1
|
Pernet O, Frederick T, Adili A, Hudgins J, Anthony P, McCaney G, Mack WJ, Noriega E, Lopez J, Balog S, Biniwale M, Yeh A, Bearden A, Ramanathan R, Kovacs A. Cord blood IgA/M reveals in utero response to SARS-CoV-2 with fluctuations in relation to circulating variants. Nat Commun 2025; 16:3551. [PMID: 40229328 PMCID: PMC11997084 DOI: 10.1038/s41467-025-58768-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
It is estimated that in utero SARS-CoV-2 infection is rare. However, few studies have systematically assessed for IgA and IgM antibodies indicating potential in utero response to SARS-CoV-2 infection using multi-isotype serology, and no studies have assessed in utero infection markers in relation to circulating variants. Between October 21, 2021 and February 15, 2023, remnant cord blood samples (CBS) from neonates born at a single hospital in Los Angeles, were systematically tested for serological markers suggesting in utero infection. SARS-CoV-2 specific fetal IgA and/or IgM antibodies were detected in 28.7% (298/1038 CBS, 95% CI: 26.0, 31.6), higher than previous in utero infection estimates that used only PCR and/or IgM. Importantly, the probability of detecting markers of in utero infection varied by month (P-value = 0.0144). The prevalence of fetal IgA/IgM varied with the emergence of new variants, increasing during the BA.1 wave with a peak in February 2022 at 36% (18/50, 95% CI: 22.7-49.3) and again during the BA.4/5 wave, with a peak at 48.8% in September 2022 (39/80, 95% CI 37.8-59.7), suggesting variant-related fluctuations. These data suggest it may be useful to identify SARS-Cov-2 in utero exposure at birth so these newborns may be more closely followed for adverse clinical outcomes.
Collapse
Affiliation(s)
- Olivier Pernet
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA.
| | - Toinette Frederick
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Amila Adili
- Southern California Clinical and Translational Science Institute (SC-CTSI), University of Southern California, Los Angeles, CA, USA
| | - Jay Hudgins
- Department of Pathology and Laboratory Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Patricia Anthony
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Gwyndolyn McCaney
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Wendy J Mack
- Southern California Clinical and Translational Science Institute (SC-CTSI), University of Southern California, Los Angeles, CA, USA
- Department of Population and Public Health Sciences, University of Southern California, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Eunice Noriega
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Jennifer Lopez
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Steven Balog
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Manoj Biniwale
- Division of Neonatal Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Amy Yeh
- Division of Neonatal Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Allison Bearden
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Rangasamy Ramanathan
- Division of Neonatal Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Andrea Kovacs
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA.
- Department of Population and Public Health Sciences, University of Southern California, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Chen JC, Hsu MH, Kuo RL, Wang LT, Kuo ML, Tseng LY, Chang HL, Chiu CH. mRNA-1273 is placenta-permeable and immunogenic in the fetus. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102489. [PMID: 40104112 PMCID: PMC11919431 DOI: 10.1016/j.omtn.2025.102489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025]
Abstract
COVID-19 mRNA vaccines are generally recognized as safe for gestational administration. However, their transplacental pharmacokinetics remain obscure. In this study, mRNA-1273 intramuscularly given to pregnant mice rapidly circulated in maternal blood and crossed the placenta within 1 h to spread in the fetal circulation. Although spike mRNA in fetal circulation faded away within 4-6 h, it could accumulate in fetal tissues, mainly the liver and get translated into spike protein. Transplacental mRNA-1273 proved immunogenic in the fetuses, as postnatally equipped with anti-spike immunoglobulin (Ig)M, paternal allotypic anti-spike IgG2a, and heightened anti-spike cellular immunity. Gestationally administered, mRNA-1273 had a dose-dependent effect on its transplacental transfer and immunogenicity in the fetuses, with higher mRNA-1273 doses leading to increased transplacental mRNA-1273 passage and greater serum titers of endogenous anti-spike IgM/IgG generated by the fetuses. Thus, gestationally maternal mRNA-1273 vaccination might endow the newborns with not only passive but also active anti-spike immunity. Our results pose new insights into transplacental capacity of mRNA vaccines and their immunogenic potential in the fetuses, advancing our knowledge of mRNA medicine to protect the unborns against pathogens in perinatal life and broaden our horizons of prenatal mRNA molecular therapy.
Collapse
Affiliation(s)
- Jeng-Chang Chen
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Division of Pediatric Surgery, Department of Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Mei-Hua Hsu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Rei-Lin Kuo
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Li-Ting Wang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ming-Ling Kuo
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Li-Yun Tseng
- Pediatric Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Hsueh-Ling Chang
- Pediatric Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Cheng-Hsun Chiu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
3
|
Makan-Murphy N, Madhi SA, Dangor Z. Safety, Efficacy, and Effectiveness of Maternal Vaccination against Respiratory Infections in Young Infants. Semin Respir Crit Care Med 2024. [PMID: 39708836 DOI: 10.1055/a-2471-6906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2024]
Abstract
Lower respiratory tract infection (LRTI) is a major cause of neonatal morbidity and mortality worldwide. Maternal vaccination is an effective strategy in protecting young infants from LRTI, particularly in the first few months after birth when infant is most vulnerable, and most primary childhood vaccinations have not been administered. Additionally, maternal vaccination protects the mother from illness during pregnancy and the postnatal period, and the developing fetus from adverse outcomes such as stillbirth and prematurity. In this paper, we review the safety, efficacy, and effectiveness of maternal vaccines against LRTIs, such as pertussis, influenza, coronavirus disease 2019, and respiratory syncytial virus.
Collapse
Affiliation(s)
- Nisha Makan-Murphy
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ziyaad Dangor
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
4
|
Rolfo A, Cosma S, Nuzzo AM, Moretti L, Tancredi A, Canosa S, Revelli A, Benedetto C. Assessment of Placental Antioxidant Defense Markers in Vaccinated and Unvaccinated COVID-19 Third-Trimester Pregnancies. Life (Basel) 2024; 14:1571. [PMID: 39768279 PMCID: PMC11677986 DOI: 10.3390/life14121571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Pregnancy has been identified as a risk factor for severe COVID-19, leading to maternal and neonatal complications. The safety and effects of the SARS-CoV-2 vaccination during pregnancy, particularly on placental function and oxidative stress (OxS), remain underexplored. We investigated the impact of vaccination on third-trimester placental antioxidant defense markers. METHODS Ninety full-term pregnant women were divided into the following groups: vaccinated (n = 27) and unvaccinated (n = 25) COVID-19-positive pregnant women; control subgroups were composed of vaccinated (n = 19) or unvaccinated (n = 19) COVID-19-negative women with a healthy term singleton pregnancy with no signs of COVID-19. Placental samples were collected after delivery. Lipid peroxidation (TBARS), gene expression of HIF-1α, and catalase (CAT), superoxide dismutase-1 (SOD1) and CAT-SOD1 enzymatic activity were measured. RESULTS COVID-19-positive placentae exhibited significantly higher TBARS and HIF-1α levels compared to controls, regardless of vaccination status. Vaccination significantly increased placental CAT and SOD1 expression and activity in COVID-19-positive women, suggesting enhanced antioxidant defense. Unvaccinated women showed a higher incidence of COVID-19 symptoms and lower antioxidant enzyme activity. CONCLUSIONS SARS-CoV-2 infection induced placental OxS, which is countered by a placental adaptive antioxidant response. Vaccination during pregnancy enhances placental defense, further supporting the safety and benefits of COVID-19 vaccination in preventing complications and protecting fetal development.
Collapse
Affiliation(s)
- Alessandro Rolfo
- Department of Surgical Sciences, Gynecology and Obstetrics 2, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (A.M.N.); (L.M.); (A.R.)
| | - Stefano Cosma
- Department of Surgical Sciences, Gynecology and Obstetrics 1, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (S.C.); (A.T.); (S.C.); (C.B.)
| | - Anna Maria Nuzzo
- Department of Surgical Sciences, Gynecology and Obstetrics 2, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (A.M.N.); (L.M.); (A.R.)
| | - Laura Moretti
- Department of Surgical Sciences, Gynecology and Obstetrics 2, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (A.M.N.); (L.M.); (A.R.)
| | - Annalisa Tancredi
- Department of Surgical Sciences, Gynecology and Obstetrics 1, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (S.C.); (A.T.); (S.C.); (C.B.)
| | - Stefano Canosa
- Department of Surgical Sciences, Gynecology and Obstetrics 1, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (S.C.); (A.T.); (S.C.); (C.B.)
| | - Alberto Revelli
- Department of Surgical Sciences, Gynecology and Obstetrics 2, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (A.M.N.); (L.M.); (A.R.)
| | - Chiara Benedetto
- Department of Surgical Sciences, Gynecology and Obstetrics 1, City of Health and Science—S. Anna University Hospital, University of Turin, 10126 Turin, Italy; (S.C.); (A.T.); (S.C.); (C.B.)
| |
Collapse
|
5
|
Jasset OJ, Lopez Zapana PA, Bahadir Z, Shook L, Dennis M, Gilbert E, Liu ZA, Yinger RV, Bald C, Bradford CG, Silfen AH, Klein SL, Pekosz A, Permar S, Konnikova L, Yonker LM, Lauffenburger D, Nelson A, Elovitz MA, Edlow AG. Enhanced placental antibody transfer efficiency with longer interval between maternal respiratory syncytial virus vaccination and birth. Am J Obstet Gynecol 2024:S0002-9378(24)01125-6. [PMID: 39515450 PMCID: PMC12056162 DOI: 10.1016/j.ajog.2024.10.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Respiratory syncytial virus is associated with significant neonatal and infant morbidity and mortality. Maternal bivalent respiratory syncytial virus prefusion F respiratory syncytial virus vaccination to protect neonates and infants was approved in September 2023 for administration between 32+0 and 36+6 weeks to protect neonates and infants. This approved timeframe is narrower than the 24 to 36 week window evaluated in the clinical trial, due to the possible association between preterm birth and vaccine administration. Currently, data are lacking on how maternal vaccine timing within the approved window affects the transfer of antibodies from mother to fetus, critical information that could influence clinical practice. OBJECTIVE We sought to examine how gestational age at vaccination and time elapsed from maternal respiratory syncytial virus vaccination to delivery impacted transfer of maternal antibodies measured in the umbilical cord at delivery and in peripheral blood of 2-month infants. We also examined differences in maternal and cord respiratory syncytial virus antibody levels achieved by vaccination vs natural RSV infection. STUDY DESIGN A prospective cohort study was conducted at 2 academic medical centers between September 20, 2023 and March 21, 2024, enrolling 124 individuals who received the respiratory syncytial virus vaccine during pregnancy. Infant capillary blood was collected at 2 months of age from 29 of the infants. Maternal and cord immunoglobulin G levels achieved by respiratory syncytial virus vaccination were compared to those associated with maternal natural respiratory syncytial virus infection, using banked blood from 20 maternal:cord dyads collected prior to the availability of the maternal respiratory syncytial virus vaccine. Levels of immunoglobulin G against respiratory syncytial virus strain A2 and B fusion (F) and attachment (G) proteins and against pertussis toxin (as a comparator antigen from a vaccine routinely administered earlier in pregnancy) were measured using a Binding Antibody Multiplex Assay. Differences in titers between vaccination and natural infection were examined using Wilcoxon rank-sum test. Differences in cord:maternal transfer ratios and 2-month infant antibody levels by timing of maternal vaccination were evaluated by Kruskal-Wallis testing. RESULTS Maternal respiratory syncytial virus vaccination resulted in significantly higher maternal and cord antirespiratory syncytial virus F antibody levels than natural infection (5.72 vs 4.82 log10 mean fluorescence intensity, P<.0001 maternal; 5.81 vs 5.03 log10 mean fluorescence intensity, P<.0001 cord). Maternal vaccination 2 to 3 weeks and 3 to 4 weeks prior to delivery was associated with significantly lower cord:maternal transfer ratios than were observed when vaccination occurred >5 weeks prior to delivery (P=.03 for 2-3 weeks, P=.007 for 3-4 weeks), and significantly lower transfer ratios than observed for pertussis vaccination administered prior to 30 weeks of gestation (P=.008 for 2-3 weeks, P=.03 for 3-4 weeks, similar at >4 weeks). CONCLUSION Vaccine administration earlier in the approved 32 to 36 week window (at least 5 weeks prior to delivery) results in the highest transplacental transfer of maternal antibodies to the neonate. These results should inform the counseling of pregnant individuals on optimal vaccination timing.
Collapse
Affiliation(s)
- Olyvia J Jasset
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Paola Andrea Lopez Zapana
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Zeynep Bahadir
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Lydia Shook
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA; Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Maria Dennis
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Emily Gilbert
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Zhaojing Ariel Liu
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Rachel V Yinger
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Caroline Bald
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Caroline G Bradford
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Alexa H Silfen
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Liza Konnikova
- Departments of Pediatrics, Immunobiology, and Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT
| | - Lael M Yonker
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Ashley Nelson
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Michal A Elovitz
- Women's Biomedical Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn school of Medicine at Mount Sinai, New York, NY
| | - Andrea G Edlow
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA; Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| |
Collapse
|
6
|
Sundell GN, Tao SC. Phage Immunoprecipitation and Sequencing-a Versatile Technique for Mapping the Antibody Reactome. Mol Cell Proteomics 2024; 23:100831. [PMID: 39168282 PMCID: PMC11417174 DOI: 10.1016/j.mcpro.2024.100831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
Characterizing the antibody reactome for circulating antibodies provide insight into pathogen exposure, allergies, and autoimmune diseases. This is important for biomarker discovery, clinical diagnosis, and prognosis of disease progression, as well as population-level insights into the immune system. The emerging technology phage display immunoprecipitation and sequencing (PhIP-seq) is a high-throughput method for identifying antigens/epitopes of the antibody reactome. In PhIP-seq, libraries with sequences of defined lengths and overlapping segments are bioinformatically designed using naturally occurring proteins and cloned into phage genomes to be displayed on the surface. These libraries are used in immunoprecipitation experiments of circulating antibodies. This can be done with parallel samples from multiple sources, and the DNA inserts from the bound phages are barcoded and subjected to next-generation sequencing for hit determination. PhIP-seq is a powerful technique for characterizing the antibody reactome that has undergone rapid advances in recent years. In this review, we comprehensively describe the history of PhIP-seq and discuss recent advances in library design and applications.
Collapse
Affiliation(s)
- Gustav N Sundell
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Ce Tao
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
7
|
Li L, Matsui Y, Prahl MK, Cassidy AG, Golan Y, Jigmeddagva U, Ozarslan N, Lin CY, Buarpung S, Gonzalez VJ, Chidboy MA, Basilio E, Lynch KL, Song D, Jegatheesan P, Rai DS, Govindaswami B, Needens J, Rincon M, Myatt L, Taha TY, Montano M, Ott M, Greene WC, Gaw SL. Neutralizing and binding antibody responses to SARS-CoV-2 with hybrid immunity in pregnancy. NPJ Vaccines 2024; 9:156. [PMID: 39191763 PMCID: PMC11349990 DOI: 10.1038/s41541-024-00948-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Hybrid immunity against SARS-CoV-2 has not been well studied in pregnancy. We conducted a comprehensive analysis of neutralizing antibodies (nAb) and binding antibodies in pregnant individuals who received mRNA vaccination, natural infection, or both. A third vaccine dose augmented nAb levels compared to the two-dose regimen or natural infection alone; this effect was more pronounced in hybrid immunity. There was reduced anti-Omicron nAb, but the maternal-fetal transfer efficiency remained comparable to that of other variants. Vaccine-induced nAbs were transferred more efficiently than infection-induced nAbs. Anti-spike receptor binding domain (RBD) IgG was associated with nAb against wild-type (Wuhan-Hu-1) following breakthrough infection. Both vaccination and infection-induced anti-RBD IgA, which was more durable than anti-nucleocapsid IgA. IgA response was attenuated in pregnancy compared to non-pregnant controls. These data provide additional evidence of augmentation of humoral immune responses in hybrid immunity in pregnancy.
Collapse
Affiliation(s)
- Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yusuke Matsui
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Mary K Prahl
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Division of Pediatric Infectious Diseases and Global Health, University of California San Francisco, San Francisco, CA, USA
| | - Arianna G Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Christine Y Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Veronica J Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Megan A Chidboy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Emilia Basilio
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Dongli Song
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Priya Jegatheesan
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Daljeet S Rai
- Stanford-O'Connor Family Medicine Residency Program, Division of Family Medicine, Stanford University, Palo Alto, CA, USA
| | - Balaji Govindaswami
- Division of Neonatology, Department of Pediatrics, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Jordan Needens
- Department of Obstetrics and Gynecology, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Monica Rincon
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA.
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA.
| | - Stephanie L Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Martínez-Quezada R, Valencia-Ledezma OE, Ramírez-Lozada T, Miguel-Rodríguez CE, Fernández-Hernández JC, Acosta-Altamirano G. Influence of Maternal and Neonatal Factors on Transplacental Passive Immunity after Vaccination against COVID-19. Vaccines (Basel) 2024; 12:860. [PMID: 39203986 PMCID: PMC11360686 DOI: 10.3390/vaccines12080860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
In the present study, we evaluated the influence of maternal and neonatal factors on the efficiency of the placental transfer of neutralizing antibodies against SARS-CoV-2. Vaccination during pregnancy provides fetal and neonatal protection through the passive transplacental transfer of maternal neutralizing antibodies. To date, little information is available regarding the factors that affect the transfer of antibodies against SARS-CoV-2. A retrospective, cross-sectional, observational, and analytical study was carried out. It was found that several biological factors could be altering transplacental passive immunity after vaccination against COVID-19. In our study population, type 2 diabetes mellitus and chronic hypertension tended to decrease efficiency, while data from women with pre-eclampsia showed better indices compared to those from women with healthy pregnancies. Neonates born prematurely showed lower transfer rates when compared to healthy neonates. The premature rupture of membranes significantly decreased antibody transfer. Taken together, the data suggest that vaccination against COVID-19 during pregnancy is effective even under certain unfavorable clinical conditions for the mother, fetus, and neonate. It is important to create and disseminate immunization strategies in vulnerable populations to reduce maternal and perinatal morbidity and mortality associated with infections preventable by vaccination.
Collapse
Affiliation(s)
- Rebeca Martínez-Quezada
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico; (R.M.-Q.); (O.E.V.-L.); (C.E.M.-R.)
- Consejo Mexiquense de Ciencia y Tecnología (COMECYT), Toluca 50120, Mexico
| | - Omar Esteban Valencia-Ledezma
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico; (R.M.-Q.); (O.E.V.-L.); (C.E.M.-R.)
| | - Tito Ramírez-Lozada
- Servicio de Ginecología y Obstetricia, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico;
| | - Carlos Emilio Miguel-Rodríguez
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico; (R.M.-Q.); (O.E.V.-L.); (C.E.M.-R.)
| | | | - Gustavo Acosta-Altamirano
- Dirección de Investigación, Hospital General de México “Eduardo Liceaga”, Ciudad de México 06726, Mexico;
| |
Collapse
|
9
|
Jarlhelt I, Hansen CB, Pérez-Alós L, Weihe P, Petersen MS, Garred P. SARS-CoV-2 anti-RBD and anti-N protein responses are differentially regulated between mother-child pairs: insight from a national study cohort at the Faroe Islands. Front Immunol 2024; 15:1418678. [PMID: 39021574 PMCID: PMC11251900 DOI: 10.3389/fimmu.2024.1418678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Background Knowledge about SARS-CoV-2 antibody dynamics in neonates and direct comparisons with maternal antibody responses are not well established. This study aimed to characterize and directly compare the maternal and infant antibody response in a national birth cohort from the Faroe Islands. Methods The levels of immunoglobulins (Ig) targeting the receptor binding domain (RBD) of the spike protein and the nucleocapsid protein (N protein) of SARS-CoV-2 were investigated in maternal blood and umbilical cord blood from neonates. The study included 537 neonates and 565 mothers from the Faroe Islands, and follow-up samples were collected 12 months after birth. Multiple linear regression models were used to assess associations of maternal parameters with maternal and neonatal Ig levels and pregnancy outcomes. Results The finding showed that neonates acquired varying levels of SARS-CoV-2 antibodies through transplacental transfer, and the levels were significantly influenced by the mother's vaccination and infection status. The study also found that maternal vaccination and the presence of SARS-CoV-2 antibodies targeting spike RBD were associated with gestational age and APGAR scores. Furthermore, the anti-RBD and -N protein-specific antibody response dynamics during 12 months after birth exhibited differences between mothers and children. RBD and N protein responses were maintained at follow-up in the mother's cohort, while only the N protein response was maintained at follow-up in the children's cohort. Conclusion In conclusion, SARS-CoV-2-specific immune responses in newborns rely on maternal immunity, while the persistence of SARS-CoV-2-specific Igs appears to be differently regulated between mothers and children. The study provides new insights into the dynamics of SARS-CoV-2-specific immune responses in newborns and underscores the nuanced relationship between maternal factors and neonatal humoral responses.
Collapse
Affiliation(s)
- Ida Jarlhelt
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Cecilie Bo Hansen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pál Weihe
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Maria Skaalum Petersen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Lin X, Botros B, Hanna M, Gurzenda E, De Mejia CM, Chavez M, Hanna N. Transplacental transmission of the COVID-19 vaccine messenger RNA: evidence from placental, maternal, and cord blood analyses postvaccination. Am J Obstet Gynecol 2024; 230:e113-e116. [PMID: 38307473 DOI: 10.1016/j.ajog.2024.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/04/2024]
Affiliation(s)
- Xinhua Lin
- Women and Children's Research Laboratory, Department of Foundations of Medicine, New York University-Grossman Long Island School of Medicine, Mineola, NY
| | - Bishoy Botros
- Women and Children's Research Laboratory, Department of Foundations of Medicine, New York University-Grossman Long Island School of Medicine, Mineola, NY
| | - Monica Hanna
- Division of Neonatology, Department of Pediatrics, New York University Langone Hospital-Long Island, New York University-Grossman Long Island School of Medicine, Mineola, NY
| | - Ellen Gurzenda
- Women and Children's Research Laboratory, Department of Foundations of Medicine, New York University-Grossman Long Island School of Medicine, Mineola, NY
| | - Claudia Manzano De Mejia
- Women and Children's Research Laboratory, Department of Foundations of Medicine, New York University-Grossman Long Island School of Medicine, Mineola, NY
| | - Martin Chavez
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, New York University Langone Hospital-Long Island, New York University-Grossman Long Island School of Medicine, Mineola, NY
| | - Nazeeh Hanna
- Women and Children's Research Laboratory, Department of Foundations of Medicine, New York University-Grossman Long Island School of Medicine, Mineola, NY; Division of Neonatology, Department of Pediatrics, New York University Langone Hospital-Long Island, New York University-Grossman Long Island School of Medicine, 259 First St, Mineola, NY 11501.
| |
Collapse
|
11
|
Sabharwal V, Taglauer E, Demos R, Snyder-Cappione J, Shaik-Dasthagirisaheb YB, Parker-Kelleher S, Hunnewell J, Boateng J, Clarke K, Yuen R, Barnett ED, Wachman EM, Yarrington CD. Comparison of Anti-SARS-CoV-2-Specific Antibody Signatures in Maternal and Infant Blood after COVID-19 Infection versus COVID-19 Vaccination during Pregnancy. Am J Perinatol 2024; 41:e2970-e2977. [PMID: 37774748 DOI: 10.1055/a-2183-9109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
OBJECTIVE The Advisory Committee on Immunization Practices and The American College of Obstetricians and Gynecologists recommend coronavirus disease 2019 (COVID-19) vaccine for pregnant persons to prevent severe illness and death. The objective was to examine levels of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG, IgM, and IgA against spike protein receptor binding domain (RBD) and nucleocapsid protein (NCP) in maternal and infant/cord blood at delivery after COVID 19 vaccination compared with SARS-CoV-2 infection at in mother-infant dyads at specified time points. STUDY DESIGN Mothers with SARS-CoV-2 infection (n = 31) or COVID-19 vaccination (n = 25) during pregnancy were enrolled between July 2020 and November 2021. Samples were collected at delivery and IgG, IgM, and IgA to RBD of spike and NCPs compared in the infected and vaccinated groups. Timing of infection/vaccination prior to delivery and correlation with antibody levels was performed. RESULTS The majority of participants received vaccination within 90 days of delivery and over half received the Pfizer BioNTech vaccine. There were no significant correlations between antibody levels and timing of infection or vaccination. Infant IgG levels to the RBD domain of spike protein were higher in the vaccinated group (n = 25) as compared with the infants born to mothers with infection (n = 31). Vaccination against COVID-19 during pregnancy was associated with detectable maternal and infant anti-RBD IgG levels at delivery irrespective of the timing of vaccination. CONCLUSION Timing of vaccination had no correlation to the antibody levels suggesting that the timing of maternal vaccination in the cohort did not matter. There was no IgM detected in infants from vaccinated mothers. Infants from vaccinated mothers had robust IgG titers to RBD, which have a lasting protective effect in infants. KEY POINTS · COVID-19 vaccination during pregnancy had detectable antibody.. · No correlation between antibody levels and timing of vaccination.. · Infants from vaccinated mothers had robust IgG titers to RBD..
Collapse
Affiliation(s)
| | | | - Riley Demos
- Boston University School of Medicine, Boston, Massachusetts
| | | | | | | | | | - Jeffery Boateng
- Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Katherine Clarke
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | - Rachel Yuen
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | | | - Elisha M Wachman
- Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | | |
Collapse
|
12
|
Norman M, Magnus MC, Söderling J, Juliusson PB, Navér L, Örtqvist AK, Håberg S, Stephansson O. Neonatal Outcomes After COVID-19 Vaccination in Pregnancy. JAMA 2024; 331:396-407. [PMID: 38319332 PMCID: PMC10848052 DOI: 10.1001/jama.2023.26945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/10/2023] [Indexed: 02/07/2024]
Abstract
Importance Better knowledge about neonatal adverse events after COVID-19 vaccination during pregnancy could help address concerns about vaccine safety. Objective To evaluate the risks of neonatal adverse events after exposure to COVID-19 vaccination during pregnancy. Design, Setting, and Participants Population-based cohort study including all infants in Sweden and Norway born from June 2021 to January 2023. Unique personal identity numbers were used to link individual information from different national registers. Exposure Administration of any mRNA vaccine against COVID-19 during pregnancy, irrespective of previous vaccination, number of doses during pregnancy, or vaccine manufacturer. Main Outcomes and Measures Outcomes were neonatal conditions with bleeding/thrombosis or inflammation/infection; disorders of the central nervous system; circulatory, respiratory, or gastrointestinal problems; and neonatal mortality. Statistical methods included logistic regression adjusted for characteristics of the pregnant individuals, with additional restricted and stratified analyses. Results Of 196 470 newborn infants included (51.3% male, 93.8% born at term, 62.5% born in Sweden), 94 303 (48.0%) were exposed to COVID-19 vaccination during pregnancy. Exposed infants exhibited no increased odds of adverse neonatal outcomes, and they exhibited lower odds for neonatal nontraumatic intracranial hemorrhage (event rate, 1.7 vs 3.2/1000; adjusted odds ratio [aOR], 0.78 [95% CI, 0.61-0.99]), hypoxic-ischemic encephalopathy (1.8 vs 2.7/1000; aOR, 0.73 [95% CI, 0.55-0.96]), and neonatal mortality (0.9 vs 1.8/1000; aOR, 0.68 [95% CI, 0.50-0.91]). Subgroup analyses found a similar association between vaccination during pregnancy and lower neonatal mortality; subgroups were restricted to infants delivered by individuals unvaccinated before pregnancy, individuals vaccinated before pregnancy, individuals vaccinated after a general recommendation of vaccination during pregnancy was issued, and individuals without COVID-19 infection during pregnancy. Analyses restricted to term infants, singleton births, or infants without birth defects yielded similar results. Stratifying the analysis by vaccine manufacturer did not attenuate the association between vaccination and low neonatal mortality. Conclusions and Relevance In this large population-based study, vaccination of pregnant individuals with mRNA COVID-19 vaccines was not associated with increased risks of neonatal adverse events in their infants.
Collapse
Affiliation(s)
- Mikael Norman
- Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Neonatal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Maria C. Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Jonas Söderling
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Petur B. Juliusson
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Health Registry Research and Development, Norwegian Institute of Public Health, Bergen, Norway
- Department of Paediatric and Adolescent Medicine, Haukeland University Hospital, Bergen, Norway
| | - Lars Navér
- Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Neonatal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Anne K. Örtqvist
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Visby County Hospital, Visby, Sweden
| | - Siri Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Olof Stephansson
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Women’s Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
13
|
Elko EA, Mead HL, Nelson GA, Zaia JA, Ladner JT, Altin JA. Recurrent SARS-CoV-2 mutations at Spike D796 evade antibodies from pre-Omicron convalescent and vaccinated subjects. Microbiol Spectr 2024; 12:e0329123. [PMID: 38189279 PMCID: PMC10871546 DOI: 10.1128/spectrum.03291-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lineages of the Omicron variant rapidly became dominant in early 2022 and frequently cause human infections despite vaccination or prior infection with other variants. In addition to antibody-evading mutations in the receptor-binding domain, Omicron features amino acid mutations elsewhere in the Spike protein; however, their effects generally remain ill defined. The Spike D796Y substitution is present in all Omicron sub-variants and occurs at the same site as a mutation (D796H) selected during viral evolution in a chronically infected patient. Here, we map antibody reactivity to a linear epitope in the Spike protein overlapping position 796. We show that antibodies binding this region arise in pre-Omicron SARS-CoV-2 convalescent and vaccinated subjects but that both D796Y and D796H abrogate their binding. These results suggest that D796Y contributes to the fitness of Omicron in hosts with pre-existing immunity to other variants of SARS-CoV-2 by evading antibodies targeting this site.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has evolved substantially through the coronavirus disease 2019 (COVID-19) pandemic: understanding the drivers and consequences of this evolution is essential for projecting the course of the pandemic and developing new countermeasures. Here, we study the immunological effects of a particular mutation present in the Spike protein of all Omicron strains and find that it prevents the efficient binding of a class of antibodies raised by pre-Omicron vaccination and infection. These findings reveal a novel consequence of a poorly understood Omicron mutation and shed light on the drivers and effects of SARS-CoV-2 evolution.
Collapse
Affiliation(s)
- Evan A. Elko
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - Heather L. Mead
- The Translational Genomics Research Institute (TGen), Flagstaff, Arizona, USA
| | - Georgia A. Nelson
- The Translational Genomics Research Institute (TGen), Flagstaff, Arizona, USA
| | - John A. Zaia
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Jason T. Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - John A. Altin
- The Translational Genomics Research Institute (TGen), Flagstaff, Arizona, USA
| |
Collapse
|
14
|
Martínez-Quezada R, Miguel-Rodríguez CE, Ramírez-Lozada T, Valencia-Ledezma OE, Acosta-Altamirano G. Placental Transfer Efficiency of Neutralizing Antibodies on SARS-CoV-2 Vaccination before and after Pregnancy in Mexican Women. Int J Mol Sci 2024; 25:1516. [PMID: 38338795 PMCID: PMC10855582 DOI: 10.3390/ijms25031516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/12/2024] Open
Abstract
The protection of the neonate against pathogens depends largely on the antibodies transferred placentally from the mother; for this reason, maternal vaccination against emerging viruses, such as SARS-CoV-2, is of vital importance. Knowing some of the immunogenic factors that could alter the placental transfer of antibodies could aid in understanding the immune response and neonatal protection after maternal vaccination. In this study, we analyzed the efficiency of the placental transfer of binding and neutralizing antibodies, as well as some factors that could alter the passive immune response, such as the trimester of gestation at the time of immunization, the number of doses received by the mother and the type of vaccine. Binding IgG antibodies were detected by ELISA, and the detection of neutralizing antibodies was carried out using flow cytometry. Our results show efficient transfer rates (>1), which are higher when maternal vaccination occurs during the third trimester of gestation. Antibodies are detectable in mothers and their neonates after 12 months of maternal immunization, suggesting than the vaccination against COVID-19 before and during pregnancy in the Mexican population induces a lasting neutralizing response in mothers and their newborns.
Collapse
Affiliation(s)
- Rebeca Martínez-Quezada
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
- Consejo Mexiquense de Ciencia y Tecnología (COMECYT), Paseo Colón N° 112-A, Ciprés, Toluca 50120, Mexico
| | - Carlos Emilio Miguel-Rodríguez
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
| | - Tito Ramírez-Lozada
- Unidad de Ginecología y Obstetricia, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico;
| | - Omar Esteban Valencia-Ledezma
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
| | - Gustavo Acosta-Altamirano
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
| |
Collapse
|
15
|
Kachikis A, Pike M, Eckert LO, Roberts E, Frank Y, Young AL, Goecker E, Gravett MG, Greninger AL, Englund JA. Timing of Maternal COVID-19 Vaccine and Antibody Concentrations in Infants Born Preterm. JAMA Netw Open 2024; 7:e2352387. [PMID: 38241046 PMCID: PMC10799259 DOI: 10.1001/jamanetworkopen.2023.52387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/30/2023] [Indexed: 01/22/2024] Open
Abstract
Importance COVID-19 vaccine-derived antibodies in pregnant people may protect infants from severe infection in the first 6 months of life via transplacental antibody transfer. Few data exist on maternally derived SARS-CoV-2 antibodies in preterm compared with full-term infants in association with vaccination timing. Objective To compare SARS-CoV-2 anti-Spike (anti-S) antibody levels in preterm and full-term infants in the context of vaccine dose timing before delivery. Design, Setting, and Participants This prospective cohort study enrolled pregnant individuals and collected paired maternal and cord blood samples at delivery at the University of Washington between February 1, 2021, and January 31, 2023. Participants who had received at least 2 doses of a messenger RNA COVID-19 vaccine before delivery and did not have a history of prior COVID-19 infection or detectable anti-SARS-CoV-2 nucleocapsid antibodies were included. Exposures Timing of the last vaccine dose and preterm or full-term gestational age at delivery. Main Outcomes and Measures Paired maternal and cord samples were tested for anti-S antibody, and linear regression was used to evaluate associations between preterm delivery and anti-S antibody levels. Covariates included timing of last dose, number of doses, insurance status, and immunosuppressing medications. Results A total of 220 participants (median [IQR] age, 34 [32-37] years; 212 [96.4%] female) with 36 preterm and 184 full-term deliveries were studied. Before delivery, 121 persons received 2 vaccine doses and 99 persons received 3 or more vaccine doses. The geometric mean concentration of maternal anti-S antibodies was 674 (95% CI, 577-787) after 2 doses and 8159 (95% CI, 6636-10 032) after 3 or more doses (P < .001). The cord anti-S antibody geometric mean concentration was 1000 (95% CI, 874-1144) after 2 doses and 9992 (95% CI, 8381-11 914) after 3 or more doses (P < .001). After adjustment for vaccine timing and number of doses before delivery, no association was found between preterm delivery and cord anti-S antibody levels (β = 0.44; 95% CI, -0.06 to 0.94). Conclusions and Relevance In this prospective cohort study of pregnant individuals with preterm and full-term deliveries, receipt of 3 or more compared with 2 doses of COVID-19 vaccine before delivery resulted in 10-fold higher cord anti-S antibody levels. Maternal antibody concentration appeared more important than delivery gestational age in determining cord anti-S antibody levels. The number of doses and timing considerations for COVID-19 vaccine in pregnancy should include individuals at risk for preterm delivery.
Collapse
Affiliation(s)
- Alisa Kachikis
- Department of Obstetrics and Gynecology, University of Washington, Seattle
| | - Mindy Pike
- Department of Obstetrics and Gynecology, University of Washington, Seattle
| | - Linda O. Eckert
- Department of Obstetrics and Gynecology, University of Washington, Seattle
- Department of Global Health, University of Washington, Seattle
| | - Emma Roberts
- Department of Obstetrics and Gynecology, University of California, San Diego
| | - Yael Frank
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amber L. Young
- Department of Obstetrics and Gynecology, University of Washington, Seattle
| | - Erin Goecker
- Department of Laboratory Medicine, University of Washington, Seattle
| | - Michael G. Gravett
- Department of Obstetrics and Gynecology, University of Washington, Seattle
- Department of Global Health, University of Washington, Seattle
| | | | - Janet A. Englund
- Seattle Children’s Hospital Research Institute, Department of Pediatrics, University of Washington, Seattle
| |
Collapse
|
16
|
Tudisco R, Garufi C, Rizzo F, Polimeno T, Lanzone A, De Carolis S. Impact of mRNA-based vaccines in the prevention of adverse outcomes of COVID-19 infection in pregnancy: a single-center cohort study. Front Pediatr 2023; 11:1214768. [PMID: 37941978 PMCID: PMC10628479 DOI: 10.3389/fped.2023.1214768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
Several data have suggested that pregnant women have an increased risk of severe COVID-19 compared to those who are not pregnant. Moreover, different studies have showed that severe COVID-19 is limited mostly to unvaccinated women. The aim of the present study was to ascertain the different maternal and fetal outcomes in pregnant women with COVID-19 according to their vaccination status. A retrospective cohort study was carried out including all women admitted to the high-risk pregnancy unit of our center with COVID-19 between December 2021 and February 2022. Among the 163 women included in the study, 60 were vaccinated with an mRNA vaccine and 103 were unvaccinated. Pregnancy outcome and obstetrical and neonatal complications were encountered. Vaccinated women showed higher educational levels and lower prevalence of cases, with BMI >25 compared to unvaccinated women. Moreover, vaccinated women were admitted mostly for obstetrical indications rather than for COVID-related symptoms. In addition, the risk of developing COVID-19 pneumonia was significantly higher in unvaccinated women (p = 0.01) compared with vaccinated ones. Furthermore, pregnancy and neonatal outcomes showed some differences in the two cohorts. In unvaccinated women, the rate of C-section was higher (p = 0.03), and the mean birthweight percentile in their infants was impaired by COVID-19 infection (p = 0.01) when compared to those born to vaccinated women. Based on these results, we suggest that women who received a full course of vaccination were protected from the severity of the disease, having milder symptoms of SARS-Cov2 infection, while also presenting a more favorable pregnancy outcome.
Collapse
Affiliation(s)
- Riccardo Tudisco
- Dipartimento Scienze Della Salute Della Donna, Del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristina Garufi
- Arthritis Center, Reumatologia, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Rome, Italy
| | - Francesca Rizzo
- Dipartimento Scienze Della Salute Della Donna, Del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Teresa Polimeno
- Dipartimento Scienze Della Salute Della Donna, Del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Lanzone
- Dipartimento Scienze Della Salute Della Donna, Del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sara De Carolis
- Dipartimento Scienze Della Salute Della Donna, Del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
17
|
Golan Y, Ilala M, Li L, Gay C, Hunagund S, Lin CY, Cassidy AG, Jigmeddagva U, Matsui Y, Ozarslan N, Asiodu IV, Ahituv N, Flaherman VJ, Gaw SL, Prahl M. Milk antibody response after 3 rd COVID-19 vaccine and SARS-CoV-2 infection and implications for infant protection. iScience 2023; 26:107767. [PMID: 37731614 PMCID: PMC10507209 DOI: 10.1016/j.isci.2023.107767] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/28/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
Little is known about the persistence of human milk anti-SARS-CoV-2 antibodies after 2nd and 3rd vaccine doses and infection following 3rd dose. In this study, human milk, saliva, and blood samples were collected from 33 lactating individuals before and after vaccination and infection. Antibody levels were measured using ELISA and symptoms were assessed using questionnaires. We found that after vaccination, milk anti-SARS-CoV-2 antibodies persisted for up to 8 months. In addition, distinct patterns of human milk IgA and IgG production and higher milk RBD-blocking activity was observed after infection compared to 3-dose vaccination. Infected mothers reported more symptoms than vaccinated mothers. We examined the persistence of milk antibodies in infant saliva after breastfeeding and found that IgA was more abundant compared to IgG. Our results emphasize the importance of improving the secretion of IgA antibodies to human milk after vaccination to improve the protection of breastfeeding infants.
Collapse
Affiliation(s)
- Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Mikias Ilala
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Caryl Gay
- Department of Family Health Care Nursing, University of California, San Francisco, San Francisco, CA, USA
| | - Soumya Hunagund
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Christine Y. Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Arianna G. Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yusuke Matsui
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Ifeyinwa V. Asiodu
- Department of Family Health Care Nursing, University of California, San Francisco, San Francisco, CA, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Valerie J. Flaherman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Mary Prahl
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Division of Pediatric Infectious Diseases and Global Health, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
18
|
Gonzalez VJ, Li L, Buarpung S, Prahl M, Robinson JF, Gaw SL. Minimal mRNA uptake and inflammatory response to COVID-19 mRNA vaccine exposure in human placental explants. iScience 2023; 26:107549. [PMID: 37664582 PMCID: PMC10470080 DOI: 10.1016/j.isci.2023.107549] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Despite universal recommendations for COVID-19 mRNA vaccination in pregnancy, uptake has been lower than desired. There have been limited studies of the direct impact of COVID-19 mRNA vaccine exposure in human placental tissue. Using a primary human placental explants model, we investigated the uptake of two common mRNA vaccines (BNT162b2 Pfizer-BioNTech or mRNA-1273 Moderna), and whether exposure altered villous cytokine responses. Explants derived from second or third trimester chorionic villi were incubated with vaccines at supraphysiologic concentrations and analyzed at two time points. We observed minimal uptake of mRNA vaccines in placental explants by in situ hybridization and quantitative RT-PCR. No specific or global cytokine response was elicited by either of the mRNA vaccines in multiplexed immunoassays. Our results suggest that the human placenta does not readily absorb the COVID-19 mRNA vaccines nor generate a significant inflammatory response after exposure.
Collapse
Affiliation(s)
- Veronica J. Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mary Prahl
- Division of Pediatric Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Joshua F. Robinson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
19
|
Marchand G, Masoud AT, Grover S, King A, Brazil G, Ulibarri H, Parise J, Arroyo A, Coriell C, Goetz S, Moir C, Govindan M, Moberly A, Proctor A, Sainz K, Blumrick R. Maternal and neonatal outcomes of COVID-19 vaccination during pregnancy, a systematic review and meta-analysis. NPJ Vaccines 2023; 8:103. [PMID: 37454153 PMCID: PMC10349851 DOI: 10.1038/s41541-023-00698-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 06/27/2023] [Indexed: 07/18/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is associated with increased pregnancy complications. Despite effective vaccination strategies for the general population, the evidence on the safety and efficacy of Coronavirus disease 2019 (COVID-19) vaccinations in pregnancy is limited due to a lack of well-powered studies. The present study compares the maternal, neonatal, and immunological outcomes between vaccinated pregnant and unvaccinated pregnant women using a systematic review and meta-analysis approach. We included 37 studies with a total of 141,107 pregnant women (36.8% vaccinated) spread across all outcomes. Our evidence indicates a higher rate of cesarean section in the 1898 vaccinated pregnant women compared to the 6180 women who did not receive vaccination (OR = 1.20, CI = (1.05, 1.38), P = 0.007, I2 = 45%). Regarding immunological outcomes, the risk of SARS-CoV-2 infection during pregnancy or postpartum was significantly reduced in 6820 vaccinated pregnant women compared to 17,010 unvaccinated pregnant women (OR = 0.25, CI = 0.13-0.48, P < 0.0001, I2 = 61%), as evident from qualitative assessment indicating significantly higher postpartum antibody titers compared to that observed in both unvaccinated mothers and mothers who have recently recovered from a SARS-CoV-2 infection. Our analysis represents high quality evidence showing that COVID-19 vaccination effectively raises antibody titers against SARS-CoV-2. This may confer protection against infection during pregnancy and the postpartum period. In addition to being protective against SARS-CoV-2, the vaccine was associated with decreased odds of preterm delivery. Furthermore, COVID-19 vaccination may also be associated with higher odds of cesarean section.
Collapse
Affiliation(s)
- Greg Marchand
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA.
| | - Ahmed Taher Masoud
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
- Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Sandeep Grover
- Center for Human Genetics, Universitatsklinikum Giessen und Marburg, Marburg, Germany
| | - Alexa King
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Giovanna Brazil
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Hollie Ulibarri
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Julia Parise
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Amanda Arroyo
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | | | - Sydnee Goetz
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Carmen Moir
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Malini Govindan
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Atley Moberly
- Marchand Institute for Minimally Invasive Surgery, Mesa, AZ, USA
| | - Anna Proctor
- University of Iowa, College of Public Health, Iowa City, IA, USA
| | - Katelyn Sainz
- Tucson Medical Center, Department of Pediatrics, Tucson, AZ, USA
| | | |
Collapse
|
20
|
Macáková K, Pšenková P, Šupčíková N, Vlková B, Celec P, Záhumenský J. Effect of SARS-CoV-2 Infection and COVID-19 Vaccination on Oxidative Status of Human Placenta: A Preliminary Study. Antioxidants (Basel) 2023; 12:1403. [PMID: 37507942 PMCID: PMC10376152 DOI: 10.3390/antiox12071403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Infection with SARS-CoV-2 during pregnancy increases the risk of pregnancy complications associated with inflammation, which could lead to oxidative stress in the placenta. Whether vaccination against COVID-19 has any effect is unclear. This study aimed to analyze the effects of SARS-CoV-2 infection and vaccination against COVID-19 during pregnancy on oxidative stress in the placenta and on extracellular DNA (ecDNA) in umbilical cord plasma. Placenta samples from healthy uninfected and unvaccinated control patients who recovered from COVID-19 and women vaccinated against COVID-19 during pregnancy were collected. Biomarkers of oxidative damage and antioxidant capacity were assessed in the placenta homogenates. EcDNA and deoxyribonuclease activity were quantified in umbilical cord plasma using real-time PCR and the single radial enzyme diffusion method, respectively. Markers of oxidative damage to lipids and proteins as well as antioxidant capacity in the placenta did not differ between the study groups. No differences were observed in total, nuclear or mitochondrial ecDNA, or deoxyribonuclease activity in the umbilical cord plasma. Taking into account the limits of a small observational study, our results suggest that the infection with SARS-CoV-2 and vaccination against COVID-19 do not induce any major disturbances in the balance between the production of free radicals and antioxidant activity in the placenta. This is in line with the minor effects on fetal outcomes and ecDNA as a suggested marker of fetal well-being.
Collapse
Affiliation(s)
- Kristína Macáková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia
| | - Petra Pšenková
- 2nd Department of Gynaecology and Obstetrics, University Hospital Bratislava and Comenius University, 82606 Bratislava, Slovakia
| | - Nadja Šupčíková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia
| | - Barbora Vlková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia
| | - Jozef Záhumenský
- 2nd Department of Gynaecology and Obstetrics, University Hospital Bratislava and Comenius University, 82606 Bratislava, Slovakia
| |
Collapse
|
21
|
Cassidy AG, Li L, Golan Y, Gay C, Lin CY, Jigmeddagva U, Chidboy MA, Ilala M, Buarpung S, Gonzalez VJ, Basilio E, Duck M, Murtha AP, Wu AHB, Lynch KL, Asiodu IV, Prahl MK, Gaw SL. Assessment of Adverse Reactions, Antibody Patterns, and 12-month Outcomes in the Mother-Infant Dyad After COVID-19 mRNA Vaccination in Pregnancy. JAMA Netw Open 2023; 6:e2323405. [PMID: 37450302 PMCID: PMC10349345 DOI: 10.1001/jamanetworkopen.2023.23405] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/28/2023] [Indexed: 07/18/2023] Open
Abstract
Importance Longitudinal data on COVID-19 messenger RNA (mRNA) vaccine reactogenicity and immunogenicity in pregnancy and for the mother-infant dyad are needed. Objective To examine COVID-19 mRNA vaccine reactogenicity and immunogenicity in pregnancy and observe longitudinal maternal and infant outcomes. Design, Setting, and Participants This prospective cohort study of pregnant individuals enrolled in the COVID-19 Vaccination in Pregnancy and Lactation study from December 1, 2020, through December 31, 2021, with follow-up through March 31, 2022, was conducted at a large academic medical center in an urban metropolitan area in California. Pregnant individuals receiving COVID-19 mRNA vaccines (mRNA-1273 [Moderna] and BNT162b2 [Pfizer-BioNTech]) were eligible. Of 81 participants enrolled, 5 were excluded after enrollment: 1 terminated pregnancy, 1 received the third vaccine dose prior to delivery, and 3 delivered prior to completing the initial vaccine series. Exposure COVID-19 mRNA vaccination at any time during pregnancy. Main Outcomes and Measures The primary outcomes were vaccine response as measured by blood Immunoglobulin G (IgG) titers after each vaccine dose and self-reported postvaccination symptoms. Patients' IgG titers were measured in cord blood and in infant blood at intervals up to 1 year of life; IgG and IgA titers were measured in maternal milk. Clinical outcomes were collected from medical records. Results Of 76 pregnant individuals included in final analyses (median [IQR] maternal age, 35 [29-41] years; 51 [67.1%] White; 28 [36.8%] primigravid; 37 [48.7%] nulliparous), 42 (55.3%) received BNT162b2 and 34 (44.7%) received mRNA-1237. There were no significant differences in maternal characteristics between the 2 vaccine groups. Systemic symptoms were more common after receipt of the second vaccine dose than after the first dose (42 of 59 [71.2%] vs 26 of 59 [44.1%]; P = .007) and after mRNA-1237 than after BNT162b2 (25 of 27 [92.6%] vs 17 of 32 53.1%; P = .001). Systemic symptoms were associated with 65.6% higher median IgG titers than no symptoms after the second vaccine dose (median [IQR], 2596 [1840-4455] vs 1568 [1114-4518] RFU; P = .007); mean cord titers in individuals with local or systemic symptoms were 6.3-fold higher than in individuals without symptoms. Vaccination in all trimesters elicited a robust maternal IgG response. The IgG transfer ratio was highest among individuals vaccinated in the second trimester. Anti-SARS-CoV-2 IgG was detectable in cord blood regardless of vaccination trimester. In milk, IgG and IgA titers remained above the positive cutoff for at least 5-6 months after birth, and infants of mothers vaccinated in the second and third trimesters had positive IgG titers for at least 5 to 6 months of life. There were no vaccine-attributable adverse perinatal outcomes. Conclusions and Relevance The findings of this cohort study suggest that mRNA COVID-19 vaccination in pregnancy provokes a robust IgG response for the mother-infant dyad for approximately 6 months after birth. Postvaccination symptoms may indicate a more robust immune response, without adverse maternal, fetal, or neonatal outcomes.
Collapse
Affiliation(s)
- Arianna G. Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco
- Institute for Human Genetics, University of California, San Francisco
| | - Caryl Gay
- Department of Family Health Care Nursing, University of California, San Francisco
| | - Christine Y. Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Unurzul Jigmeddagva
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Megan A. Chidboy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Mikias Ilala
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
- Division of Pediatric Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Veronica J. Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Emilia Basilio
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Meghan Duck
- UCSF Benioff Children’s Hospital, University of California, San Francisco
| | - Amy P. Murtha
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| | - Alan H. B. Wu
- Department of Laboratory Medicine, University of California, San Francisco
| | - Kara L. Lynch
- Department of Laboratory Medicine, University of California, San Francisco
| | - Ifeyinwa V. Asiodu
- Department of Family Health Care Nursing, University of California, San Francisco
| | - Mary K. Prahl
- Division of Pediatric Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco
| |
Collapse
|
22
|
Juliá-Burchés C, Martínez-Varea A. An Update on COVID-19 Vaccination and Pregnancy. J Pers Med 2023; 13:jpm13050797. [PMID: 37240967 DOI: 10.3390/jpm13050797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Pregnant women are more prone to experience severe COVID-19 disease, including intensive care unit (ICU) admission, use of invasive ventilation, extracorporeal membrane oxygenation (ECMO), and mortality compared to non-pregnant individuals. Additionally, research suggests that SARS-CoV-2 infection during pregnancy is linked to adverse pregnancy outcomes, such as preterm birth, preeclampsia, and stillbirth, as well as adverse neonatal outcomes, including hospitalization and admission to the neonatal intensive care unit. This review assessed the available literature from November 2021 to 19 March 2023, concerning the safety and effectiveness of COVID-19 vaccination during pregnancy. COVID-19 vaccination administered during pregnancy is not linked to significant adverse events related to the vaccine or negative obstetric, fetal, or neonatal outcomes. Moreover, the vaccine has the same effectiveness in preventing severe COVID-19 disease in pregnant individuals as in the general population. Additionally, COVID-19 vaccination is the safest and most effective method for pregnant women to protect themselves and their newborns from severe COVID-19 disease, hospitalization, and ICU admission. Thus, vaccination should be recommended for pregnant patients. While the immunogenicity of vaccination in pregnancy appears to be similar to that in the general population, more research is needed to determine the optimal timing of vaccination during pregnancy for the benefit of the neonate.
Collapse
Affiliation(s)
- Cristina Juliá-Burchés
- Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Alicia Martínez-Varea
- Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain
| |
Collapse
|
23
|
Dawood FS, Tita A, Stockwell MS, Newes-Adeyi G, Wielgosz K, Gyamfi-Bannerman C, Battarbee A, Reichle L, Thornburg N, Ellington S, Galang RR, Vorwaller K, Vargas CY, Morrill T, Parks M, Powers E, Gibson M, Varner M. Neutralizing Antibody Responses to Messenger RNA Coronavirus Disease 2019 Vaccines Versus Severe Acute Respiratory Syndrome Coronavirus 2 Infection Among Pregnant Women and Vaccine-Induced Antibody Transfer to Infants. Open Forum Infect Dis 2023; 10:ofad204. [PMID: 37187508 PMCID: PMC10167992 DOI: 10.1093/ofid/ofad204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Background Early coronavirus disease 2019 (COVID-19) vaccine trials excluded pregnant women, resulting in limited data about immunogenicity and maternal-fetal antibody transfer, particularly by gestational timing of vaccination. Methods In this multicenter observational immunogenicity study, pregnant and nonpregnant women receiving COVID-19 vaccines were prospectively enrolled. Participants had sera collected before vaccination, at 14-28 days after each vaccine dose, at delivery (umbilical cord and peripheral), and from their infants at 3 and 6 months. Geometric mean titers (GMTs) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ID50 neutralizing antibody (nAb) against D614G-like viruses were compared by participant characteristics. Results Overall, 23 nonpregnant and 85 pregnant participants (trimester of first vaccine dose: 10 first, 47 second, 28 third) were enrolled. Ninety-three percent (76/82 with blood samples) of pregnant participants had detectable SARS-CoV-2 nAb after 2 vaccine doses, but GMTs (95% confidence intervals) were lower in pregnant participants than nonpregnant participants (1722 [1136-2612] vs 4419 [2012-9703]; P = .04). By 3 and 6 months, 28% and 74% of infants, respectively, of vaccinated participants had no detectable nAb to D614G-like viruses. Among the 71 pregnant participants without detectable nAb before vaccination, cord blood GMTs at delivery were 5-fold higher among participants vaccinated during the third versus first trimester, and cord blood nAb titers appeared inversely correlated with weeks since first vaccine dose (R2 = 0.06, P = .06). Conclusions Though most pregnant women develop nAb after 2 doses of mRNA COVID-19 vaccines, this analysis suggests that infant protection from maternal vaccination varies by gestational timing of vaccination and wanes. Additional prevention strategies such as caregiver vaccination may warrant consideration to optimize infant protection.
Collapse
Affiliation(s)
- Fatimah S Dawood
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Alan Tita
- Center for Women's Reproductive Health and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Melissa S Stockwell
- New York–Presbyterian Hospital, New York, New York, USA
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
- Department of Population and Family Health, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Kristina Wielgosz
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Cynthia Gyamfi-Bannerman
- New York–Presbyterian Hospital, New York, New York, USA
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, California, USA
| | - Ashley Battarbee
- Center for Women's Reproductive Health and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Natalie Thornburg
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sascha Ellington
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Romeo R Galang
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Kelly Vorwaller
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | | | | | - Mickey Parks
- Center for Women's Reproductive Health and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Emily Powers
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
| | - Marie Gibson
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Michael Varner
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| |
Collapse
|
24
|
Lipschuetz M, Guedalia J, Cohen SM, Sompolinsky Y, Shefer G, Melul E, Ergaz-Shaltiel Z, Goldman-Wohl D, Yagel S, Calderon-Margalit R, Beharier O. Maternal third dose of BNT162b2 mRNA vaccine and risk of infant COVID-19 hospitalization. Nat Med 2023; 29:1155-1163. [PMID: 36959421 DOI: 10.1038/s41591-023-02270-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/22/2023] [Indexed: 03/25/2023]
Abstract
Infants are at a higher risk of Coronavirus Disease 2019 (COVID-19)-related hospitalizations compared to older children. In this study, we investigated the effect of the recommended third maternal dose of BNT162b2 COVID-19 vaccine during pregnancy on rates of infant COVID-19-related hospitalizations. We conducted a nationwide cohort study of all live-born infants delivered in Israel between 24 August 2021 and 15 March 2022 to estimate the effectiveness of the third booster dose versus the second dose against infant COVID-19-related hospitalizations. Data were analyzed for the overall study period, and the Delta and Omicron periods were analyzed separately. Cox proportional hazard regression models estimated hazard ratios and 95% confidence intervals (CIs) for infant hospitalizations according to maternal vaccination status at delivery. Among 48,868 live-born infants included in the analysis, rates of COVID-19 hospitalization were 0.4%, 0.6% and 0.7% in the third-dose, second-dose and unvaccinated groups, respectively. Compared to the second dose, the third dose was associated with reduced infant hospitalization with estimated effectiveness of 53% (95% CI: 36-65%). Greater protection was associated with a shorter interval between vaccination and delivery. A third maternal dose during pregnancy reduced the risk of infant hospitalization for COVID-19 during the first 4 months of life, supporting clinical and public health guidance for maternal booster vaccination to prevent infant COVID-19 hospitalization.
Collapse
Affiliation(s)
- Michal Lipschuetz
- Obstetrics & Gynecology Division Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel
- Henrietta Szold Hadassah Hebrew University School of Nursing in the Faculty of Medicine, Jerusalem, Israel
| | - Joshua Guedalia
- Obstetrics & Gynecology Division Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sarah M Cohen
- Obstetrics & Gynecology Division Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yishai Sompolinsky
- Obstetrics & Gynecology Division Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Galit Shefer
- TIMNA-Israel Ministry of Health's Big Data Platform, Israel Ministry of Health, Jerusalem, Israel
| | - Eli Melul
- TIMNA-Israel Ministry of Health's Big Data Platform, Israel Ministry of Health, Jerusalem, Israel
| | | | - Debra Goldman-Wohl
- Obstetrics & Gynecology Division Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Simcha Yagel
- Obstetrics & Gynecology Division Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ronit Calderon-Margalit
- Braun School of Public Health, Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofer Beharier
- Obstetrics & Gynecology Division Hadassah Medical Center, Faculty of Medicine of the Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
25
|
Ahmed AK, Sijercic VC, Sayad R, Ruthig GR, Abdelwahab SF, El-Mokhtar MA, Sayed IM. Risks and Preventions for Pregnant Women and Their Preterm Infants in a World with COVID-19: A Narrative Review. Vaccines (Basel) 2023; 11:640. [PMID: 36992224 PMCID: PMC10056995 DOI: 10.3390/vaccines11030640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/05/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
(1) Background and Aim: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is linked to increasing cases of coronavirus disease 2019 (COVID-19) around the world. COVID-19 infections have an important impact on pregnancy, preterm birth (PTB) and delivery. Although several complications have been reported in infected pregnant women, the effect of infection on PTB is controversial. The purpose of this study was to summarize the existing literature on the effects and complications of COVID-19 on the health of pregnant women and preterm babies and its impact on the incidence of PTB. We also discuss the effect of current COVID-19 vaccines during pregnancy. (2) Methods: We carried out a systematic search of MEDLINE, Embase, and PubMed for studies on preterm births associated with COVID-19. (3) Results and Conclusions: We discovered contradictory results regarding the prevalence of PTB during the pandemic compared to earlier years. While most studies indicated an increase in PTBs with COVID-19, some indicated a decline in the preterm delivery rate during this time. During pregnancy, COVID-19 infection can increase the incidence of cesarean section, stillbirth, ICU admission, preeclampsia/eclampsia, and mortality rates. In the treatment of pregnant women with severe COVID-19, methylprednisolone was favored over prednisolone, and a brief course of dexamethasone is advised for pregnant women with anticipated PTB to accelerate the development of the fetal lung. Generally, vaccination for COVID-19 in pregnant and lactating women stimulates anti-SARS-CoV2 immune responses, and it does not result in any noteworthy negative reactions or outcomes for the mother or baby.
Collapse
Affiliation(s)
| | | | - Reem Sayad
- Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Gregory R. Ruthig
- Department of Biology, North Central College, Naperville, IL 60540, USA
| | - Sayed F. Abdelwahab
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Mohamed A. El-Mokhtar
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
- Microbiology and Immunology Department, Faculty of Pharmacy, Sphinx University, Assiut 71515, Egypt
| | - Ibrahim M. Sayed
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| |
Collapse
|
26
|
Zilver SJM, de Groot CJM, Grobben M, Remmelzwaal S, Burgers E, Nunez Velasco D, Juncker HG, van Keulen BJ, van Goudoever JB, de Leeuw RA, van Gils MJ, Ris-Stalpers C, van Leeuwen E. Vaccination from the early second trimester onwards gives a robust SARS-CoV-2 antibody response throughout pregnancy and provides antibodies for the neonate. Int J Infect Dis 2023; 130:126-135. [PMID: 36868302 PMCID: PMC9977072 DOI: 10.1016/j.ijid.2023.02.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
OBJECTIVES Preventative measures against Coronavirus Disease 2019 (COVID-19) are essential for pregnant women; they are particularly vulnerable to emerging infectious pathogens due to alterations in their physiology. We aimed to determine the optimum timing of vaccination to protect pregnant women and their neonates from COVID-19. METHODS A prospective observational longitudinal cohort study in pregnant women who received COVID-19 vaccination. We collected blood samples to evaluate levels of anti-spike, receptor binding domain (RBD) and nucleocapsid antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) before vaccination, and 15 days after first and second vaccination. We determined neutralizing antibodies from mother-infant dyads in maternal and umbilical cord blood at birth. If available, IgA was measured in human milk. RESULTS We included 178 pregnant women. Median anti-spike IgG levels increased significantly from 1.8 to 5431 binding antibody units/milliliter (BAU/mL) and RBD from 6 to 4466 BAU/mL. Virus neutralization showed similar results between different weeks' gestations at vaccination (p > 0.3). CONCLUSIONS We advise vaccination in the early second trimester of pregnancy for the optimum balance between maternal antibody response and placental antibody transfer to the neonate.
Collapse
Affiliation(s)
- S J M Zilver
- Amsterdam UMC location Vrije Universiteit Amsterdam, department of obstetrics and gynecology, Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Reproduction and Development research institute, Amsterdam, The Netherlands.
| | - C J M de Groot
- Amsterdam Reproduction and Development research institute, Amsterdam, The Netherlands; Amsterdam UMC location University of Amsterdam, department of obstetrics and gynecology, Meibergdreef 9, Amsterdam, the Netherlands
| | - M Grobben
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, The Netherlands
| | - S Remmelzwaal
- Amsterdam UMC location Vrije Universiteit Amsterdam, Epidemiology and Data Science, De Boelelaan 1117, Amsterdam, Netherlands
| | - E Burgers
- Amsterdam UMC location Vrije Universiteit Amsterdam, department of obstetrics and gynecology, Boelelaan 1117, Amsterdam, the Netherlands
| | - D Nunez Velasco
- Amsterdam UMC location University of Amsterdam, department of obstetrics and gynecology, Meibergdreef 9, Amsterdam, the Netherlands
| | - H G Juncker
- Amsterdam Reproduction and Development research institute, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit, University of Amsterdam, department of pediatrics, Emma Children's Hospital, Meibergdreef 9, Amsterdam, The Netherlands
| | - B J van Keulen
- Amsterdam Reproduction and Development research institute, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit, University of Amsterdam, department of pediatrics, Emma Children's Hospital, Meibergdreef 9, Amsterdam, The Netherlands
| | - J B van Goudoever
- Amsterdam Reproduction and Development research institute, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit, University of Amsterdam, department of pediatrics, Emma Children's Hospital, Meibergdreef 9, Amsterdam, The Netherlands
| | - R A de Leeuw
- Amsterdam UMC location University of Amsterdam, department of obstetrics and gynecology, Meibergdreef 9, Amsterdam, the Netherlands
| | - M J van Gils
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, The Netherlands
| | - C Ris-Stalpers
- Amsterdam Reproduction and Development research institute, Amsterdam, The Netherlands; Amsterdam UMC location University of Amsterdam, department of obstetrics and gynecology, Meibergdreef 9, Amsterdam, the Netherlands
| | - E van Leeuwen
- Amsterdam Reproduction and Development research institute, Amsterdam, The Netherlands; Amsterdam UMC location University of Amsterdam, department of obstetrics and gynecology, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
27
|
Romero-Ibarguengoitia ME, Flores-Salazar ZL, Arroyo-García KD, Soto-Gámez R, Leal-Meléndez JA, René Garza-Herrera M, Bennett-Vidales G, Cabrera MH, González-Habib R, Jiménez LP, Garza-Bulnes R, Barco-Flores IA, Castillo-Figueroa LF, Garza-Silva A, Rivera-Cavazos A, Rivera-Salinas D, González-Cantú A, Sanz-Sánchez MÁ. Evaluation of Transplacental Antibody Transfer in Pregnant Women Immunized with Different SARS-CoV-2 Homologous or Heterologous Schemes. Vaccines (Basel) 2023; 11:vaccines11020415. [PMID: 36851294 PMCID: PMC9961240 DOI: 10.3390/vaccines11020415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
There is scarce information related to transplacental antibody transfer against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with different homologous and heterologous vaccination schemes. This study aimed to correlate the magnitude of transplacental transfer anti-SARS-CoV-2 antibodies in different homologous and heterologous schemes. An observational cross-sectional study was developed to identify pregnant women vaccinated against SARS-CoV-2. They were questioned about their immunization status; blood samples from the mother, umbilical cord during labor, and the newborn 72 h after birth were taken to measure anti-S1 and anti-S2 specific IgG antibodies for SARS-CoV-2. We recruited 104 women with a median age of 29 (SD 1.17). We found antibodies in all newborns with vaccinated mothers. Homologous BNT162b2 mRNA regimen had the highest mean (SD) antibody titers (AU/mL) in maternal (994.93 (3.08), p = 0.039), umbilical cord (1316.43 (2.79), p = 0.016), and newborn (1192.02 (3.55), p = 0.020) blood. The generalized linear model showed a positive effect over antibodies with at least one dose in maternal (β = -1.1, p = 0.002) and newborn (β= -0.717, p = 0.044) blood, and with two doses (β = -0.684, p = 0.026) in umbilical cord blood. In conclusion, antibodies were detected in all vaccinated women and their newborns. Transfer of antibodies was found from the first dose, and the levels increased with the number of vaccine doses. Vaccination should be encouraged in pregnant women with any available scheme.
Collapse
Affiliation(s)
- Maria Elena Romero-Ibarguengoitia
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Dirección de Enseñanza e Investigación en Salud, Hospital Christus Muguerza Sistema de Salud, Calle Miguel Hidalgo y Costilla 2525, Obispado, Monterrey 64000, Nuevo León, Mexico
- Correspondence: ; Tel.: +52-81-8865-5656
| | - Zulema Lourdes Flores-Salazar
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Kimberly Dariela Arroyo-García
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Departamento de Ginecología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Rafael Soto-Gámez
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Departamento de Ginecología, Hospital Clínica Nova de Monterrey, Avenida del Bosque 139, Cuauhtémoc, Cuauhtémoc, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | | | | | - Gordon Bennett-Vidales
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Mauricio Hurtado Cabrera
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Roberto González-Habib
- Departamento de Ginecología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
- Dirección de Enseñanza e Investigación en Salud, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Liliann Peña Jiménez
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Raúl Garza-Bulnes
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | | | - Luis Fernando Castillo-Figueroa
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
- Departamento de Neonatología, Hospital Clínica Nova de Monterrey, Avenida del Bosque 139, Cuauhtémoc, Cuauhtémoc, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Arnulfo Garza-Silva
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Andrea Rivera-Cavazos
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Diego Rivera-Salinas
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Arnulfo González-Cantú
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Dirección de Enseñanza e Investigación en Salud, Hospital Christus Muguerza Sistema de Salud, Calle Miguel Hidalgo y Costilla 2525, Obispado, Monterrey 64000, Nuevo León, Mexico
| | - Miguel Ángel Sanz-Sánchez
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| |
Collapse
|
28
|
Rzymski P, Szuster-Ciesielska A, Dzieciątkowski T, Gwenzi W, Fal A. mRNA vaccines: The future of prevention of viral infections? J Med Virol 2023; 95:e28572. [PMID: 36762592 DOI: 10.1002/jmv.28572] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023]
Abstract
Messenger RNA (mRNA) vaccines against COVID-19 are the first authorized biological preparations developed using this platform. During the pandemic, their administration has been proven to be a life-saving intervention. Here, we review the main advantages of using mRNA vaccines, identify further technological challenges to be met during the development of the mRNA platform, and provide an update on the clinical progress on leading mRNA vaccine candidates against different viruses that include influenza viruses, human immunodeficiency virus 1, respiratory syncytial virus, Nipah virus, Zika virus, human cytomegalovirus, and Epstein-Barr virus. The prospects and challenges of manufacturing mRNA vaccines in low-income countries are also discussed. The ongoing interest and research in mRNA technology are likely to overcome some existing challenges for this technology (e.g., related to storage conditions and immunogenicity of some components of lipid nanoparticles) and enhance the portfolio of vaccines against diseases for which classical formulations are already authorized. It may also open novel pathways of protection against infections and their consequences for which no safe and efficient immunization methods are currently available.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.,Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), Poznań, Poland
| | - Agnieszka Szuster-Ciesielska
- Department of Virology and Immunology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | | | - Willis Gwenzi
- Alexander von Humboldt Fellow & Guest Professor, Grassland Science and Renewable Plant Resources, Faculty of Organic Agricultural Sciences, Universität Kassel, Witzenhausen, Germany.,Alexander von Humboldt Fellow & Guest Professor, Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
| | - Andrzej Fal
- Collegium Medicum, Warsaw Faculty of Medicine, Cardinal Stefan Wyszynski University, Warsaw, Poland.,Department of Public Health, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
29
|
Pongsatha S, Chawansuntati K, Sakkhachornphop S, Tongsong T. Placental Transfer Immunity to the Newborns in a Twin Pregnant Women Vaccinated with Heterologous CoronaVac-ChAdOx1. Vaccines (Basel) 2023; 11:116. [PMID: 36679961 PMCID: PMC9864723 DOI: 10.3390/vaccines11010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
Pregnant women who receive the COVID-19 vaccine develop anti-SARS-CoV-2 antibodies, which can be transferred to the fetus. However, the effectiveness of placental transfer has not been evaluated in twin pregnancy, especially in cases vaccinated with heterologous CoronaVac (Sinovac)-ChAdOx1 (Oxford-AstraZeneca) regimen, which was commonly used in many countries. Case: A 34-year-old Thai woman with a twin pregnancy attended our antenatal care clinic at 21 + 2 weeks of gestation and requested COVID-19 vaccination. Her medical history and physical examination were unremarkable. She had not received COVID-19 vaccination before. Ultrasound screening for fetal anomaly revealed a dichorion diamnion twin pregnancy. Both twins showed no structural anomaly. She received the CoronaVac vaccine at 21 + 2 weeks of gestation without serious side effects and the ChAdOx1 vaccine at 24 + 2 weeks of gestation. Cesarean delivery was performed at 36 + 5 weeks of gestation, giving birth to the two healthy babies. The levels of anti-spike protein IgG levels (BAU/mL) in maternal blood just before delivery and umbilical cord blood of the two newborns were 313.349, 678.219, and 874.853, respectively. The levels of % inhibition (wild-type and delta) in the two newborns were also higher than those in the mother. In conclusion, heterologous CoronaVac-ChAdOx1-S vaccination in a twin pregnancy could effectively provide protective immunity to both twin newborns. The antibody levels in both were approximately two times higher than those in the mothers. This case report may serve as a reference in counseling couples with a twin pregnancy, while the studies on placental transfer of vaccine-derived antibodies in twin pregnancy are currently not available, especially in countries experiencing a vaccine shortage or unavailability of mRNA vaccines.
Collapse
Affiliation(s)
- Saipin Pongsatha
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | | | - Theera Tongsong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
30
|
Zhang D, Huang T, Chen Z, Zhang L, Gao Q, Liu G, Zheng J, Ding F. Systematic review and meta-analysis of neonatal outcomes of COVID-19 vaccination in pregnancy. Pediatr Res 2023:10.1038/s41390-022-02421-0. [PMID: 36596943 PMCID: PMC9808682 DOI: 10.1038/s41390-022-02421-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND The safety of coronavirus disease 2019 (COVID-19) vaccines during pregnancy is a particular concern. Here, we addressed the neonatal outcomes after maternal vaccination of COVID-19 during pregnancy. METHODS We systematically searched PubMed, EMBASE, and the WHO COVID-19 Database for studies on neonatal outcomes after maternal COVID-19 vaccination from inception to 3 July 2022. Main neonatal outcomes were related to preterm, small for gestation (SGA), NICU admission, low Apgar score at 5 min (<7), and additional neonatal outcomes such as gestation <34 weeks, low birth weight and some neonatal morbidity were all also analyzed. RESULTS A total of 15 studies were included. We found that maternal vaccination during pregnancy was related to the reduction rates of Preterm, SGA, Low Apgar score at 5 min (<7). In addition, there was no evidence of a higher risk of adverse neonatal outcomes after maternal vaccination of COVID-19 during pregnancy, including NICU admission, preterm birth with gestation <34 weeks, low birth weight, very low birth weight, congenital anomalies, and so on. CONCLUSIONS COVID-19 vaccination in pregnant women does not raise significant adverse effects on neonatal outcomes and is related to a protective effect on some neonatal outcomes. IMPACT Present study has addressed the neonatal outcomes after maternal vaccination of COVID-19 during pregnancy. COVID-19 vaccination in pregnant women does not raise significant adverse effects on neonatal outcomes and is related to a protective effect on some neonatal outcomes. The present study could encourage pregnant women to be vaccinated against COVID-19.
Collapse
Affiliation(s)
- Dingning Zhang
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000 Tianjin, China ,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000 Tianjin, China ,grid.216938.70000 0000 9878 7032Department of Neonatology, Nankai University Maternity Hospital, 300000 Tianjin, China
| | - Tingting Huang
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000 Tianjin, China ,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000 Tianjin, China ,grid.216938.70000 0000 9878 7032Department of Neonatology, Nankai University Maternity Hospital, 300000 Tianjin, China
| | - Zhihui Chen
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000 Tianjin, China ,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000 Tianjin, China ,grid.216938.70000 0000 9878 7032Department of Neonatology, Nankai University Maternity Hospital, 300000 Tianjin, China
| | - Lulu Zhang
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000 Tianjin, China ,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000 Tianjin, China ,grid.216938.70000 0000 9878 7032Department of Neonatology, Nankai University Maternity Hospital, 300000 Tianjin, China
| | - Qi Gao
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000 Tianjin, China ,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000 Tianjin, China ,grid.216938.70000 0000 9878 7032Department of Neonatology, Nankai University Maternity Hospital, 300000 Tianjin, China
| | - Ge Liu
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000 Tianjin, China ,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000 Tianjin, China ,grid.216938.70000 0000 9878 7032Department of Neonatology, Nankai University Maternity Hospital, 300000 Tianjin, China
| | - Jun Zheng
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000, Tianjin, China. .,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000, Tianjin, China. .,Department of Neonatology, Nankai University Maternity Hospital, 300000, Tianjin, China.
| | - Fangrui Ding
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, 300000, Tianjin, China. .,Tianjin Key Laboratory of Human Development and Reproductive Regulation, 300000, Tianjin, China. .,Department of Neonatology, Nankai University Maternity Hospital, 300000, Tianjin, China.
| |
Collapse
|
31
|
Uda K, Tsuge M, Yashiro M, Honda T, Tsukahara H. Need for re-evaluating the risk of coronavirus disease 2019 transmission to neonates. Pediatr Int 2023; 65:e15460. [PMID: 36567643 PMCID: PMC9880739 DOI: 10.1111/ped.15460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/21/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022]
Affiliation(s)
- Kazuhiro Uda
- Department of PediatricsOkayama University Hospital
| | - Mitsuru Tsuge
- Department of Pediatric Acute Diseases, Academic Field of Medicine, Dentistry, and Pharmaceutical ScienceOkayama University
| | | | - Tomoyuki Honda
- Department of Virology, Faculty of Medicine, Dentistry, and Pharmaceutical SciencesOkayama University
- Department of Virology, Graduate School of Medicine, Dentistry and Pharmaceutical ScienceOkayama University
| | - Hirokazu Tsukahara
- Department of Pediatrics, Faculty of Medicine, Dentistry, and Pharmaceutical SciencesOkayama University
| |
Collapse
|
32
|
Golan Y, Ilala M, Gay C, Hunagund S, Lin CY, Cassidy AG, Jigmeddagva U, Li L, Ozarslan N, Asiodu IV, Ahituv N, Flaherman VJ, Gaw SL, Prahl M. Milk antibody response after 3rd dose of COVID-19 mRNA vaccine and SARS-CoV-2 breakthrough infection and implications for infant protection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.12.12.22283367. [PMID: 36561188 PMCID: PMC9774223 DOI: 10.1101/2022.12.12.22283367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Anti-SARS-CoV-2 antibodies have been found in human-milk after COVID-19 infection and vaccination. However, little is known about their persistence in milk after booster vaccination and breakthrough infection. In this study, human-milk, saliva and blood samples were collected from 33 lactating individuals before and after mRNA-based vaccination and COVID-19 breakthrough infections. Antibody levels were measured using ELISA and symptoms were assessed using questionnaires. Evaluation of maternal and infant symptomatology revealed that infected mothers reported more symptoms than vaccinated mothers. We found that after vaccination, human-milk anti-SARS-CoV-2 antibodies persisted for up to 8 months. In addition, distinct patterns of human milk IgA and IgG production we observed after breakthrough infection compared to 3-dose vaccination series alone, indicating a differential central and mucosal immune profiles in hybrid compared with vaccine-induced immunity. To investigate passively-derived milk antibody protection in infants, we examined the persistence of these antibodies in infant saliva after breastfeeding. We found that IgA was more abundant in infant saliva compared to IgG and persist in infant saliva longer after feeding. Our results delineate the differences in milk antibody response to vaccination as compared to breakthrough infection and emphasize the importance of improving the secretion of IgA antibodies to human milk after vaccination to improve the protection of breastfeeding infants.
Collapse
Affiliation(s)
- Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, California, United States of America
| | - Mikias Ilala
- Department of Pediatrics, University of California, San Francisco, California, United States of America
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, United States of America
| | - Caryl Gay
- Department of Family Health Care Nursing, University of California, San Francisco, California, United States of America
| | - Soumya Hunagund
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Christine Y. Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Arianna G. Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Unurzul Jigmeddagva
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Ifeyinwa V. Asiodu
- Department of Family Health Care Nursing, University of California, San Francisco, California, United States of America
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, California, United States of America
| | - Valerie J. Flaherman
- Department of Pediatrics, University of California, San Francisco, California, United States of America
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Mary Prahl
- Department of Pediatrics, University of California, San Francisco, California, United States of America
- Division of Pediatric Infectious Diseases and Global Health, University of California, San Francisco, California, United States of America
| |
Collapse
|
33
|
Alcendor DJ, Matthews-Juarez P, Smoot D, Hildreth JEK, Tabatabai M, Wilus D, Brown KY, Juarez PD. The COVID-19 Vaccine and Pregnant Minority Women in the US: Implications for Improving Vaccine Confidence and Uptake. Vaccines (Basel) 2022; 10:2122. [PMID: 36560532 PMCID: PMC9784552 DOI: 10.3390/vaccines10122122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/25/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
The American College of Obstetricians and Gynecologists (AGOG) recommends the FDA-approved Pfizer and Moderna mRNA COVID-19 vaccines and boosters for all eligible pregnant women in the US. However, COVID-19 vaccine confidence and uptake among pregnant minority women have been poor. While the underlying reasons are unclear, they are likely to be associated with myths and misinformation about the vaccines. Direct and indirect factors that deter minority mothers in the US from receiving the mRNA COVID-19 vaccines require further investigation. Here, we examine the historical perspectives on vaccinations during pregnancy. We will examine the following aspects: (1) the influenza and tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis (Tdap) vaccinations during pregnancy; (2) the exclusion of pregnant and lactating women from COVID-19 vaccine trials; (3) COVID-19 vaccine safety during pregnancy, obstetric complications associated with symptomatic COVID-19 during pregnancy, COVID-19 vaccine hesitancy among pregnant minority women, and racial disparities experienced by pregnant minority women due to the COVID-19 pandemic as well as their potential impact on pregnancy care; and (4) strategies to improve COVID-19 vaccine confidence and uptake among pregnant minority women in the US. COVID-19 vaccine hesitancy among minority mothers can be mitigated by community engagement efforts that focus on COVID-19 vaccine education, awareness campaigns by trusted entities, and COVID-19-appropriate perinatal counseling aimed to improve COVID-19 vaccine confidence and uptake.
Collapse
Affiliation(s)
- Donald J. Alcendor
- Department of Microbiology, Immunology and Physiology, Center for AIDS Health Disparities Research, School of Medicine, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| | - Patricia Matthews-Juarez
- Department of Family & Community Medicine, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| | - Duane Smoot
- Department of Internal Medicine, School of Medicine, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| | - James E. K. Hildreth
- Department of Microbiology, Immunology and Physiology, Center for AIDS Health Disparities Research, School of Medicine, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
- Department of Family & Community Medicine, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
- Department of Internal Medicine, School of Medicine, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| | - Mohammad Tabatabai
- School of Graduate Studies and Research, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| | - Derek Wilus
- School of Graduate Studies and Research, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| | - Katherine Y. Brown
- Department of Family & Community Medicine, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| | - Paul D. Juarez
- Department of Family & Community Medicine, Meharry Medical College, 1005 D.B. Todd Jr. Blvd., Nashville, TN 37208, USA
| |
Collapse
|
34
|
Sunder A, Alqatari HM, Taha OE, Keshta MS, Bughamar FK, Darwish B. COVID-19 vaccinations in pregnancy: Save mother and baby from COVID-19 pandemic. Int J Gynaecol Obstet 2022; 160:864-873. [PMID: 36306398 PMCID: PMC9874773 DOI: 10.1002/ijgo.14532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVE The current study investigated the immune response of maternal coronavirus disease 2019 (COVID-19) vaccination and vertical transmission of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) and nucleocapsid (N) proteins. STUDY DESIGN This retrospective study included pregnant women in Bahrain Defense Force Hospital from March 2021 to September 2021 who were vaccinated with Sinopharm or Pfizer/BioNTech. Testing of anti-N and -S levels from paired samples of maternal and umbilical cord blood was performed at the time of delivery. The immune response to vaccination, association with maternal and fetal factors, and vertical transmission of antibodies were studied. RESULTS The current study included 79 pregnant women. The median gestational age for those vaccinated with Sinopharm was 28 weeks and those vaccinated with Pfizer was 31 weeks, with 100% of the vaccinated population generating antibodies and showing vertical transmission. The anti-N and -S titers and interval frequencies varied in both vaccinations. The anti-N and -S and transfer ratio statistically correlated with maternal age, gestational age at delivery, latency period, and birth weight of the neonates differently in both vaccines. In addition, the peak level of antibodies and transfer ratios varied. CONCLUSION Although variations are exhibited in both types of vaccination, the vaccinated pregnant population generated a significant level of anti-N and -S and showed vertical transmission.
Collapse
Affiliation(s)
| | - Huda Merza Alqatari
- School of Medicine, Royal College of Surgeons in Ireland‐BahrainBusaiteenBahrain
| | - Omer E. Taha
- School of Medicine, Royal College of Surgeons in Ireland‐BahrainBusaiteenBahrain
| | - Mohamed S. Keshta
- School of Medicine, Royal College of Surgeons in Ireland‐BahrainBusaiteenBahrain
| | | | | |
Collapse
|