2 Antibody tests can be useful for the purpose of estimating community prevalence of SARS-CoV-2 infection. PRACTICE POINT 3 Current evidence is uncertain to predict presence, level, or durability of natural immunity conferred by SARS-CoV-2 antibodies against reinfection (after SARS-CoV-2 infection).
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 and 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
|
65
|
Flannery DD, Gouma S, Dhudasia MB, Mukhopadhyay S, Pfeifer MR, Woodford EC, Triebwasser JE, Gerber JS, Morris JS, Weirick ME, McAllister CM, Bolton MJ, Arevalo CP, Anderson EM, Goodwin EC, Hensley SE, Puopolo KM. Assessment of Maternal and Neonatal Cord Blood SARS-CoV-2 Antibodies and Placental Transfer Ratios. JAMA Pediatr 2021; 175:594-600. [PMID: 33512440 PMCID: PMC7846944 DOI: 10.1001/jamapediatrics.2021.0038] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022]
Abstract
Importance Maternally derived antibodies are a key element of neonatal immunity. Understanding the dynamics of maternal antibody responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy and subsequent transplacental antibody transfer can inform neonatal management as well as maternal vaccination strategies. Objective To assess the association between maternal and neonatal SARS-CoV-2-specific antibody concentrations. Design, Setting, and Participants This cohort study took place at Pennsylvania Hospital in Philadelphia, Pennsylvania. A total of 1714 women delivered at the study site between April 9 and August 8, 2020. Maternal and cord blood sera were available for antibody measurement for 1471 mother/newborn dyads. Exposures SARS-CoV-2. Main Outcomes and Measures IgG and IgM antibodies to the receptor-binding domain of the SARS-CoV-2 spike protein were measured by enzyme-linked immunosorbent assay. Antibody concentrations and transplacental transfer ratios were analyzed in combination with demographic and clinical data. Results The study cohort consisted of 1714 parturient women, with median (interquartile range) age of 32 (28-35) years, of whom 450 (26.3%) identified as Black/non-Hispanic, 879 (51.3%) as White/non-Hispanic, 203 (11.8%) as Hispanic, 126 (7.3%) as Asian, and 56 (3.3%) as other race/ethnicity. Among 1471 mother/newborn dyads for which matched sera were available, SARS-CoV-2 IgG and/or IgM antibodies were detected in 83 of 1471 women (6%; 95% CI, 5%-7%) at the time of delivery, and IgG was detected in cord blood from 72 of 83 newborns (87%; 95% CI, 78%-93%). IgM was not detected in any cord blood specimen, and antibodies were not detected in any infant born to a seronegative mother. Eleven infants born to seropositive mothers were seronegative: 5 of 11 (45%) were born to mothers with IgM antibody only, and 6 of 11 (55%) were born to mothers with significantly lower IgG concentrations compared with those found among mothers of seropositive infants. Cord blood IgG concentrations were positively correlated with maternal IgG concentrations (r = 0.886; P < .001). Placental transfer ratios more than 1.0 were observed among women with asymptomatic SARS-CoV-2 infections as well as those with mild, moderate, and severe coronavirus disease 2019. Transfer ratios increased with increasing time between onset of maternal infection and delivery. Conclusions and Relevance In this cohort study, maternal IgG antibodies to SARS-CoV-2 were transferred across the placenta after asymptomatic as well as symptomatic infection during pregnancy. Cord blood antibody concentrations correlated with maternal antibody concentrations and with duration between onset of infection and delivery. Our findings demonstrate the potential for maternally derived SARS-CoV-2 specific antibodies to provide neonatal protection from coronavirus disease 2019.
Collapse
Affiliation(s)
- Dustin D. Flannery
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - 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
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - 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
| | - Jourdan E. Triebwasser
- Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Jeffrey S. Gerber
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- 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
| | | | - Marcus J. Bolton
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Claudia P. Arevalo
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Elizabeth M. Anderson
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Eileen C. Goodwin
- 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
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
66
|
Arkhipova-Jenkins I, Helfand M, Armstrong C, Gean E, Anderson J, Paynter RA, Mackey K. Antibody Response After SARS-CoV-2 Infection and Implications for Immunity : A Rapid Living Review. Ann Intern Med 2021; 174:811-821. [PMID: 33721517 PMCID: PMC8025942 DOI: 10.7326/m20-7547] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The clinical significance of the antibody response after SARS-CoV-2 infection remains unclear. PURPOSE To synthesize evidence on the prevalence, levels, and durability of detectable antibodies after SARS-CoV-2 infection and whether antibodies to SARS-CoV-2 confer natural immunity. DATA SOURCES MEDLINE (Ovid), Embase, CINAHL, Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, World Health Organization global literature database, and Covid19reviews.org from 1 January through 15 December 2020, limited to peer-reviewed publications available in English. STUDY SELECTION Primary studies characterizing the prevalence, levels, and duration of antibodies in adults with SARS-CoV-2 infection confirmed by reverse transcriptase polymerase chain reaction (RT-PCR); reinfection incidence; and unintended consequences of antibody testing. DATA EXTRACTION Two investigators sequentially extracted study data and rated quality. DATA SYNTHESIS Moderate-strength evidence suggests that most adults develop detectable levels of IgM and IgG antibodies after infection with SARS-CoV-2 and that IgG levels peak approximately 25 days after symptom onset and may remain detectable for at least 120 days. Moderate-strength evidence suggests that IgM levels peak at approximately 20 days and then decline. Low-strength evidence suggests that most adults generate neutralizing antibodies, which may persist for several months like IgG. Low-strength evidence also suggests that older age, greater disease severity, and presence of symptoms may be associated with higher antibody levels. Some adults do not develop antibodies after SARS-CoV-2 infection for reasons that are unclear. LIMITATIONS Most studies were small and had methodological limitations; studies used immunoassays of variable accuracy. CONCLUSION Most adults with SARS-CoV-2 infection confirmed by RT-PCR develop antibodies. Levels of IgM peak early in the disease course and then decline, whereas IgG peaks later and may remain detectable for at least 120 days. PRIMARY FUNDING SOURCE Agency for Healthcare Research and Quality. (PROSPERO: CRD42020207098).
Collapse
Affiliation(s)
- Irina Arkhipova-Jenkins
- Scientific Resource Center for the AHRQ Evidence-based Practice Center Program, Portland VA Research Foundation, and VA Portland Health Care System, Portland, Oregon (I.A.J., C.A., E.G., R.A.P.)
| | - Mark Helfand
- Scientific Resource Center for the AHRQ Evidence-based Practice Center Program, Portland VA Research Foundation, VA Evidence Synthesis Program, and VA Portland Health Care System, Portland, Oregon (M.H.)
| | - Charlotte Armstrong
- Scientific Resource Center for the AHRQ Evidence-based Practice Center Program, Portland VA Research Foundation, and VA Portland Health Care System, Portland, Oregon (I.A.J., C.A., E.G., R.A.P.)
| | - Emily Gean
- Scientific Resource Center for the AHRQ Evidence-based Practice Center Program, Portland VA Research Foundation, and VA Portland Health Care System, Portland, Oregon (I.A.J., C.A., E.G., R.A.P.)
| | - Joanna Anderson
- VA Evidence Synthesis Program and VA Portland Health Care System, Portland, Oregon (J.A., K.M.)
| | - Robin A Paynter
- Scientific Resource Center for the AHRQ Evidence-based Practice Center Program, Portland VA Research Foundation, and VA Portland Health Care System, Portland, Oregon (I.A.J., C.A., E.G., R.A.P.)
| | - Katherine Mackey
- VA Evidence Synthesis Program and VA Portland Health Care System, Portland, Oregon (J.A., K.M.)
| |
Collapse
|
67
|
Saadaoui M, Kumar M, Al Khodor S. COVID-19 Infection during Pregnancy: Risk of Vertical Transmission, Fetal, and Neonatal Outcomes. J Pers Med 2021; 11:483. [PMID: 34071251 PMCID: PMC8227688 DOI: 10.3390/jpm11060483] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/18/2022] Open
Abstract
The COVID-19 pandemic is a worldwide, critical public health challenge and is considered one of the most communicable diseases that the world had faced so far. Response and symptoms associated with COVID-19 vary between the different cases recorded, but it is amply described that symptoms become more aggressive in subjects with a weaker immune system. This includes older subjects, patients with chronic diseases, patients with immunosuppression treatment, and pregnant women. Pregnant women are receiving more attention not only because of their altered physiological and immunological function but also for the potential risk of viral vertical transmission to the fetus or infant. However, very limited data about the impact of maternal infection during pregnancy, such as the possibility of vertical transmission in utero, during birth, or via breastfeeding, is available. Moreover, the impact of infection on the newborn in the short and long term remains poorly understood. Therefore, it is vital to collect and analyze data from pregnant women infected with COVID-19 to understand the viral pathophysiology during pregnancy and its effects on the offspring. In this article, we review the current knowledge about pre-and post-natal COVID-19 infection, and we discuss whether vertical transmission takes place in pregnant women infected with the virus and what are the current recommendations that pregnant women should follow in order to be protected from the virus.
Collapse
Affiliation(s)
| | | | - Souhaila Al Khodor
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (M.S.); (M.K.)
| |
Collapse
|
68
|
Ma KC, Menkir TF, Kissler S, Grad YH, Lipsitch M. Modeling the impact of racial and ethnic disparities on COVID-19 epidemic dynamics. eLife 2021; 10:e66601. [PMID: 34003112 PMCID: PMC8221808 DOI: 10.7554/elife.66601] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Background The impact of variable infection risk by race and ethnicity on the dynamics of SARS-CoV-2 spread is largely unknown. Methods Here, we fit structured compartmental models to seroprevalence data from New York State and analyze how herd immunity thresholds (HITs), final sizes, and epidemic risk change across groups. Results A simple model where interactions occur proportionally to contact rates reduced the HIT, but more realistic models of preferential mixing within groups increased the threshold toward the value observed in homogeneous populations. Across all models, the burden of infection fell disproportionately on minority populations: in a model fit to Long Island serosurvey and census data, 81% of Hispanics or Latinos were infected when the HIT was reached compared to 34% of non-Hispanic whites. Conclusions Our findings, which are meant to be illustrative and not best estimates, demonstrate how racial and ethnic disparities can impact epidemic trajectories and result in unequal distributions of SARS-CoV-2 infection. Funding K.C.M. was supported by National Science Foundation GRFP grant DGE1745303. Y.H.G. and M.L. were funded by the Morris-Singer Foundation. M.L. was supported by SeroNet cooperative agreement U01 CA261277.
Collapse
Affiliation(s)
- Kevin C Ma
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public HealthBostonUnited States
| | - Tigist F Menkir
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public HealthBostonUnited States
| | - Stephen Kissler
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public HealthBostonUnited States
| | - Yonatan H Grad
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public HealthBostonUnited States
- Division of Infectious Diseases, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Marc Lipsitch
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public HealthBostonUnited States
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public HealthBostonUnited States
| |
Collapse
|
69
|
Song D, Prahl M, Gaw SL, Narasimhan S, Rai D, Huang A, Flores C, Lin CY, Jigmeddagva U, Wu AH, Warrier L, Levan J, Nguyen CB, Callaway P, Farrington L, Acevedo GR, Gonzalez VJ, Vaaben A, Nguyen P, Atmosfera E, Marleau C, Anderson C, Misra S, Stemmle M, Cortes M, McAuley J, Metz N, Patel R, Nudelman M, Abraham S, Byrne J, Jegatheesan P. Passive and active immunity in infants born to mothers with SARS-CoV-2 infection during pregnancy: Prospective cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.05.01.21255871. [PMID: 33972953 PMCID: PMC8109203 DOI: 10.1101/2021.05.01.21255871] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To investigate maternal immunoglobulins' (IgM, IgG) response to SARS-CoV-2 infection during pregnancy and IgG transplacental transfer, to characterize neonatal antibody response to SARS-CoV-2 infection, and to longitudinally follow actively- and passively-acquired SARS-CoV-2 antibodies in infants. DESIGN A prospective observational study. SETTING A public healthcare system in Santa Clara County (CA, USA). PARTICIPANTS Women with SARS-CoV-2 infection during pregnancy and their infants were enrolled between April 15, 2020 and March 31, 2021. OUTCOMES SARS-CoV-2 serology analyses in the cord and maternal blood at delivery and longitudinally in infant blood between birth and 28 weeks of life. RESULTS Of 145 mothers who tested positive for SARS-CoV-2 during pregnancy, 86 had symptomatic infections: 78 with mild-moderate symptoms, and eight with severe-critical symptoms. Of the 147 newborns, two infants showed seroconversion at two weeks of age with high levels of IgM and IgG, including one premature infant with confirmed intrapartum infection. The seropositivity rates of the mothers at delivery was 65% (95% CI 0.56-0.73) and the cord blood was 58% (95% CI 0.49-0.66). IgG levels significantly correlated between the maternal and cord blood (Rs= 0.93, p< 0.0001). IgG transplacental transfer ratio was significantly higher when the first maternal positive PCR was 60-180 days before delivery compared to <60 days (1.2 vs. 0.6, p=<0.0001). Infant IgG negative conversion rate over follow-up periods of 1-4, 5-12, and 13-28 weeks were 8% (4/48), 12% (3/25), and 38% (5/13), respectively. The IgG seropositivity in the infants was positively related to IgG levels in the cord blood and persisted up to six months of age. CONCLUSIONS Maternal SARS-CoV-2 IgG is efficiently transferred across the placenta when infections occur more than two months before delivery. Maternally-derived passive immunity may protect infants up to six months of life. Neonates mount a strong antibody response to perinatal SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Dongli Song
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mary Prahl
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Global Health, University of California, San Francisco, CA, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - SudhaRani Narasimhan
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Daljeet Rai
- Department of Family Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Angela Huang
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Claudia Flores
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Christine Y. Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Alan H.B. Wu
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Lakshmi Warrier
- Department of Medicine, University of California San Francisco, CA, USA
| | - Justine Levan
- Department of Medicine, University of California San Francisco, CA, USA
| | | | - Perri Callaway
- Department of Medicine, University of California San Francisco, CA, USA
| | - Lila Farrington
- Department of Medicine, University of California San Francisco, CA, USA
| | | | - Veronica J. Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Anna Vaaben
- Department of Medicine, University of California San Francisco, CA, USA
| | - Phuong Nguyen
- Department of Pathology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Elda Atmosfera
- Department of Pathology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Constance Marleau
- Department of Pathology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Christina Anderson
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sonya Misra
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Monica Stemmle
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria Cortes
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Jennifer McAuley
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Nicole Metz
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Rupalee Patel
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Matthew Nudelman
- Department of Pediatrics, Marshall University, Huntington, WV, USA
| | - Susan Abraham
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - James Byrne
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Santa Clara Valley, CA, USA
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Priya Jegatheesan
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
70
|
Godinas L, Iyer K, Meszaros G, Quarck R, Escribano-Subias P, Vonk Noordegraaf A, Jansa P, D'Alto M, Luknar M, Milutinov Ilic S, Belge C, Sitbon O, Reis A, Rosenkranz S, Pepke-Zaba J, Humbert M, Delcroix M. PH CARE COVID survey: an international patient survey on the care for pulmonary hypertension patients during the early phase of the COVID-19 pandemic. Orphanet J Rare Dis 2021; 16:196. [PMID: 33933110 PMCID: PMC8087873 DOI: 10.1186/s13023-021-01752-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/16/2021] [Indexed: 12/23/2022] Open
Abstract
Background During the COVID-19 pandemic, most of the health care systems suspended their non-urgent activities. This included the cancellation of consultations for patients with rare diseases, such as severe pulmonary hypertension (PH), resulting in potential medication shortage and loss of follow-up. Thus, the aim of the study was to evaluate PH patient health status evolution, access to health care and mental health experience during the early phase of the pandemic. Methods We conducted an online patient survey, available in 16 languages, between 22/05/2020 and 28/06/2020. The survey included questions corresponding to demographic, COVID-19 and PH related information. Results 1073 patients (or relatives, 27%) from 52 countries all over the world participated in the survey. Seventy-seven percent (77%) of responders reported a diagnosis of pulmonary arterial hypertension and 15% of chronic thromboembolic PH. The COVID-19 related events were few: only 1% of all responders reported a diagnosis of COVID-19. However, 8% of patients reported health deterioration possibly related to PH, and 4% hospitalization for PH. Besides, 11% of the patients reported difficulties to access their PH expert centre, and 3% interruption of treatment due to shortage of medication. Anxiety or depression was reported by 67% of the participants. Conclusion Although COVID-19 incidence in PH patients was low, PH related problems occurred frequently as the pandemic progressed, including difficulties to have access to specialized care. The importance of primary health care was emphasized. Further studies are needed to evaluate the long-term consequences of COVID-related PH care disruption. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01752-1.
Collapse
Affiliation(s)
- Laurent Godinas
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium. .,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium.
| | - Keerthana Iyer
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France.,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de L'Hypertension Pulmonaire, ERN-LUNG, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | | | - Rozenn Quarck
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Pilar Escribano-Subias
- Cardiology Department and Spanish Cardiovascular Research Network (CIBER-CV), Hospital Universitario, 12 de Octubre, Madrid, Spain
| | - Anton Vonk Noordegraaf
- Departement of Pulmonary Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, Amsterdam, Netherlands
| | - Pavel Jansa
- Department of Medicine - Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Michele D'Alto
- Department of Cardiology, Monaldi Hospital, Naples, Italy
| | - Milan Luknar
- National Cardiovascular Institute, Comenius University School of Medicine, Pod Krasnou Horkou 1, Bratislava, Slovakia
| | | | - Catharina Belge
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Olivier Sitbon
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France.,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de L'Hypertension Pulmonaire, ERN-LUNG, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Abílio Reis
- Medicine Department, Pulmonary Vascular Diseases Unit, Centro Hospitalar Universitário Do Porto, Porto, Portugal
| | - Stephan Rosenkranz
- Department III of Internal Medicine and, Cologne Cardiovascular Research Center (CCRC), Cologne University Heart Center, Cologne, Germany
| | - Joanna Pepke-Zaba
- Pulmonary Vascular Diseases Unit, Royal Papworth Hospital, Cambridge, UK
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France.,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de L'Hypertension Pulmonaire, ERN-LUNG, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Marion Delcroix
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
71
|
Lara-Cinisomo S, D'Anna-Hernandez K, Non AL. Recommendations for Clinical Practice, Research, and Policy to Address the Effects of the COVID-19 Pandemic on Anxiety Symptoms in Immigrant and U.S.-Born Latina Mothers. Womens Health Issues 2021; 31:301-305. [PMID: 33893016 DOI: 10.1016/j.whi.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Sandraluz Lara-Cinisomo
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois.
| | | | - Amy L Non
- Department of Anthropology, University of California San Diego, La Jolla, California
| |
Collapse
|
72
|
Gouma S, Weirick ME, Bolton MJ, Arevalo CP, Goodwin EC, Anderson EM, McAllister CM, Christensen SR, Dunbar D, Fiore D, Brock A, Weaver J, Millar J, DerOhannessian S, The UPenn COVID Processing Unit, Frank I, Rader DJ, Wherry EJ, Hensley SE. Sero-monitoring of health care workers reveals complex relationships between common coronavirus antibodies and SARS-CoV-2 severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.04.12.21255324. [PMID: 33907765 PMCID: PMC8077588 DOI: 10.1101/2021.04.12.21255324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recent common coronavirus (CCV) infections are associated with reduced COVID-19 severity upon SARS-CoV-2 infection, however the immunological mechanisms involved are unknown. We completed serological assays using samples collected from health care workers to identify antibody types associated with SARS-CoV-2 protection and COVID-19 severity. Rare SARS-CoV-2 cross-reactive antibodies elicited by past CCV infections were not associated with protection; however, the duration of symptoms following SARS-CoV-2 infections was significantly reduced in individuals with higher common betacoronavirus (βCoV) antibody titers. Since antibody titers decline over time after CCV infections, individuals in our cohort with higher βCoV antibody titers were more likely recently infected with common βCoVs compared to individuals with lower antibody titers. Therefore, our data suggest that recent βCoV infections potentially limit the severity of SARS-CoV-2 infections through mechanisms that do not involve cross-reactive antibodies. Our data are consistent with the emerging hypothesis that cellular immune responses elicited by recent common βCoV infections transiently reduce disease severity following SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Madison E. Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Marcus J. Bolton
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Claudia P. Arevalo
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eileen C. Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth M. Anderson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christopher M. McAllister
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shannon R. Christensen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Debora Dunbar
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Danielle Fiore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amanda Brock
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - JoEllen Weaver
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John Millar
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephanie DerOhannessian
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - The UPenn COVID Processing Unit
- The UPenn COVID Processing Unit is a unit of individuals from diverse laboratories at the University of Pennsylvania who volunteered time and effort to enable study of COVID-19 patients during the pandemic. Members are listed in the acknowledgement section
| | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel J. Rader
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
73
|
Nande A, Sheen J, Walters EL, Klein B, Chinazzi M, Gheorghe AH, Adlam B, Shinnick J, Tejeda MF, Scarpino SV, Vespignani A, Greenlee AJ, Schneider D, Levy MZ, Hill AL. The effect of eviction moratoria on the transmission of SARS-CoV-2. Nat Commun 2021; 12:2274. [PMID: 33859196 PMCID: PMC8050248 DOI: 10.1038/s41467-021-22521-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/17/2021] [Indexed: 12/21/2022] Open
Abstract
Massive unemployment during the COVID-19 pandemic could result in an eviction crisis in US cities. Here we model the effect of evictions on SARS-CoV-2 epidemics, simulating viral transmission within and among households in a theoretical metropolitan area. We recreate a range of urban epidemic trajectories and project the course of the epidemic under two counterfactual scenarios, one in which a strict moratorium on evictions is in place and enforced, and another in which evictions are allowed to resume at baseline or increased rates. We find, across scenarios, that evictions lead to significant increases in infections. Applying our model to Philadelphia using locally-specific parameters shows that the increase is especially profound in models that consider realistically heterogenous cities in which both evictions and contacts occur more frequently in poorer neighborhoods. Our results provide a basis to assess eviction moratoria and show that policies to stem evictions are a warranted and important component of COVID-19 control.
Collapse
Affiliation(s)
- Anjalika Nande
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, USA
| | - Justin Sheen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Emma L Walters
- Department of Urban and Regional Planning, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Brennan Klein
- Network Science Institute, Northeastern University, Boston, MA, USA
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, USA
| | - Matteo Chinazzi
- Network Science Institute, Northeastern University, Boston, MA, USA
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, USA
| | - Andrei H Gheorghe
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, USA
| | - Ben Adlam
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, USA
| | - Julianna Shinnick
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maria Florencia Tejeda
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Alessandro Vespignani
- Network Science Institute, Northeastern University, Boston, MA, USA
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, USA
| | - Andrew J Greenlee
- Department of Urban and Regional Planning, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Daniel Schneider
- Department of Urban and Regional Planning, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Michael Z Levy
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Alison L Hill
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, USA.
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
74
|
Goel RR, Apostolidis SA, Painter MM, Mathew D, Pattekar A, Kuthuru O, Gouma S, Hicks P, Meng W, Rosenfeld AM, Dysinger S, Lundgreen KA, Kuri-Cervantes L, Adamski S, Hicks A, Korte S, Oldridge DA, Baxter AE, Giles JR, Weirick ME, McAllister CM, Dougherty J, Long S, D'Andrea K, Hamilton JT, Betts MR, Luning Prak ET, Bates P, Hensley SE, Greenplate AR, Wherry EJ. Distinct antibody and memory B cell responses in SARS-CoV-2 naïve and recovered individuals following mRNA vaccination. Sci Immunol 2021; 6:eabi6950. [PMID: 33858945 PMCID: PMC8158969 DOI: 10.1126/sciimmunol.abi6950] [Citation(s) in RCA: 496] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022]
Abstract
Novel mRNA vaccines for SARS-CoV-2 have been authorized for emergency use. Despite their efficacy in clinical trials, data on mRNA vaccine-induced immune responses are mostly limited to serological analyses. Here, we interrogated antibody and antigen-specific memory B cells over time in 33 SARS-CoV-2 naïve and 11 SARS-CoV-2 recovered subjects. SARS-CoV-2 naïve individuals required both vaccine doses for optimal increases in antibodies, particularly for neutralizing titers against the B.1.351 variant. Memory B cells specific for full-length spike protein and the spike receptor binding domain (RBD) were also efficiently primed by mRNA vaccination and detectable in all SARS-CoV-2 naive subjects after the second vaccine dose, though the memory B cell response declined slightly with age. In SARS-CoV-2 recovered individuals, antibody and memory B cell responses were significantly boosted after the first vaccine dose; however, there was no increase in circulating antibodies, neutralizing titers, or antigen-specific memory B cells after the second dose. This robust boosting after the first vaccine dose strongly correlated with levels of pre-existing memory B cells in recovered individuals, identifying a key role for memory B cells in mounting recall responses to SARS-CoV-2 antigens. Together, our data demonstrated robust serological and cellular priming by mRNA vaccines and revealed distinct responses based on prior SARS-CoV-2 exposure, whereby COVID-19 recovered subjects may only require a single vaccine dose to achieve peak antibody and memory B cell responses. These findings also highlight the utility of defining cellular responses in addition to serologies and may inform SARS-CoV-2 vaccine distribution in a resource-limited setting.
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
| | - 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
| | - 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
| | - 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
| | - Ajinkya Pattekar
- 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
| | - 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
| | - 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
| | - Sarah Dysinger
- Department of Microbiology, 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
| | - 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
| | - Sharon Adamski
- Immune Health™, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amanda Hicks
- 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
| | - 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
| | - Amy E Baxter
- Institute for Immunology, 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
| | - 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
| | - 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
| | - Paul Bates
- 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
| | - 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
|
75
|
Anderson EM, Goodwin EC, Verma A, Arevalo CP, Bolton MJ, Weirick ME, Gouma S, McAllister CM, Christensen SR, Weaver J, Hicks P, Manzoni TB, Oniyide O, Ramage H, Mathew D, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, Alanio C, D'Andrea K, Kuthuru O, Dougherty J, Pattekar A, Kim J, Han N, Apostolidis SA, Huang AC, Vella LA, Kuri-Cervantes L, Pampena MB, Betts MR, Wherry EJ, Meyer NJ, Cherry S, Bates P, Rader DJ, Hensley SE. Seasonal human coronavirus antibodies are boosted upon SARS-CoV-2 infection but not associated with protection. Cell 2021; 184:1858-1864.e10. [PMID: 33631096 PMCID: PMC7871851 DOI: 10.1016/j.cell.2021.02.010] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread within the human population. Although SARS-CoV-2 is a novel coronavirus, most humans had been previously exposed to other antigenically distinct common seasonal human coronaviruses (hCoVs) before the coronavirus disease 2019 (COVID-19) pandemic. Here, we quantified levels of SARS-CoV-2-reactive antibodies and hCoV-reactive antibodies in serum samples collected from 431 humans before the COVID-19 pandemic. We then quantified pre-pandemic antibody levels in serum from a separate cohort of 251 individuals who became PCR-confirmed infected with SARS-CoV-2. Finally, we longitudinally measured hCoV and SARS-CoV-2 antibodies in the serum of hospitalized COVID-19 patients. Our studies indicate that most individuals possessed hCoV-reactive antibodies before the COVID-19 pandemic. We determined that ∼20% of these individuals possessed non-neutralizing antibodies that cross-reacted with SARS-CoV-2 spike and nucleocapsid proteins. These antibodies were not associated with protection against SARS-CoV-2 infections or hospitalizations, but they were boosted upon SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Elizabeth M Anderson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eileen C Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anurag Verma
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Claudia P Arevalo
- 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
| | - Madison E Weirick
- 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
| | - Christopher M McAllister
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shannon R Christensen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - JoEllen Weaver
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Philip Hicks
- School of Veterinary 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
| | - Oluwatosin Oniyide
- Division of Pulmonary, Allergy, and Critical Care Medicine and Center for Translational Lung Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Holly Ramage
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Divij Mathew
- Institute for Immunology, 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
| | - Amy E Baxter
- Institute for Immunology, 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
| | - Derek A Oldridge
- Institute for Immunology, 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
| | - Allison R Greenplate
- Institute for Immunology, 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
| | - Jennifer E Wu
- Institute for Immunology, 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, Philadelphia, PA 19104, USA
| | - Cécile Alanio
- Institute for Immunology, 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, Philadelphia, PA 19104, USA
| | - Kurt D'Andrea
- Institute for Immunology, 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
| | - Oliva Kuthuru
- Institute for Immunology, 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
| | - Jeanette Dougherty
- Institute for Immunology, 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
| | - Ajinkya Pattekar
- Institute for Immunology, 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
| | - Justin Kim
- Institute for Immunology, 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
| | - Nicholas Han
- Institute for Immunology, 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; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex C Huang
- Institute for Immunology, 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
| | - Laura A Vella
- Institute for Immunology, 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; Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Betina Pampena
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael R Betts
- 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, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nuala J Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine and Center for Translational Lung Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Center for Research on Coronavirus and Other Emerging Pathogens, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J Rader
- Department of Genetics, 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.
| |
Collapse
|
76
|
Abstract
PURPOSE OF REVIEW The aim of this review was to describe the process of and challenges in developing national guidance for management of infants born to mothers with COVID-19. RECENT FINDINGS Beginning in January 2020, infection with the novel coronavirus SARS-CoV-2 spread across the USA, causing the illness COVID-19. As pregnant women began to present for delivery while sick with COVID-19, the American Academy of Pediatrics (AAP) convened a writing group in March 2020 to develop guidance for the management of their newborns. The initial guidance was developed emergently and was forced to rely on extremely limited data from China. The initial guidance advocated for a conservative approach that included temporary physical separation of infected mother and newborn. To address the knowledge deficit, the AAP sponsored a volunteer registry to collect data on perinatal infection and management. As data have emerged informing the natural history of COVID-19, the performance of PCR-based diagnostics, the value of infection control measures and the risk of infant disease, AAP has issued serial updates to newborn guidance. SUMMARY Evolving knowledge on the epidemiology of perinatal COVID-19 has informed newborn guidance. The most recent guidance focuses on the use of infection control measures to support maternal-newborn contact and breastfeeding.
Collapse
Affiliation(s)
- Dustin D. Flannery
- Division of Neonatology, Children's Hospital of Philadelphia
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Karen M. Puopolo
- Division of Neonatology, Children's Hospital of Philadelphia
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
77
|
Kiseleva AA, Troisi EM, Hensley SE, Kohli RM, Epstein JA. SARS-CoV-2 spike protein binding selectively accelerates substrate-specific catalytic activity of ACE2. J Biochem 2021; 170:299-306. [PMID: 33774672 PMCID: PMC8083718 DOI: 10.1093/jb/mvab041] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/22/2021] [Indexed: 11/26/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that has given rise to the devastating global pandemic. In most cases, SARS-CoV-2 infection results in the development of viral pneumonia and acute respiratory distress syndrome, known as ‘coronavirus disease 2019’ or COVID-19. Intriguingly, besides the respiratory tract, COVID-19 affects other organs and systems of the human body. COVID-19 patients with pre-existing cardiovascular disease have a higher risk of death, and SARS-CoV-2 infection itself may cause myocardial inflammation and injury. One possible explanation of such phenomena is the fact that SARS-CoV-2 utilizes angiotensin-converting enzyme 2 (ACE2) as the receptor required for viral entry. ACE2 is expressed in the cells of many organs, including the heart. ACE2 functions as a carboxypeptidase that can cleave several endogenous substrates, including angiotensin II, thus regulating blood pressure and vascular tone. It remains largely unknown if the SARS-CoV-2 infection alters the enzymatic properties of ACE2, thereby contributing to cardiovascular complications in patients with COVID-19. Here, we demonstrate that ACE2 cleavage of des-Arg9-bradykinin substrate analogue is markedly accelerated, while cleavage of angiotensin II analogue is minimally affected by the binding of spike protein. These findings may have implications for a better understanding of COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Anna A Kiseleva
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth M Troisi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Rahul M Kohli
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jonathan A Epstein
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
78
|
Goel RR, Apostolidis SA, Painter MM, Mathew D, Pattekar A, Kuthuru O, Gouma S, Kuri-Cervantes L, Meng W, Adamski S, Baxter AE, Giles JR, Weirick ME, McAllister CM, Hicks A, Korte S, Dougherty J, Long S, D’Andrea K, Hamilton JT, Prak ETL, Betts MR, Bates P, Hensley SE, Greenplate AR, Wherry EJ. Longitudinal Analysis Reveals Distinct Antibody and Memory B Cell Responses in SARS-CoV2 Naïve and Recovered Individuals Following mRNA Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.03.03.21252872. [PMID: 33688691 PMCID: PMC7941668 DOI: 10.1101/2021.03.03.21252872] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Novel mRNA vaccines for SARS-CoV2 have been authorized for emergency use and are currently being administered to millions of individuals worldwide. Despite their efficacy in clinical trials, there is limited data on vaccine-induced immune responses in individuals with a prior SARS-CoV2 infection compared to SARS-CoV2 naïve subjects. Moreover, how mRNA vaccines impact the development of antibodies as well as memory B cells in COVID-19 experienced versus COVID-19 naïve subjects remains poorly understood. In this study, we evaluated antibody responses and antigen-specific memory B cell responses over time in 33 SARS-CoV2 naïve and 11 SARS-CoV2 recovered subjects. mRNA vaccination induced significant antibody and memory B cell responses against full-length SARS-CoV2 spike protein and the spike receptor binding domain (RBD). SARS-CoV2 naïve individuals benefitted from both doses of mRNA vaccine with additional increases in antibodies and memory B cells following booster immunization. In contrast, SARS-CoV2 recovered individuals had a significant immune response after the first dose with no increase in circulating antibodies or antigen-specific memory B cells after the second dose. Moreover, the magnitude of the memory B cell response induced by vaccination was lower in older individuals, revealing an age-dependence to mRNA vaccine-induced B cell memory. Side effects also tended to associate with post-boost antibody levels, but not with post-boost memory B cells, suggesting that side effect severity may be a surrogate of short-term antibody responses. The frequency of pre-vaccine antigen-specific memory B cells in SARS-CoV2 recovered individuals strongly correlated with post-vaccine antibody levels, supporting a key role for memory B cells in humoral recall responses to SARS-CoV2. This observation may have relevance for future booster vaccines and for responses to viral variants that partially escape pre-existing antibodies and require new humoral responses to be generated from memory B cells. Finally, post-boost antibody levels were not correlated with post-boost memory responses in SARS-CoV2 naïve individuals, indicating that short-term antibody levels and memory B cells are complementary immunological endpoints that should be examined in tandem when evaluating vaccine response. Together, our data provide evidence of both serological response and immunological memory following mRNA vaccination that is distinct based on prior SARS-CoV2 exposure. These findings may inform vaccine distribution in a resource-limited setting.
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
| | - 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
| | - 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
| | - 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
| | - Ajinkya Pattekar
- 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
| | - 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
| | - 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
| | - Sharon Adamski
- 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
| | - 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
| | - Amanda Hicks
- 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
| | - 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
- Immune Health™, 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
| | - 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
| | - 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
| | - Paul Bates
- 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
| | - 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
|
79
|
Vella LA, Giles JR, Baxter AE, Oldridge DA, Diorio C, Kuri-Cervantes L, Alanio C, Pampena MB, Wu JE, Chen Z, Huang YJ, Anderson EM, Gouma S, McNerney KO, Chase J, Burudpakdee C, Lee JH, Apostolidis SA, Huang AC, Mathew D, Kuthuru O, Goodwin EC, Weirick ME, Bolton MJ, Arevalo CP, Ramos A, Jasen CJ, Conrey PE, Sayed S, Giannini HM, D'Andrea K, Meyer NJ, Behrens EM, Bassiri H, Hensley SE, Henrickson SE, Teachey DT, Betts MR, Wherry EJ. Deep immune profiling of MIS-C demonstrates marked but transient immune activation compared to adult and pediatric COVID-19. Sci Immunol 2021; 6:eabf7570. [PMID: 33653907 PMCID: PMC8128303 DOI: 10.1126/sciimmunol.abf7570] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/25/2021] [Indexed: 12/14/2022]
Abstract
Pediatric COVID-19 following SARS-CoV-2 infection is associated with fewer hospitalizations and often milder disease than in adults. A subset of children, however, present with Multisystem Inflammatory Syndrome in Children (MIS-C) that can lead to vascular complications and shock, but rarely death. The immune features of MIS-C compared to pediatric COVID-19 or adult disease remain poorly understood. We analyzed peripheral blood immune responses in hospitalized SARS-CoV-2 infected pediatric patients (pediatric COVID-19) and patients with MIS-C. MIS-C patients had patterns of T cell-biased lymphopenia and T cell activation similar to severely ill adults, and all patients with MIS-C had SARS-CoV-2 spike-specific antibodies at admission. A distinct feature of MIS-C patients was robust activation of vascular patrolling CX3CR1+ CD8+ T cells that correlated with the use of vasoactive medication. Finally, whereas pediatric COVID-19 patients with acute respiratory distress syndrome (ARDS) had sustained immune activation, MIS-C patients displayed clinical improvement over time, concomitant with decreasing immune activation. Thus, non-MIS-C versus MIS-C SARS-CoV-2 associated illnesses are characterized by divergent immune signatures that are temporally distinct from one another and implicate CD8+ T cells in the clinical presentation and trajectory of MIS-C.
Collapse
Affiliation(s)
- Laura A Vella
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Josephine R Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Amy E Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Derek A Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Caroline Diorio
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Leticia Kuri-Cervantes
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Cécile Alanio
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - M Betina Pampena
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jennifer E Wu
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Zeyu Chen
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Yinghui Jane Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Elizabeth M Anderson
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sigrid Gouma
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Kevin O McNerney
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Julie Chase
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Chakkapong Burudpakdee
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jessica H Lee
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sokratis A Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Alexander C Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Eileen C Goodwin
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Madison E Weirick
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Marcus J Bolton
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Claudia P Arevalo
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Andre Ramos
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - C J Jasen
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,19104, USA
| | - Peyton E Conrey
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,19104, USA
| | - Samir Sayed
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,19104, USA
| | - Heather M Giannini
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kurt D'Andrea
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nuala J Meyer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Edward M Behrens
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hamid Bassiri
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Scott E Hensley
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sarah E Henrickson
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,19104, USA
| | - David T Teachey
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Michael R Betts
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| |
Collapse
|
80
|
Mackey K, Ayers CK, Kondo KK, Saha S, Advani SM, Young S, Spencer H, Rusek M, Anderson J, Veazie S, Smith M, Kansagara D. Racial and Ethnic Disparities in COVID-19-Related Infections, Hospitalizations, and Deaths : A Systematic Review. Ann Intern Med 2021; 174:362-373. [PMID: 33253040 PMCID: PMC7772883 DOI: 10.7326/m20-6306] [Citation(s) in RCA: 773] [Impact Index Per Article: 193.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Data suggest that the effects of coronavirus disease 2019 (COVID-19) differ among U.S. racial/ethnic groups. PURPOSE To evaluate racial/ethnic disparities in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection rates and COVID-19 outcomes, factors contributing to disparities, and interventions to reduce them. DATA SOURCES English-language articles in MEDLINE, PsycINFO, CINAHL, Cochrane Central Register of Controlled Trials, Cochrane Database of Systematic Reviews, and Scopus, searched from inception through 31 August 2020. Gray literature sources were searched through 2 November 2020. STUDY SELECTION Observational studies examining SARS-CoV-2 infections, hospitalizations, or deaths by race/ethnicity in U.S. settings. DATA EXTRACTION Single-reviewer abstraction confirmed by a second reviewer; independent dual-reviewer assessment of quality and strength of evidence. DATA SYNTHESIS 37 mostly fair-quality cohort and cross-sectional studies, 15 mostly good-quality ecological studies, and data from the Centers for Disease Control and Prevention and APM Research Lab were included. African American/Black and Hispanic populations experience disproportionately higher rates of SARS-CoV-2 infection, hospitalization, and COVID-19-related mortality compared with non-Hispanic White populations, but not higher case-fatality rates (mostly reported as in-hospital mortality) (moderate- to high-strength evidence). Asian populations experience similar outcomes to non-Hispanic White populations (low-strength evidence). Outcomes for other racial/ethnic groups have been insufficiently studied. Health care access and exposure factors may underlie the observed disparities more than susceptibility due to comorbid conditions (low-strength evidence). LIMITATIONS Selection bias, missing race/ethnicity data, and incomplete outcome assessments in cohort and cross-sectional studies must be considered. In addition, adjustment for key demographic covariates was lacking in ecological studies. CONCLUSION African American/Black and Hispanic populations experience disproportionately higher rates of SARS-CoV-2 infection and COVID-19-related mortality but similar rates of case fatality. Differences in health care access and exposure risk may be driving higher infection and mortality rates. PRIMARY FUNDING SOURCE Department of Veterans Affairs, Veterans Health Administration, Health Services Research & Development. (PROSPERO: CRD42020187078).
Collapse
Affiliation(s)
- Katherine Mackey
- VA Evidence Synthesis Program, VA Portland Health Care System and Oregon Health & Science University, Portland, Oregon (K.M., K.K.K., S.S., D.K.)
| | - Chelsea K Ayers
- VA Evidence Synthesis Program, VA Portland Health Care System, Portland, Oregon (C.K.A., S.Y., J.A., S.V.)
| | - Karli K Kondo
- VA Evidence Synthesis Program, VA Portland Health Care System and Oregon Health & Science University, Portland, Oregon (K.M., K.K.K., S.S., D.K.)
| | - Somnath Saha
- VA Evidence Synthesis Program, VA Portland Health Care System and Oregon Health & Science University, Portland, Oregon (K.M., K.K.K., S.S., D.K.)
| | - Shailesh M Advani
- Social Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland (S.M.A.)
| | - Sarah Young
- VA Evidence Synthesis Program, VA Portland Health Care System, Portland, Oregon (C.K.A., S.Y., J.A., S.V.)
| | - Hunter Spencer
- Oregon Health & Science University, Portland, Oregon (H.S., M.R.)
| | - Max Rusek
- Oregon Health & Science University, Portland, Oregon (H.S., M.R.)
| | - Johanna Anderson
- VA Evidence Synthesis Program, VA Portland Health Care System, Portland, Oregon (C.K.A., S.Y., J.A., S.V.)
| | - Stephanie Veazie
- VA Evidence Synthesis Program, VA Portland Health Care System, Portland, Oregon (C.K.A., S.Y., J.A., S.V.)
| | - Mia Smith
- Oregon Health & Science University-Portland State University School of Public Health, Portland, Oregon (M.S.)
| | - Devan Kansagara
- VA Evidence Synthesis Program, VA Portland Health Care System and Oregon Health & Science University, Portland, Oregon (K.M., K.K.K., S.S., D.K.)
| |
Collapse
|
81
|
Screening of severe acute respiratory syndrome coronavirus-2 infection during labor and delivery using polymerase chain reaction and immunoglobulin testing. Life Sci 2021; 271:119200. [PMID: 33577855 PMCID: PMC7871853 DOI: 10.1016/j.lfs.2021.119200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
Aims To assess severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection during labor and delivery with polymerase chain reaction (PCR) and using immunoglobulin G and M testing to correlate with maternal and perinatal outcomes. Main methods Pregnant women admitted for labor and delivery at two Spanish hospitals were screened for SARS-CoV-2 infection by PCR test and by detection of serum immunoglobulins G and M. Maternal and perinatal outcomes were compared in women with laboratory evidence of SARS-CoV-2 infection with those with negative tests. Key findings Between March 31st and September 30th, 2020, 1211 pregnant women were screened for SARS-CoV-2 infection. The prevalence of laboratory evidence of SARS-CoV-2 infections was 5.4% (n = 65), corresponding to (i) 22 ongoing infections at admission, including two with mild clinical symptoms and 20 asymptomatic women; (ii) 43 cases of previous SARS-CoV-2 exposure; (iii) and 1146 women who were negative for both SARS-CoV-2 PCR and serological test. None of the screened mothers required hospital admission for coronavirus disease before or after delivery, nor were any of the newborns admitted to the intensive care unit. All newborns from mothers with positive PCR on admission were PCR negative. There were no significant differences in maternal or perinatal outcomes among the three studied groups. Significance Ongoing or previous SARS-CoV-2 infection with asymptomatic or mild clinical symptoms detected during screening in pregnant women at labor and delivery do not have a higher rate of adverse maternal or perinatal outcomes.
Collapse
|
82
|
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CA, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella L, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, The UPenn COVID Processing Unit, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer N, DeMichele AM, Vardhana SA, Mamtani R, Huang AC. CD8 T cells compensate for impaired humoral immunity in COVID-19 patients with hematologic cancer. RESEARCH SQUARE 2021:rs.3.rs-162289. [PMID: 33564756 PMCID: PMC7872363 DOI: 10.21203/rs.3.rs-162289/v1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer patients have increased morbidity and mortality from Coronavirus Disease 2019 (COVID-19), but the underlying immune mechanisms are unknown. In a cohort of 100 cancer patients hospitalized for COVID-19 at the University of Pennsylvania Health System, we found that patients with hematologic cancers had a significantly higher mortality relative to patients with solid cancers after accounting for confounders including ECOG performance status and active cancer status. We performed flow cytometric and serologic analyses of 106 cancer patients and 113 non-cancer controls from two additional cohorts at Penn and Memorial Sloan Kettering Cancer Center. Patients with solid cancers exhibited an immune phenotype similar to non-cancer patients during acute COVID-19 whereas patients with hematologic cancers had significant impairment of B cells and SARS-CoV-2-specific antibody responses. High dimensional analysis of flow cytometric data revealed 5 distinct immune phenotypes. An immune phenotype characterized by CD8 T cell depletion was associated with a high viral load and the highest mortality of 71%, among all cancer patients. In contrast, despite impaired B cell responses, patients with hematologic cancers and preserved CD8 T cells had a lower viral load and mortality. These data highlight the importance of CD8 T cells in acute COVID-19, particularly in the setting of impaired humoral immunity. Further, depletion of B cells with anti-CD20 therapy resulted in almost complete abrogation of SARS-CoV-2-specific IgG and IgM antibodies, but was not associated with increased mortality compared to other hematologic cancers, when adequate CD8 T cells were present. Finally, higher CD8 T cell counts were associated with improved overall survival in patients with hematologic cancers. Thus, CD8 T cells likely compensate for deficient humoral immunity and influence clinical recovery of COVID-19. These observations have important implications for cancer and COVID-19-directed treatments, immunosuppressive therapies, and for understanding the role of B and T cells in acute COVID-19.
Collapse
Affiliation(s)
- Erin M. Bange
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Nicholas A. Han
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Paul Wileyto
- Abramson Cancer Center, University of Pennsylvania
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania
| | - Justin Y. Kim
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - Allison R. Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Florence Porterfield
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Olutosin Owoyemi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Karan Naik
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Cathy Zheng
- Abramson Cancer Center, University of Pennsylvania
| | | | - Ariel R. Weisman
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Caroline A.G. Ittner
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Emily M. Kugler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Amy E. Baxter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Olutwatosin Oniyide
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Roseline S. Agyekum
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Thomas G. Dunn
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Tiffanie K. Jones
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Heather M. Giannini
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Madison E. Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - N. Esther Babady
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Adam J Widman
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | | | | | | | - Carla Wright
- Abramson Cancer Center, University of Pennsylvania
| | - Tara Perloff
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Josephine R. Giles
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Derek A. Oldridge
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Jennifer E. Wu
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Cécile Alanio
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Sharon Adamski
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Alfred L. Garfall
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Laura Vella
- Department of Pediatrics, Perelman School of Medicine, Children’s Hospital of Philadelphia
| | - Samuel J. Kerr
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Justine V. Cohen
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Randall A. Oyer
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Ryan Massa
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital
| | - Ivan P. Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | | | - Kara N. Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - John P. Reilly
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Peter G. Maslak
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Robert H. Vonderheide
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Jedd D. Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Scott E. Hensley
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Nuala Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Angela M. DeMichele
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Santosha A. Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Parker Institute for Cancer Immunotherapy
| | - Ronac Mamtani
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Alexander C. Huang
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| |
Collapse
|
83
|
Cavalcante MB, Cavalcante CTDMB, Sarno M, Barini R, Kwak-Kim J. Maternal immune responses and obstetrical outcomes of pregnant women with COVID-19 and possible health risks of offspring. J Reprod Immunol 2021; 143:103250. [PMID: 33249335 PMCID: PMC7676367 DOI: 10.1016/j.jri.2020.103250] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/30/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022]
Abstract
Coronavirus disease 2019 (COVID-19) pandemic has spread rapidly across the world. The vast majority of patients with COVID-19 manifest mild to moderate symptoms but may progress to severe cases or even mortalities. Young adults of reproductive age are the most affected population by SARS-CoV-2 infection. However, there is no consensus yet if pregnancy contributes to the severity of COVID-19. Initial studies of pregnant women have found that COVID-19 significantly increases the risk of preterm birth, intrauterine growth restriction, and low birth weight, which have been associated with non-communicable diseases in offspring. Besides, maternal viral infections with or without vertical transmission have been allied with neurological and behavioral disorders of the offspring. In this review, obstetrical outcomes of women with COVID-19 and possible risks for their offspring are discussed by reviewing maternal immune responses to COVID-19 based on the current evidence. Structural and systemic follow-up of offspring who are exposed to SARS-CoV-2 in-utero is suggested.
Collapse
Affiliation(s)
- Marcelo Borges Cavalcante
- Department of Obstetrics and Gynecology, Fortaleza University (UNIFOR), Fortaleza, CE, 60.811-905, Brazil; CONCEPTUS - Reproductive Medicine, Fortaleza, CE, 60.170-240, Brazil.
| | | | - Manoel Sarno
- Department of Obstetrics and Gynecology, Federal University of Bahia (UFBA), Salvador, BA, 40.026-010, Brazil; Harris Birthright Research Center for Fetal Medicine, "'King's College Hospital and Department of Fetal Medicine, University College, London, United Kingdom
| | - Ricardo Barini
- Department of Obstetrics and Gynecology, Campinas University (UNICAMP), Campinas, SP, 13.083-887, Brazil
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, 60061, USA
| |
Collapse
|
84
|
Nande A, Sheen J, Walters EL, Klein B, Chinazzi M, Gheorghe A, Adlam B, Shinnick J, Tejeda MF, Scarpino SV, Vespignani A, Greenlee AJ, Schneider D, Levy MZ, Hill AL. The effect of eviction moratoria on the transmission of SARS-CoV-2. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2020.10.27.20220897. [PMID: 33140067 PMCID: PMC7605580 DOI: 10.1101/2020.10.27.20220897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Massive unemployment during the COVID-19 pandemic could result in an eviction crisis in US cities. Here we model the effect of evictions on SARS-CoV-2 epidemics, simulating viral transmission within and among households in a theoretical metropolitan area. We recreate a range of urban epidemic trajectories and project the course of the epidemic under two counterfactual scenarios, one in which a strict moratorium on evictions is in place and enforced, and another in which evictions are allowed to resume at baseline or increased rates. We find, across scenarios, that evictions lead to significant increases in infections. Applying our model to Philadelphia using locally-specific parameters shows that the increase is especially profound in models that consider realistically heterogenous cities in which both evictions and contacts occur more frequently in poorer neighborhoods. Our results provide a basis to assess municipal eviction moratoria and show that policies to stem evictions are a warranted and important component of COVID-19 control.
Collapse
Affiliation(s)
- Anjalika Nande
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, 02138
| | - Justin Sheen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Emma L Walters
- Department of Urban and Regional Planning, University of Illinois at Urbana-Champaign, Champaign, IL 61820
| | - Brennan Klein
- Network Science Institute, Northeastern University, Boston, MA, USA
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, USA
| | - Matteo Chinazzi
- Network Science Institute, Northeastern University, Boston, MA, USA
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, USA
| | - Andrei Gheorghe
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, 02138
| | - Ben Adlam
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, 02138
| | - Julianna Shinnick
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Maria Florencia Tejeda
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | | | - Alessandro Vespignani
- Network Science Institute, Northeastern University, Boston, MA, USA
- Laboratory for the Modeling of Biological and Socio-technical Systems, Northeastern University, Boston, USA
| | - Andrew J Greenlee
- Department of Urban and Regional Planning, University of Illinois at Urbana-Champaign, Champaign, IL 61820
| | - Daniel Schneider
- Department of Urban and Regional Planning, University of Illinois at Urbana-Champaign, Champaign, IL 61820
| | - Michael Z Levy
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Alison L Hill
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA, 02138
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
85
|
la Cour Freiesleben N, Egerup P, Hviid KVR, Severinsen ER, Kolte AM, Westergaard D, Fich Olsen L, Prætorius L, Zedeler A, Christiansen AMH, Nielsen JR, Bang D, Berntsen S, Ollé-López J, Ingham A, Bello-Rodríguez J, Storm DM, Ethelberg-Findsen J, Hoffmann ER, Wilken-Jensen C, Jørgensen FS, Westh H, Jørgensen HL, Nielsen HS. SARS-CoV-2 in first trimester pregnancy: a cohort study. Hum Reprod 2021; 36:40-47. [PMID: 33145598 PMCID: PMC7665455 DOI: 10.1093/humrep/deaa311] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/16/2020] [Indexed: 12/22/2022] Open
Abstract
Study question Does maternal infection with SARS-CoV-2 in first trimester pregnancy have an impact on the fetal development as measured by nuchal translucency thickness and pregnancy loss? Summary answer Nuchal translucency thickness at the first trimester scan was not significantly different in pregnant women with versus without SARS-CoV-2 infection in early pregnancy and there was no significant increased risk of pregnancy loss in women with SARS-CoV-2 infection in the first trimester. What is known already Pregnant women are more vulnerable to viral infections. Previous coronavirus epidemics have been associated with increased maternal morbidity, mortality and adverse obstetric outcomes. Currently, no evidence exists regarding possible effects of SARS-CoV-2 in first trimester pregnancies. Study design, size, duration Cohort study of 1,019 women with a double test taken between Feb. 17 and Apr. 23, 2020, as a part of the combined first trimester risk assessment, and 36 women with a first trimester pregnancy loss between Apr. 14 and May 21, 2020, prior to the double test. The study period was during the first SARS-CoV-2 epidemic wave in Denmark. Participants/materials, setting, methods Cohort 1 included pregnant women with a double test taken within the study period. The excess serum from each double test was analyzed for SARS-CoV-2 antibodies. Results were correlated to the nuchal translucency thickness and the number of pregnancy losses before or at the time of the first trimester scan. Cohort 2 included women with a pregnancy loss before the gestational age for double test sample. Serum from a blood test taken the day the pregnancy loss was identified was analyzed for SARS-CoV-2 antibodies. The study was conducted at a public university hospital serving approximately 12% of pregnant women and births in Denmark. All participants in the study provided written informed consent. Main results and the role of chance Eighteen (1.8%) women had SARS-CoV-2 antibodies in the serum from the double test suggestive of SARS-CoV-2 infection in early pregnancy. There was no significant difference in nuchal translucency thickness for women testing positive for previous SARS-CoV-2 infection (n = 18) versus negative (n = 994) (p = 0.62). There was no significant increased risk of pregnancy loss for women with positive antibodies (n = 1) (OR 3.4, 0.08-24.3 95% CI, p = 0.27). None of the women had been hospitalized due to SARS-CoV-2 infection. None of the women with pregnancy loss prior to the double test (Cohort 2) had SARS-CoV-2 antibodies. Limitations, reasons for caution These results may only apply to similar populations and to patients who do not require hospitalization due to SARS-CoV-2 infection. A limitation of the study is that only 1.8% of the study population had SARS-CoV-2 antibodies suggestive of previous infection. Wider implication of the findings Maternal SARS-CoV-2 infection had no effect on the nuchal translucency thickness and there was no significant increased risk of pregnancy loss for women with SARS-CoV-2 infection in first trimester pregnancy. Evidence concerning Covid-19 in pregnancy is still limited. These data indicate that infection with SARS-CoV-2 in not hospitalized women does not pose a significant threat in first trimester pregnancies. Follow up studies are needed to establish any risk to a fetus exposed to maternal SARS-CoV-2 infection. Study funding/competing interest(s) Prof. Henriette Svarre Nielsen (HSN) and colleagues received a grant from the Danish Government for research of Covid-19 among pregnant women. The Danish government was not involved in the study design, data collection, analysis, interpretation of data, writing of the report or decision to submit the paper for publication. AI, JOL, JBR, DMS, JEF, and ERH received funding from a Novo Nordisk Foundation (NNF) Young Investigator Grant (NNF15OC0016662) and a Danish National Science Foundation Center Grant (6110-00344B). AI received a Novo Scholarship. JOL is funded by an NNF Pregraduate Fellowship (NNF19OC0058982). DW is funded by the NNF (NNF18SA0034956, NNF14CC0001, NNF17OC0027594). AMK is funded by a grant from the Rigshospitalet’s research fund. Henriette Svarre Nielsen has received speakeŕs fees from Ferring Pharmaceuticals, Merck Denmark A/S and Ibsa Nordic (outside the submitted work). Nina la Cour Freiesleben has received a grant from Gedeon Richter (outside the submitted work). Astrid Marie Kolte has received speakeŕs from Merck (outside the submitted work). The other authors did not report any potential conflicts of interest.
Collapse
Affiliation(s)
- N la Cour Freiesleben
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - P Egerup
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - K V R Hviid
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - E R Severinsen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - A M Kolte
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.,The Recurrent Pregnancy Loss Unit, The Capital Region, Copenhagen University Hospitals Rigshospitalet & Hvidovre Hospital, Denmark
| | - D Westergaard
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark.,Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Methods and Analysis, Statistics Denmark, DK-2100, Copenhagen, Denmark
| | - L Fich Olsen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - L Prætorius
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - A Zedeler
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - A-M H Christiansen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - J R Nielsen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - D Bang
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - S Berntsen
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - J Ollé-López
- DNRF Center for Chromosome Stability (CCS), Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - A Ingham
- DNRF Center for Chromosome Stability (CCS), Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - J Bello-Rodríguez
- DNRF Center for Chromosome Stability (CCS), Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - D M Storm
- DNRF Center for Chromosome Stability (CCS), Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - J Ethelberg-Findsen
- DNRF Center for Chromosome Stability (CCS), Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - E R Hoffmann
- DNRF Center for Chromosome Stability (CCS), Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - C Wilken-Jensen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - F S Jørgensen
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Fetal Medicine Unit, Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - H Westh
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - H L Jørgensen
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - H S Nielsen
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark.,The Recurrent Pregnancy Loss Unit, The Capital Region, Copenhagen University Hospitals Rigshospitalet & Hvidovre Hospital, Denmark
| |
Collapse
|
86
|
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. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.15.21249810. [PMID: 33469597 PMCID: PMC7814843 DOI: 10.1101/2021.01.15.21249810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
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. To gauge the effectiveness of these measures at the University of Pennsylvania, we conducted a longitudinal study of SARS-CoV-2 antibody seropositivity and seroconversion in patients presenting to infusion centers for cancer-directed therapy between 5/21/2020 and 10/8/2020. Participants completed questionnaires and had up to five serial blood collections. 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 0.0-4.1%) over 14.8 person-years of follow up, with a median of 13 healthcare visits per patient. These results suggest that cancer patients receiving in-person care at a facility with aggressive mitigation efforts have an extremely low likelihood of COVID-19 infection.
Collapse
|
87
|
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Antibodies at Delivery in Women, Partners, and Newborns. Obstet Gynecol 2021; 137:49-55. [PMID: 33116054 DOI: 10.1097/aog.0000000000004199] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the frequency of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies in parturient women, their partners, and their newborns and the association of such antibodies with obstetric and neonatal outcomes. METHODS From April 4 to July 3, 2020, in a single university hospital in Denmark, all parturient women and their partners were invited to participate in the study, along with their newborns. Participating women and partners had a pharyngeal swab and a blood sample taken at admission; immediately after delivery, a blood sample was drawn from the umbilical cord. The swabs were analyzed for SARS-CoV-2 RNA by polymerase chain reaction, and the blood samples were analyzed for SARS-CoV-2 antibodies. Full medical history and obstetric and neonatal information were available. RESULTS A total of 1,313 parturient women (72.5.% of all women admitted for delivery at the hospital in the study period), 1,188 partners, and 1,206 newborns participated in the study. The adjusted serologic prevalence was 2.6% in women and 3.5% in partners. Seventeen newborns had SARS-CoV-2 immunoglobulin G (IgG) antibodies, and none had immunoglobulin M antibodies. No associations between SARS-CoV-2 antibodies and obstetric or neonatal complications were found (eg, preterm birth, preeclampsia, cesarean delivery, Apgar score, low birth weight, umbilical arterial pH, need for continuous positive airway pressure, or neonatal admission), but statistical power to detect such differences was low. Full serologic data from 1,051 families showed an absolute risk of maternal infection of 39% if the partner had antibodies. CONCLUSION We found no association between SARS-CoV-2 infection and obstetric or neonatal complications. Sixty-seven percent of newborns delivered by mothers with antibodies had SARS-CoV-2 IgG antibodies. A limitation of our study is that we lacked statistical power to detect small but potentially meaningful differences between those with and without evidence of infection.
Collapse
|
88
|
Brüssow H. COVID-19 by numbers - infections, cases and deaths. Environ Microbiol 2021; 23:1322-1333. [PMID: 33368993 DOI: 10.1111/1462-2920.15377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 01/28/2023]
Abstract
During the COVID-19 pandemic, governments face difficult decisions when being confronted with public health threats and economic needs. Decision making is further complicated by the different perception of the pandemic by the public. Politicians as well as the public need objective facts for guidance and numbers play here a crucial role. The current contribution compiles numbers for infections, cases and deaths with SARS-CoV-2 to serve as an orientation help.
Collapse
Affiliation(s)
- Harald Brüssow
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| |
Collapse
|
89
|
Hicks SM, Pohl K, Neeman T, McNamara HA, Parsons KM, He JS, Ali SA, Nazir S, Rowntree LC, Nguyen THO, Kedzierska K, Doolan DL, Vinuesa CG, Cook MC, Coatsworth N, Myles PS, Kurth F, Sander LE, Mann GJ, Gruen RL, George AJ, Gardiner EE, Cockburn IA, SARS-CoV-2 Testing in Elective Surgery Collaborators. A Dual-Antigen Enzyme-Linked Immunosorbent Assay Allows the Assessment of Severe Acute Respiratory Syndrome Coronavirus 2 Antibody Seroprevalence in a Low-Transmission Setting. J Infect Dis 2021; 223:10-14. [PMID: 33009908 PMCID: PMC7665523 DOI: 10.1093/infdis/jiaa623] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/29/2020] [Indexed: 01/07/2023] Open
Abstract
Estimates of seroprevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies have been hampered by inadequate assay sensitivity and specificity. Using an enzyme-linked immunosorbent assay-based approach that combines data about immunoglobulin G responses to both the nucleocapsid and spike receptor binding domain antigens, we show that excellent sensitivity and specificity can be achieved. We used this assay to assess the frequency of virus-specific antibodies in a cohort of elective surgery patients in Australia and estimated seroprevalence in Australia to be 0.28% (95% Confidence Interval, 0-1.15%). These data confirm the low level of transmission of SARS-CoV-2 in Australia before July 2020 and validate the specificity of our assay.
Collapse
Affiliation(s)
- Sarah M Hicks
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Kai Pohl
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitatsmedizin Berlin, Berlin, Germany
| | - Teresa Neeman
- Biological Data Science Institute, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Hayley A McNamara
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Kate M Parsons
- Australian National University Centre for Therapeutic Discovery, The Australian National University, Canberra, Australia
| | - Jin-shu He
- Australian National University Centre for Therapeutic Discovery, The Australian National University, Canberra, Australia
| | - Sidra A Ali
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Samina Nazir
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Carola G Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- China Australia Centre for Personalised Immunology, Shanghai Renji Hospital, Jiaotong University, Shanghai, China
- Department of Immunology Canberra Hospital, Canberra, Australia
| | - Matthew C Cook
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Department of Immunology Canberra Hospital, Canberra, Australia
| | - Nicholas Coatsworth
- ANU Medical School, Australian National University, Canberra, Australia
- The Canberra Hospital, Infectious Diseases, Canberra, Australia
| | - Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, Alfred Hospital, Melbourne, Australia
- Department of Anaesthesiology and Perioperative Medicine, Monash University, Melbourne, Australia
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitatsmedizin Berlin, Berlin, Germany
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leif E Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitatsmedizin Berlin, Berlin, Germany
| | - Graham J Mann
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Russell L Gruen
- College of Health and Medicine, The Australian National University, Canberra, Australia
| | - Amee J George
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Australian National University Centre for Therapeutic Discovery, The Australian National University, Canberra, Australia
| | - Elizabeth E Gardiner
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | | |
Collapse
|
90
|
Joseph NT, Rasmussen SA, Jamieson DJ. The effects of COVID-19 on pregnancy and implications for reproductive medicine. Fertil Steril 2021; 115:824-830. [PMID: 33676752 PMCID: PMC7775651 DOI: 10.1016/j.fertnstert.2020.12.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
COVID-19 was officially declared a pandemic in March 2020. Since then, our understanding of its effects on pregnancy have evolved rapidly. Emerging surveillance data and large cohort studies suggest that pregnancy is associated with an increased risk of intensive care unit hospitalization, invasive ventilation, and death. Pregnancies complicated by SARS-CoV-2 infection are associated with increased likelihood of cesarean delivery and preterm birth. Intrauterine transmission occurs, but seems to be rare. Critical gaps remain, and rigorous high-quality data are needed to better ascertain pregnancy risks and to inform antenatal and obstetrical management.
Collapse
Affiliation(s)
- Naima T Joseph
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Sonja A Rasmussen
- Departments of Pediatrics, Obstetrics and Gynecology, and Epidemiology, University of Florida College of Medicine and College of Public Health and Health Professions, Gainesville, Florida
| | - Denise J Jamieson
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
91
|
Uyoga S, Adetifa IMO, Karanja HK, Nyagwange J, Tuju J, Wanjiku P, Aman R, Mwangangi M, Amoth P, Kasera K, Ng'ang'a W, Rombo C, Yegon C, Kithi K, Odhiambo E, Rotich T, Orgut I, Kihara S, Otiende M, Bottomley C, Mupe ZN, Kagucia EW, Gallagher KE, Etyang A, Voller S, Gitonga JN, Mugo D, Agoti CN, Otieno E, Ndwiga L, Lambe T, Wright D, Barasa E, Tsofa B, Bejon P, Ochola-Oyier LI, Agweyu A, Scott JAG, Warimwe GM. Seroprevalence of anti-SARS-CoV-2 IgG antibodies in Kenyan blood donors. Science 2021; 371:79-82. [PMID: 33177105 PMCID: PMC7877494 DOI: 10.1126/science.abe1916] [Citation(s) in RCA: 193] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022]
Abstract
The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Africa is poorly described. The first case of SARS-CoV-2 in Kenya was reported on 12 March 2020, and an overwhelming number of cases and deaths were expected, but by 31 July 2020, there were only 20,636 cases and 341 deaths. However, the extent of SARS-CoV-2 exposure in the community remains unknown. We determined the prevalence of anti-SARS-CoV-2 immunoglobulin G among blood donors in Kenya in April-June 2020. Crude seroprevalence was 5.6% (174 of 3098). Population-weighted, test-performance-adjusted national seroprevalence was 4.3% (95% confidence interval, 2.9 to 5.8%) and was highest in urban counties Mombasa (8.0%), Nairobi (7.3%), and Kisumu (5.5%). SARS-CoV-2 exposure is more extensive than indicated by case-based surveillance, and these results will help guide the pandemic response in Kenya and across Africa.
Collapse
Affiliation(s)
- Sophie Uyoga
- 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, Keppel Street, London, UK
| | | | | | - James Tuju
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Rashid Aman
- Ministry of Health, Government of Kenya, Nairobi, Kenya
| | | | - Patrick Amoth
- Ministry of Health, Government of Kenya, Nairobi, Kenya
| | | | - Wangari Ng'ang'a
- Presidential Policy and Strategy Unit, The Presidency, Government of Kenya, Nairobi, Kenya
| | - Charles Rombo
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Christine Yegon
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Khamisi Kithi
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Elizabeth Odhiambo
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Thomas Rotich
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Irene Orgut
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Sammy Kihara
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Mark Otiende
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Christian Bottomley
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - Zonia N Mupe
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Katherine E Gallagher
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | | | - Shirine Voller
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | | | - Daisy Mugo
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Edward Otieno
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Teresa Lambe
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Daniel Wright
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Edwine Barasa
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| | | | | | - J Anthony G Scott
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - George M Warimwe
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| |
Collapse
|
92
|
Haizler-Cohen L, Davidov A, Blitz MJ, Fruhman G. Severe acute respiratory syndrome coronavirus 2 antibodies in pregnant women admitted to labor and delivery units. Am J Obstet Gynecol 2021; 224:112-114. [PMID: 32971014 PMCID: PMC7503125 DOI: 10.1016/j.ajog.2020.09.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/13/2020] [Accepted: 09/18/2020] [Indexed: 12/19/2022]
|
93
|
Uyoga S, Adetifa IMO, Karanja HK, Nyagwange J, Tuju J, Wanjiku P, Aman R, Mwangangi M, Amoth P, Kasera K, Ng'ang'a W, Rombo C, Yegon C, Kithi K, Odhiambo E, Rotich T, Orgut I, Kihara S, Otiende M, Bottomley C, Mupe ZN, Kagucia EW, Gallagher KE, Etyang A, Voller S, Gitonga JN, Mugo D, Agoti CN, Otieno E, Ndwiga L, Lambe T, Wright D, Barasa E, Tsofa B, Bejon P, Ochola-Oyier LI, Agweyu A, Scott JAG, Warimwe GM. Seroprevalence of anti-SARS-CoV-2 IgG antibodies in Kenyan blood donors. Science 2021; 371:79-82. [PMID: 33177105 DOI: 10.1101/2020.07.27.20162693] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/06/2020] [Indexed: 05/24/2023]
Abstract
The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Africa is poorly described. The first case of SARS-CoV-2 in Kenya was reported on 12 March 2020, and an overwhelming number of cases and deaths were expected, but by 31 July 2020, there were only 20,636 cases and 341 deaths. However, the extent of SARS-CoV-2 exposure in the community remains unknown. We determined the prevalence of anti-SARS-CoV-2 immunoglobulin G among blood donors in Kenya in April-June 2020. Crude seroprevalence was 5.6% (174 of 3098). Population-weighted, test-performance-adjusted national seroprevalence was 4.3% (95% confidence interval, 2.9 to 5.8%) and was highest in urban counties Mombasa (8.0%), Nairobi (7.3%), and Kisumu (5.5%). SARS-CoV-2 exposure is more extensive than indicated by case-based surveillance, and these results will help guide the pandemic response in Kenya and across Africa.
Collapse
Affiliation(s)
- Sophie Uyoga
- 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, Keppel Street, London, UK
| | | | | | - James Tuju
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Rashid Aman
- Ministry of Health, Government of Kenya, Nairobi, Kenya
| | | | - Patrick Amoth
- Ministry of Health, Government of Kenya, Nairobi, Kenya
| | | | - Wangari Ng'ang'a
- Presidential Policy and Strategy Unit, The Presidency, Government of Kenya, Nairobi, Kenya
| | - Charles Rombo
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Christine Yegon
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Khamisi Kithi
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Elizabeth Odhiambo
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Thomas Rotich
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Irene Orgut
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Sammy Kihara
- Kenya National Blood Transfusion Services, Ministry of Health, Nairobi, Kenya
| | - Mark Otiende
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Christian Bottomley
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - Zonia N Mupe
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Katherine E Gallagher
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | | | - Shirine Voller
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | | | - Daisy Mugo
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Edward Otieno
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Teresa Lambe
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Daniel Wright
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Edwine Barasa
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| | | | | | - J Anthony G Scott
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - George M Warimwe
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Oxford University, Oxford, UK
| |
Collapse
|
94
|
Evidence of thrombotic microangiopathy in children with SARS-CoV-2 across the spectrum of clinical presentations. Blood Adv 2020; 4:6051-6063. [PMID: 33290544 PMCID: PMC7724906 DOI: 10.1182/bloodadvances.2020003471] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022] Open
Abstract
Most children with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have mild or minimal disease, with a small proportion developing severe disease or multisystem inflammatory syndrome in children (MIS-C). Complement-mediated thrombotic microangiopathy (TMA) has been associated with SARS-CoV-2 infection in adults but has not been studied in the pediatric population. We hypothesized that complement activation plays an important role in SARS-CoV-2 infection in children and sought to understand if TMA was present in these patients. We enrolled 50 hospitalized pediatric patients with acute SARS-CoV-2 infection (n = 21, minimal coronavirus disease 2019 [COVID-19]; n = 11, severe COVID-19) or MIS-C (n = 18). As a biomarker of complement activation and TMA, soluble C5b9 (sC5b9, normal 247 ng/mL) was measured in plasma, and elevations were found in patients with minimal disease (median, 392 ng/mL; interquartile range [IQR], 244-622 ng/mL), severe disease (median, 646 ng/mL; IQR, 203-728 ng/mL), and MIS-C (median, 630 ng/mL; IQR, 359-932 ng/mL) compared with 26 healthy control subjects (median, 57 ng/mL; IQR, 9-163 ng/mL; P < .001). Higher sC5b9 levels were associated with higher serum creatinine (P = .01) but not age. Of the 19 patients for whom complete clinical criteria were available, 17 (89%) met criteria for TMA. A high proportion of tested children with SARS-CoV-2 infection had evidence of complement activation and met clinical and diagnostic criteria for TMA. Future studies are needed to determine if hospitalized children with SARS-CoV-2 should be screened for TMA, if TMA-directed management is helpful, and if there are any short- or long-term clinical consequences of complement activation and endothelial damage in children with COVID-19 or MIS-C.
Collapse
|
95
|
Pothineni NVK, Starkey S, Conn K, Evans C, Shah R, Hyman MC, Frankel DS, Halaby R, Johnston-Cox HA, Kunkel K, Nathan AS, Seigerman ME, Herrmann HC, Giri J, Marchlinski FE, Santangeli P, Fanaroff AC. Patient and Staff Perceptions of Universal Severe Acute Respiratory Syndrome Coronavirus 2 Screening Prior to Cardiac Catheterization and Electrophysiology Laboratory Procedures. Circ Cardiovasc Interv 2020; 13:e009975. [PMID: 33272035 PMCID: PMC7732150 DOI: 10.1161/circinterventions.120.009975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Naga Venkata K. Pothineni
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Samantha Starkey
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Kristine Conn
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Christina Evans
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Ronak Shah
- Department of Anesthesiology and Critical Care (R.S., M.C.H., D.S.F., R.H., H.A.J.-C.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Matthew C. Hyman
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Department of Anesthesiology and Critical Care (R.S., M.C.H., D.S.F., R.H., H.A.J.-C.), University of Pennsylvania, Philadelphia
| | - David S. Frankel
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Department of Anesthesiology and Critical Care (R.S., M.C.H., D.S.F., R.H., H.A.J.-C.), University of Pennsylvania, Philadelphia
| | - Rim Halaby
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Department of Anesthesiology and Critical Care (R.S., M.C.H., D.S.F., R.H., H.A.J.-C.), University of Pennsylvania, Philadelphia
| | - Hillary A. Johnston-Cox
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Department of Anesthesiology and Critical Care (R.S., M.C.H., D.S.F., R.H., H.A.J.-C.), University of Pennsylvania, Philadelphia
| | - Katherine Kunkel
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Ashwin S. Nathan
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Cardiovascular Outcomes Quality and Evaluative Research Center and Leonard Davis Institute (A.S.N., J.G., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Matthew E. Seigerman
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Howard C. Herrmann
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Jay Giri
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Cardiovascular Outcomes Quality and Evaluative Research Center and Leonard Davis Institute (A.S.N., J.G., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Francis E. Marchlinski
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Pasquale Santangeli
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| | - Alexander C. Fanaroff
- Division of Cardiovascular Medicine (N.V.K.P., M.C.H., D.S.F., R.H., H.A.J.-C., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.), University of Pennsylvania, Philadelphia
- Cardiovascular Outcomes Quality and Evaluative Research Center and Leonard Davis Institute (A.S.N., J.G., A.C.F.), University of Pennsylvania, Philadelphia
- Penn Heart and Vascular Center, Philadelphia, PA (N.V.K.P., S.S., K.C., C.E., R.S., K.K., A.S.N., M.E.S., H.C.H., J.G., F.E.M., P.S., A.C.F.)
| |
Collapse
|
96
|
Anderson EM, Diorio C, Goodwin EC, McNerney KO, Weirick ME, Gouma S, Bolton MJ, Arevalo CP, Chase J, Hicks P, Manzoni TB, Baxter AE, Andrea KP, Burudpakdee C, Lee JH, Vella LA, Henrickson SE, Harris RM, Wherry EJ, Bates P, Bassiri H, Behrens EM, Teachey DT, Hensley SE. Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) Antibody Responses in Children With Multisystem Inflammatory Syndrome in Children (MIS-C) and Mild and Severe Coronavirus Disease 2019 (COVID-19). J Pediatric Infect Dis Soc 2020; 10:669-673. [PMID: 33263756 PMCID: PMC7799010 DOI: 10.1093/jpids/piaa161] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) antibody responses in children remain poorly characterized. Here, we show that pediatric patients with multisystem inflammatory syndrome in children (MIS-C) possess higher SARS-CoV-2 spike immunoglobulin G (IgG) titers compared with those with severe coronavirus disease 2019, likely reflecting a longer time since the onset of infection in MIS-C patients.
Collapse
Affiliation(s)
- Elizabeth M Anderson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Caroline Diorio
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA,Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eileen C Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Kevin O McNerney
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA,Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Madison E Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Marcus J Bolton
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Claudia P Arevalo
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Julie Chase
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA,Division of Rheumatology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip Hicks
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA,School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Tomaz B Manzoni
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Amy E Baxter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Kurt P Andrea
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chakkapong Burudpakdee
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica H Lee
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura A Vella
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA,Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah E Henrickson
- Division of Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rebecca M Harris
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA,Penn Center for Research on Coronavirus and Other Emerging Pathogens, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Hamid Bassiri
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA,Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Edward M Behrens
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA,Division of Rheumatology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David T Teachey
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA,Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA,Correspondence:
| |
Collapse
|
97
|
Ogunwole SM, Bennett WL, Williams AN, Bower KM. Community-Based Doulas and COVID-19: Addressing Structural and Institutional Barriers to Maternal Health Equity. PERSPECTIVES ON SEXUAL AND REPRODUCTIVE HEALTH 2020; 52:199-204. [PMID: 33399272 DOI: 10.1363/psrh.12169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 05/06/2023]
Affiliation(s)
- S Michelle Ogunwole
- Division of General Internal Medicine, School of Medicine, Johns Hopkins University, Baltimore
| | - Wendy L Bennett
- Division of General Internal Medicine, School of Medicine, Johns Hopkins University, Baltimore
| | - Andrea N Williams
- Nzuri Malkia Birth Cooperative and Baltimore Community Doulas, Baltimore
| | - Kelly M Bower
- School of Nursing and Johns Hopkins Center for Health Disparities Solutions, Johns Hopkins University, Baltimore
| |
Collapse
|
98
|
Menezes MO, Takemoto MLS, Nakamura‐Pereira M, Katz L, Amorim MMR, Salgado HO, Melo A, Diniz CSG, de Sousa LAR, Magalhaes CG, Knobel R, Andreucci CB, Brazilian Group of Studies for COVID‐19, Pregnancy. Risk factors for adverse outcomes among pregnant and postpartum women with acute respiratory distress syndrome due to COVID-19 in Brazil. Int J Gynaecol Obstet 2020; 151:415-423. [PMID: 33011966 PMCID: PMC9087686 DOI: 10.1002/ijgo.13407] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/04/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To evaluate whether clinical and social risk factors are associated with negative outcomes for COVID-19 disease among Brazilian pregnant and postpartum women. METHODS A secondary analysis was conducted of the official Acute Respiratory Syndrome Surveillance System database. Pregnant and postpartum women diagnosed with COVID-19 ARDS until July 14, 2020, were included. Adverse outcomes were a composite endpoint of either death, admission to the intensive care unit (ICU), or mechanical ventilation. Risk factors were examined by multiple logistic regression. RESULTS There were 2475 cases of COVID-19 ARDS. Among them, 23.8% of women had the composite endpoint and 8.2% died. Of those who died, 5.9% were not hospitalized, 39.7% were not admitted to the ICU, 42.6% did not receive mechanical ventilation, and 25.5% did not have access to respiratory support. Multivariate analysis showed that postpartum period, age over 35 years, obesity, diabetes, black ethnicity, living in a peri-urban area, no access to Family Health Strategy, or living more than 100 km from the notification hospital were associated with an increased risk of adverse outcomes. CONCLUSION Clinical and social risk factors and barriers to access health care are associated with adverse outcomes among maternal cases of COVID-19 ARDS in Brazil.
Collapse
Affiliation(s)
| | | | - Marcos Nakamura‐Pereira
- Instituto Nacional de Saúde da Mulherda Criança e do Adolescente Fernandes FigueiraFundação Oswaldo CruzRio de JaneiroRJBrazil
| | - Leila Katz
- Instituto de Medicina Integral Prof. Fernando Figueira (IMIP)RecifePEBrazil
| | | | - Heloisa O. Salgado
- Departamento de Medicina SocialFaculdade de Medicina de Ribeirão PretoUniversidade de São Paulo (USP)Ribeirão PretoSPBrazil
| | - Adriana Melo
- Universidade Federal de Campina Grande (UFCG)Campina GrandePBBrazil
| | - Carmen S. G. Diniz
- Department of HealthLife Cycles and SocietySchool of Public HealthUniversity of São Paulo (USP)São PauloSPBrazil
| | | | | | - Roxana Knobel
- Department of Gynecology and ObstetricsUniversidade Federal de Santa Catarina (UFSC)FlorianópolisSCBrazil
| | - Carla B. Andreucci
- Department of MedicineUniversidade Federal de São Carlos (UFSCAR)São CarlosSPBrazil
| | | |
Collapse
|
99
|
Savirón-Cornudella R, Villalba A, Zapardiel J, Andeyro-Garcia M, Esteban LM, Pérez-López FR. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) universal screening in gravids during labor and delivery. Eur J Obstet Gynecol Reprod Biol 2020; 256:400-404. [PMID: 33285496 PMCID: PMC7706716 DOI: 10.1016/j.ejogrb.2020.11.069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/08/2020] [Accepted: 11/30/2020] [Indexed: 12/25/2022]
Abstract
Objective To screen pregnant women at risk of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during delivery using reverse-transcription polymerase chain reaction (RT-PCR) test and serum immunoglobulin (Ig) testing. Method Between March 31 st and August 31 st of 2020, consecutive pregnant women admitted for labor and delivery in a single hospital were screened for SARS-CoV-2 with nasopharyngeal RT-PCR swab tests and detection of serum IgG and IgM. Results We studied 266 pregnant women admitted for labor and delivery. The prevalence of acute or past SARS-CoV-2 infection was 9.0 %, including (i) two cases with respiratory symptoms of SARS-Co-V-2 infection and positive RT-PCR; (ii) four asymptomatic women with positive RT-PCR without clinical symptoms and negative serological tests between two and 15 weeks later; and (iii) two women with false positive RT-PCR due to technical problems. All newborns of the 6 pregnant women with RT-PCR positive had negative RT-PCR and did not require Neonatal Intensive Care Unit admission. There were eighteen asymptomatic women with positive serological IgG tests and negative RT-PCR. Conclusion In our cohort of gravids, we found 2.2 % of women with positive RT-PRC tests and 6.7 % with positive serological tests during the first wave of the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
| | - Ana Villalba
- Department of Obstetrics and Gynecology, Hospital Universitario General de Villalba, Madrid, Spain
| | - Javier Zapardiel
- Department of Microbiology, Hospital Universitario General de Villalba, Madrid, Spain
| | - Mercedes Andeyro-Garcia
- Department of Obstetrics and Gynecology, Hospital Universitario General de Villalba, Madrid, Spain
| | - Luis M Esteban
- Escuela Universitaria Politécnica de La Almunia, Universidad de Zaragoza, Zaragoza, Spain
| | - Faustino R Pérez-López
- Instituto de Investigación Sanitaria de Aragón and University of Zaragoza, Faculty of Medicine, Zaragoza, Spain
| |
Collapse
|
100
|
Root H, Boyce R, Robinson WR. Learning from LMICs: best practices for leveraging sentinel surveillance systems to track the US COVID-19 pandemic. BMJ Glob Health 2020; 5:e004685. [PMID: 33380417 PMCID: PMC7780421 DOI: 10.1136/bmjgh-2020-004685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 01/29/2023] Open
Affiliation(s)
- Heather Root
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Ross Boyce
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Whitney R Robinson
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
- University of North Carolina at Chapel Hill Carolina Population Center, Chapel Hill, North Carolina, USA
| |
Collapse
|