108
the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 1 RESEARCH Clinical manifestations, risk factors, and maternal and perinatal outcomes of coronavirus disease 2019 in pregnancy: living systematic review and meta-analysis John Allotey, 1,2 Elena Stallings, 3,4 Mercedes Bonet, 5 Magnus Yap, 6 Shaunak Chatterjee, 6 Tania Kew, 6 Luke Debenham, 6 Anna Clavé Llavall, 6 Anushka Dixit, 6 Dengyi Zhou, 6 Rishab Balaji, 6 Siang Ing Lee, 1 Xiu Qiu, 7,8,9 Mingyang Yuan, 1,7 Dyuti Coomar, 1 Madelon van Wely, 10 Elizabeth van Leeuwen, 11 Elena Kostova, 10 Heinke Kunst, 12,13 Asma Khalil, 14 Simon Tiberi, 12,13 Vanessa Brizuela, 5 Nathalie Broutet, 5 Edna Kara, 3 Caron Rahn Kim, 5 Anna Thorson, 5 Olufemi T Oladapo, 5 Lynne Mofenson, 15 Javier Zamora, 3,4,16 Shakila Thangaratinam, 2,17 for PregCOV-19 Living Systematic Review Consortium ABSTRACT OBJECTIVE To determine the clinical manifestations, risk factors, and maternal and perinatal outcomes in pregnant and recently pregnant women with suspected or confirmed coronavirus disease 2019 (covid-19). DESIGN Living systematic review and meta-analysis. DATA SOURCES Medline, Embase, Cochrane database, WHO COVID-19 database, China National Knowledge Infrastructure (CNKI), and Wanfang databases from 1 December 2019 to 26 June 2020, along with preprint servers, social media, and reference lists. STUDY SELECTION Cohort studies reporting the rates, clinical manifestations (symptoms, laboratory and radiological findings), risk factors, and maternal and perinatal outcomes in pregnant and recently pregnant women with suspected or confirmed covid-19. DATA EXTRACTION At least two researchers independently extracted the data and assessed study quality. Random effects meta-analysis was performed, with estimates pooled as odds ratios and proportions with 95% confidence intervals. All analyses will be updated regularly. RESULTS 77 studies were included. Overall, 10% (95% confidence interval 7% to14%; 28 studies, 11 432 women) of pregnant and recently pregnant women attending or admitted to hospital for any reason were diagnosed as having suspected or confirmed covid-19. The most common clinical manifestations of covid-19 in pregnancy were fever (40%) and cough (39%). Compared with non-pregnant women of reproductive age, pregnant and recently pregnant women with covid-19 were less likely to report symptoms of fever (odds ratio 0.43, 95% confidence interval 0.22 to 0.85; I 2 =74%; 5 studies; 80 521 women) and myalgia (0.48, 0.45 to 0.51; I 2 =0%; 3 studies; 80 409 women) and were more likely to need admission to an intensive care unit (1.62, 1.33 to 1.96; I 2 =0%) and invasive ventilation (1.88, 1.36 to 2.60; I 2 =0%; 4 studies, 91 606 women). 73 pregnant women (0.1%, 26 studies, 11 580 women) with confirmed covid-19 died from any cause. Increased maternal age (1.78, 1.25 to 2.55; I 2 =9%; 4 studies; 1058 women), high body mass index (2.38, 1.67 to 3.39; I 2 =0%; 3 studies; 877 women), chronic hypertension (2.0, 1.14 to 3.48; I 2 =0%; 2 studies; 858 women), and pre-existing diabetes (2.51, 1.31 to 4.80; I 2 =12%; 2 studies; 858 women) were associated with severe covid-19 in pregnancy. Pre-existing maternal comorbidity was a risk factor for admission to an intensive care unit (4.21, 1.06 to 16.72; I 2 =0%; 2 studies; 320 women) and invasive ventilation (4.48, 1.40 to 14.37; I 2 =0%; 2 studies; 313 women). Spontaneous preterm birth rate was 6% (95% confidence interval 3% to 9%; I 2 =55%; 10 studies; 870 women) in women with covid-19. The odds of any preterm birth (3.01, 95% confidence interval 1.16 to 7.85; I 2 =1%; 2 studies; 339 women) was high in pregnant women with covid-19 compared with those without the disease. A quarter of all neonates born to mothers with covid-19 were admitted to the neonatal unit (25%) and were at increased risk of admission (odds ratio 3.13, 95% confidence interval 2.05 to 4.78, I 2 =not estimable; 1 study, 1121 neonates) than those born to mothers without covid-19. For numbered affiliations see end of the article. Correspondence to: S Thangaratinam [email protected] (or @thangaratinam on Twitter: ORCID 0000-0002-4254-460X) Additional material is published online only. To view please visit the journal online. Cite this as: BMJ 2020;370:m3320 http://dx.doi.org/10.1136/bmj.m3320 Accepted: 23 August 2020 WHAT IS ALREADY KNOWN ON THIS TOPIC Pregnant women are considered to be a high risk group for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and the potential adverse effects of the virus on maternal and perinatal outcomes are of concern In non-pregnant populations admitted to hospital with coronavirus disease 2019 (covid-19) the most common symptoms are fever, cough, and dyspnoea, reported in more than two thirds of individuals Advancing age, high body mass index, non-white ethnicity, and pre-existing comorbidities are risk factors for severe covid-19 in the general population WHAT THIS STUDY ADDS Pregnant and recently pregnant women with covid-19 diagnosed in hospital are less likely to manifest symptoms of fever and myalgia than non-pregnant women of reproductive age and might be at increased risk of admission to an intensive care unit Risk factors for severe covid-19 in pregnancy include increasing maternal age, high body mass index, and pre-existing comorbidities Pregnant women with covid-19 are more likely to experience preterm birth and their neonates are more likely to be admitted to a neonatal unit copyright. on 15 September 2020 at Bibliotheque Faculte Medecine Geneve. Protected by http://www.bmj.com/ BMJ: first published as 10.1136/bmj.m3320 on 1 September 2020. Downloaded from

Clinical manifestations, risk factors, and maternal and

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Clinical manifestations, risk factors, and maternal and

the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 1

RESEARCH

Clinical manifestations, risk factors, and maternal and perinatal outcomes of coronavirus disease 2019 in pregnancy: living systematic review and meta-analysisJohn Allotey,1,2 Elena Stallings,3,4 Mercedes Bonet,5 Magnus Yap,6 Shaunak Chatterjee,6 Tania Kew,6 Luke Debenham,6 Anna Clavé Llavall,6 Anushka Dixit,6 Dengyi Zhou,6 Rishab Balaji,6 Siang Ing Lee,1 Xiu Qiu,7,8,9 Mingyang Yuan,1,7 Dyuti Coomar,1 Madelon van Wely,10 Elizabeth van Leeuwen,11 Elena Kostova,10 Heinke Kunst,12,13 Asma Khalil,14 Simon Tiberi,12,13 Vanessa Brizuela,5 Nathalie Broutet,5 Edna Kara,3 Caron Rahn Kim,5 Anna Thorson,5 Olufemi T Oladapo,5 Lynne Mofenson,15 Javier Zamora,3,4,16 Shakila Thangaratinam,2,17 for PregCOV-19 Living Systematic Review Consortium

AbstrActObjectiveTo determine the clinical manifestations, risk factors, and maternal and perinatal outcomes in pregnant and recently pregnant women with suspected or confirmed coronavirus disease 2019 (covid-19).DesignLiving systematic review and meta-analysis.Data sOurcesMedline, Embase, Cochrane database, WHO COVID-19 database, China National Knowledge Infrastructure (CNKI), and Wanfang databases from 1 December 2019 to 26 June 2020, along with preprint servers, social media, and reference lists.stuDy selectiOnCohort studies reporting the rates, clinical manifestations (symptoms, laboratory and radiological findings), risk factors, and maternal and perinatal outcomes in pregnant and recently pregnant women with suspected or confirmed covid-19.Data extractiOnAt least two researchers independently extracted the data and assessed study quality. Random effects

meta-analysis was performed, with estimates pooled as odds ratios and proportions with 95% confidence intervals. All analyses will be updated regularly.results77 studies were included. Overall, 10% (95% confidence interval 7% to14%; 28 studies, 11 432 women) of pregnant and recently pregnant women attending or admitted to hospital for any reason were diagnosed as having suspected or confirmed covid-19. The most common clinical manifestations of covid-19 in pregnancy were fever (40%) and cough (39%). Compared with non-pregnant women of reproductive age, pregnant and recently pregnant women with covid-19 were less likely to report symptoms of fever (odds ratio 0.43, 95% confidence interval 0.22 to 0.85; I2=74%; 5 studies; 80 521 women) and myalgia (0.48, 0.45 to 0.51; I2=0%; 3 studies; 80 409 women) and were more likely to need admission to an intensive care unit (1.62, 1.33 to 1.96; I2=0%) and invasive ventilation (1.88, 1.36 to 2.60; I2=0%; 4 studies, 91 606 women). 73 pregnant women (0.1%, 26 studies, 11 580 women) with confirmed covid-19 died from any cause. Increased maternal age (1.78, 1.25 to 2.55; I2=9%; 4 studies; 1058 women), high body mass index (2.38, 1.67 to 3.39; I2=0%; 3 studies; 877 women), chronic hypertension (2.0, 1.14 to 3.48; I2=0%; 2 studies; 858 women), and pre-existing diabetes (2.51, 1.31 to 4.80; I2=12%; 2 studies; 858 women) were associated with severe covid-19 in pregnancy. Pre-existing maternal comorbidity was a risk factor for admission to an intensive care unit (4.21, 1.06 to 16.72; I2=0%; 2 studies; 320 women) and invasive ventilation (4.48, 1.40 to 14.37; I2=0%; 2 studies; 313 women). Spontaneous preterm birth rate was 6% (95% confidence interval 3% to 9%; I2=55%; 10 studies; 870 women) in women with covid-19. The odds of any preterm birth (3.01, 95% confidence interval 1.16 to 7.85; I2=1%; 2 studies; 339 women) was high in pregnant women with covid-19 compared with those without the disease. A quarter of all neonates born to mothers with covid-19 were admitted to the neonatal unit (25%) and were at increased risk of admission (odds ratio 3.13, 95% confidence interval 2.05 to 4.78, I2=not estimable; 1 study, 1121 neonates) than those born to mothers without covid-19.

For numbered affiliations see end of the article.Correspondence to: S Thangaratinam [email protected] (or @thangaratinam on Twitter: ORCID 0000-0002-4254-460X)Additional material is published online only. To view please visit the journal online.cite this as: BMJ2020;370:m3320http://dx.doi.org/10.1136/bmj.m3320

Accepted: 23 August 2020

WhAt is AlreAdy knoWn on this topicPregnant women are considered to be a high risk group for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and the potential adverse effects of the virus on maternal and perinatal outcomes are of concernIn non-pregnant populations admitted to hospital with coronavirus disease 2019 (covid-19) the most common symptoms are fever, cough, and dyspnoea, reported in more than two thirds of individualsAdvancing age, high body mass index, non-white ethnicity, and pre-existing comorbidities are risk factors for severe covid-19 in the general population

WhAt this study AddsPregnant and recently pregnant women with covid-19 diagnosed in hospital are less likely to manifest symptoms of fever and myalgia than non-pregnant women of reproductive age and might be at increased risk of admission to an intensive care unitRisk factors for severe covid-19 in pregnancy include increasing maternal age, high body mass index, and pre-existing comorbiditiesPregnant women with covid-19 are more likely to experience preterm birth and their neonates are more likely to be admitted to a neonatal unit

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 2: Clinical manifestations, risk factors, and maternal and

RESEARCH

2 doi: 10.1136/bmj.m3320 | BMJ 2020;370:m3320 | the bmj

cOnclusiOnPregnant and recently pregnant women are less likely to manifest covid-19 related symptoms of fever and myalgia than non-pregnant women of reproductive age and are potentially more likely to need intensive care treatment for covid-19. Pre-existing comorbidities, high maternal age, and high body mass index seem to be risk factors for severe covid-19. Preterm birth rates are high in pregnant women with covid-19 than in pregnant women without the disease.systematic review registratiOnPROSPERO CRD42020178076.reaDers’ nOteThis article is a living systematic review that will be updated to reflect emerging evidence. Updates may occur for up to two years from the date of original publication.

introductionSince the first report (December 2019) of the novel coronavirus disease 2019 (covid-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the number of confirmed cases and associated mortality and morbidity have increased rapidly.1 2 Pregnant women are considered a high risk group because of concerns about the effect of covid-19 on them during and after pregnancy, and on their neonates.3 Quantification of the rates of covid-19, its risk factors, clinical manifestations, and outcomes is key to planning clinical maternal care and management in an evolving pandemic scenario.4

Publications on covid-19 in pregnancy have risen steeply through individual case reports, case series, observational studies, and systematic reviews. As of 26 June 2020, more than 86 reviews have been published in this area,5-10 with a further 94 registered in PROSPERO.8 11 The early reviews mostly included case reports and case series that were often inappropriately meta-analysed, leading to biased estimates.12 Subsequent reviews differed little from each other, often including similar primary studies, many with duplicate data. These reviews became quickly outdated as new evidence emerged. To date, no review has comprehensively evaluated the comparative data concerning pregnant and recently pregnant women and non-pregnant women with covid-19. Moreover, the sampling frames in primary studies have varied, ranging from universal SARS-CoV-2 testing for all pregnant women admitted to hospital13 14 to symptom based testing.15 16 Testing strategies have also differed within and between countries, with diagnosis in many early studies based on epidemiological risk assessment and clinical features without confirmed infection, which need to be considered in the analysis.17 Limitations in the external and internal validity of studies make it challenging for guideline developers and policy makers to make evidence based recommendations for the management of pregnant and recently pregnant women with covid-19.

We began a living systematic review to determine the clinical manifestations of covid-19 in pregnant and recently pregnant women, identify the risk factors for complications, and quantify maternal and perinatal outcomes. This systematic review will be updated on a regular basis.

MethodsOur systematic review is based on a prospectively registered protocol (PROSPERO CRD42020178076; registered 22 April 2020)18 to evaluate a series of research questions on covid-19 during and after pregnancy. We report our findings on the rates, clinical manifestations, risk factors, and maternal and perinatal outcomes in women with covid-19 in line with the preferred reporting items for systematic reviews and meta-analyses (PRISMA) recommendations (see appendix 1). As more relevant data become available, we shall address the research questions in our published protocol. Each cycle of our living systematic review involves weekly search updates (rounds), with analysis performed every 2-4 weeks for our monthly reporting through a dedicated website, with early analysis if new definitive evidence emerges. We plan to regularly review the planned frequency of updates.

literature searchWe performed a systematic search of major databases: Medline, Embase, Cochrane database, WHO (World Health Organization) COVID-19 database, China National Knowledge Infrastructure (CNKI), and Wanfang databases from 1 December 2019 to 26 June 2020 for relevant studies on covid-19 in pregnant and recently pregnant women. To identify potential studies, we coordinated our search efforts with the Evidence for Policy and Practice Information and Co-ordinating Centre (EPPI-Centre), the WHO Library, and the Cochrane Gynaecology and Fertility group. Additional searches were conducted of preprint servers, blogs, websites that serve as repositories for covid-19 studies, social media, guidelines, and reference lists of included studies and unpublished data. We also searched the Living Overview of the Evidence (LOVE) platform from 11 to 26 June 2020.19 We contacted established groups that were coordinating or conducting surveillance and studies in pregnant women with covid-19, such as the WHO Maternal, Newborn, Child and Adolescent health (MNCAH) covid-19 research network and the International Network of Obstetric Survey Systems (INOSS) for information on published and upcoming data. No language restrictions were applied. Appendix 2 provides details of the search strategies and databases searched.

study selectionTwo reviewers independently selected studies using a two stage process: they first screened the titles and abstracts of studies and then assessed the full text of the selected studies in detail for eligibility. A total of eight reviewers contributed to study selection.

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 3: Clinical manifestations, risk factors, and maternal and

RESEARCH

the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 3

Disagreements were resolved through discussion with a third reviewer (ST or JA). We excluded studies if the duplicate data for all outcomes of interest were published elsewhere, as reported by the study authors, or when the characteristics of the mother or neonate matched the setting, characteristics, and duration of another study. When we suspected an overlap of data between studies, the study that provided comparative data was included. When there was uncertainty about duplicate data, we contacted the authors of primary studies.

We defined women as having confirmed covid-19 if they had laboratory confirmation of covid-19 infection irrespective of clinical signs and symptoms.20 Women with a diagnosis based only on clinical or radiological findings were defined as having suspected covid-19. The recently pregnant group comprised women in the postpartum and post-abortion period. We included studies that compared covid-19 rates, clinical manifestations (symptoms, laboratory and radiological results), risk factors, and associated mortality and morbidity between pregnant and recently pregnant and non-pregnant women of reproductive age, and those that compared maternal and perinatal outcomes in pregnant women with and without covid-19. Studies on non-comparative cohorts with a minimum of 10 participants were included if they reported on the rates and clinical manifestations of covid-19 and relevant outcomes in pregnant and recently pregnant women. We defined cohort studies as those that sampled participants on the basis of exposure, followed-up participants over time, and ascertained the outcomes.21 The PROSPERO protocol provides a full list of the risk factors, clinical features, and outcomes evaluated.18

The sampling frames for detecting covid-19 included universal screening and testing, when all women were assessed for covid-19 using reverse transcriptase polymerase chain reaction (RT-PCR) for SARS-CoV-2 or chest computed tomography; risk based testing on the basis of epidemiological history and clinical manifestations by National Health Commission of China (NHCC) guidelines17; and symptom based when testing was performed on women with symptoms and those with a history of contact with affected individuals. We defined the population as being selected when only specific groups of women were included, such as those undergoing caesarean section or in the third trimester. We categorised studies as a high risk group if only women with any pre-existing medical or obstetric risk factors were included, low risk if women did not have any risk factors, and any risk if all women were included.

study quality assessment and data extractionThe quality of the comparative cohort studies was assessed for selection, comparability, and outcome ascertainment bias using the Newcastle Ottawa scale.22 Studies achieving four stars for selection, two for comparability, and three for ascertainment of the outcome were considered to have a low risk of bias.

Studies achieving two or three stars for selection, one for comparability, and two for outcome ascertainment were considered to have a medium risk of bias, and any study achieving one star for selection or outcome ascertainment, or zero for any of the three domains, was regarded as having a high risk of bias. We assessed the quality of studies reporting on the prevalence of clinical manifestations or outcomes for internal and external validity using an existing tool.23 The following were considered as low risk of bias for external validity: representative of national population for relevant variables (population), representative of target population (sampling frame), random selection (selection bias), and more than 75% response rate in individuals with and without the outcome (non-response bias).23 Two indepen-dent reviewers extracted data using a pre-piloted form.

statistical analysisWe pooled the comparative dichotomous data using random effects meta-analysis and summarised the findings as odds ratios with 95% confidence intervals. To combine comparative continuous data with dichotomous data we transformed standardised mean differences to logarithm odds ratios, assuming a normal underlying distribution.24 We pooled the dichotomous non-comparative data for rates of clinical manifestations and maternal and perinatal outcomes as proportions with 95% confidence intervals using Dersimonian and Laird random effects meta-analysis after transforming data using Freeman-Tukey double arcsin transformation. Heterogeneity was reported as I2 statistics. We undertook subgroup analysis by country status (high versus low and middle income), sampling frame (universal, risk based, and symptom based testing, including not reported), and risk status of women in the studies (high, low, any). Sensitivity analysis was performed by restricting the analysis to women with confirmed covid-19, study quality (high, low), and population (unselected, selected). All analyses were done with Stata (version 16).

Patient and public involvementThe study was supported by Katie’s Team, a dedicated patients and public involvement group in Women’s Health. The team was involved in the conduct, interpretation, and reporting of this living systematic review through participation in virtual meetings.

resultsAfter removing duplicates from 49 684 citations, 20 625 unique citations were identified and 77 cohort studies (55 comparative, 22 non-comparative) were included in the systematic review (fig 1).

characteristics of included studiesOf the 77 studies, 26 (34%) were from the United States, 24 from China (31%), seven from Italy, six from Spain, three each from the United Kingdom and France, and one each from Belgium, Brazil, Denmark, Israel, Japan,

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 4: Clinical manifestations, risk factors, and maternal and

RESEARCH

4 doi: 10.1136/bmj.m3320 | BMJ 2020;370:m3320 | the bmj

Mexico, the Netherlands, and Portugal. All the studies tested respiratory samples using RT-PCR to confirm the presence of SARS-CoV-2; 23 studies additionally diagnosed covid-19 based on clinical suspicion. Eight studies (95 247 women) compared pregnant populations with non-pregnant populations,25-32 and four studies (2230 women) compared pregnant women with covid-19 versus pregnant women without covid-19.33-36 Forty cohort studies reported on clinical manifestations (13 018 pregnant, 85 084 non-pregnant women),25-32 35-66 45 studies reported on covid-19 related maternal outcomes (14 094 pregnant, 85 169 non-pregnant women),25-32 35-51 53-59 61-74 and 35 studies reported on pregnancy related maternal (6279 women) and perinatal outcomes (2557 neonates)13 25 27 29 30 32-41 43-47 49-50 54 55 57 59 61 62 64-67 69 70 75 (see appendix 3). The sampling frames included universal testing (29 studies), risk based NHCC guidelines (22 studies), and symptom based (19 studies) strategies. Eleven studies did not report the sampling strategy.

Quality of included studiesOverall, 67% (37/55) of the comparative cohort studies evaluated using the Newcastle Ottawa scale had an overall low risk of bias (see appendix 4a). Forty nine (89%) had a low risk of bias for study selection and six (11%) had a medium risk. The risk of bias for comparability of cohorts was low in nine of the studies (16%), medium in 45 (82%), and high in one (2%). For outcome assessment of the cohorts, 12 (22%) studies had a low risk of bias, 42 (76%) a medium risk, and one (2%) a high risk. Quality assessment of the prevalence studies for external validity showed a low risk of bias for representativeness in 13% (10/76) of the studies, sampling in 26% (20/76), selection in 74% (56/76), and non-response in 96% (73/76). For internal validity, there was low risk of bias for data collection in 95% (72/76) of the studies, case definition in 36% (27/76), measurement in 99% (75/76), differential verification in 86% (65/76), adequate follow-up in 22% (17/76), and appropriate numerator and denominator in 83% (63/76) (see appendix 4b).

Articles excludedIrrelevant articlesDuplicates

19 46029 059

Full text articles assessed for eligibility

Citations identified

1165

49 684

48 519

Articles excludedInappropriate populationInappropriate study designDuplicate publicationInappropriate outcomeInappropriate exposureArticle not found

457452125

3518

1

Electronic databases from inception to 26 June 2020Other sources* and reference lists

49 538146

Studies included (13 118 pregnant and recently pregnant women with covid-19;83 486 non-pregnant women of reproductive age with covid-19)

Prevalence of covid-19Risk factors for covid-19 and complicationsClinical manifestations of covid-19Covid-19 related outcomesPregnancy related maternal and perinatal outcomes

2652404535

1088

77

Fig 1 | study selection process. *twitter, national reports, blog by j thornton, Obg Project, cOviD-19 and Pregnancy cases, www.obgproject.com/2020/04/07/covid-19-research-watch-with-dr-jim-thornton/ (accessed 12 may 2020); ePPi-centre, cOviD-19: a living systematic map of evidence, http://eppi.ioe.ac.uk/cms/Projects/DepartmentofHealthandsocialcare/Publishedreviews/cOviD-19livingsystematicmapoftheevidence/tabid/3765/Default.aspx (accessed 12 may 2020); norwegian institute of Public Health, niPH systematic and living map on cOviD-19 evidence, www.nornesk.no/forskningskart/niPH_mainmap.html (accessed 19 may 2020); johns Hopkins university center for Humanitarian Health; cOviD-19, maternal and child Health, nutrition, http://hopkinshumanitarianhealth.org/empower/advocacy/covid-19/covid-19-children-and-nutrition/ (accessed 2 june 2020); researchgate, cOviD-19 research community, www.researchgate.net/community/cOviD-19 (accessed 2 june 2020); and living Overview of the evidence, coronavirus disease (cOviD-19), https://app.iloveevidence.com/loves/5e6fdb9669c00e4ac072701d?population=5d062d5fc80dd41e58ba8459 (accessed 16 june 2020)

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 5: Clinical manifestations, risk factors, and maternal and

RESEARCH

the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 5

rates of covid-19 in pregnant and recently pregnant womenThe overall rate of covid-19 diagnosis in pregnant and recently pregnant women attending or admitted to hospital for any reason was 10% (95% confidence interval 7% to 14%; 26 studies, 11 432 women; fig 2). Rates varied by sampling strategy: of the women sampled by universal screening, 7% (4% to 10%; 18 studies, 6247 women) were diagnosed as having covid-19 compared with 18% (10% to 28%; 8 studies, 4928 women) of women sampled on the basis of symptoms. All studies with a prevalence rate for covid-19 greater than 15% were from the US, except for one study, which was from France.76 One in 20 asymptomatic mothers (5%, 2% to 9%; 11 studies)

attending or admitted to hospital had a diagnosis of covid-19 (see appendix 5a). Three quarters (74%, 51% to 93%; 11 studies) of the 162 pregnant women with covid-19 in the universal screening population were asymptomatic (see appendix 5b). Based on data from a small number of studies, a diagnosis of covid-19 in pregnancy was associated with maternal obesity (odds ratio 1.75, 95% confidence interval 1.34 to 2.30; 1 study, 1080 women), pre-existing comorbidities (1.64, 1.25 to 2.13; 1 study, 1121 women), asthma (1.71, 1.03 to 2.84; 2 studies, 1250 women), history of covid-19 in the support person (44.56, 14.90 to 133.28; 1 study, 199 women), and gestational diabetes (2.42, 1.55 to 3.79; 1 study, 1121 women) (see appendix 6a).

Universal screening

Sutton 2020

Vintzileos 2020

Tassis 2020

Khalil 2020

Gagliardi 2020

Naqvi 2020

Ceulemans 2020

Miller 2020

Doria 2020

London 2020

Bianco 2020

Goldfarb 2020

LaCourse 2020

Ochiai 2020

Freiesleben 2020

Cosma 2020

Crovetto 2020

Emeruwa 2020

Subtotal: P=0.00; I2=95.1%

Symptom based screening

Blitz 2020

Campbell 2020

Fox 2020

Qadri 2020

Duffy 2020

London 2020

LaCourse 2020

Griffin 2020

Subtotal: P=0.00; I2=97.9%

Not known

Cohen

Overall: I2=96.99%, P=0.00;

estimated predictive interval

0.15 (0.11 to 0.21)

0.20 (0.14 to 0.27)

0.02 (0.01 to 0.06)

0.07 (0.04 to 0.13)

0.01 (0.00 to 0.02)

0.01 (0.00 to 0.07)

0.03 (0.02 to 0.05)

0.04 (0.02 to 0.05)

0.12 (0.07 to 0.19)

0.13 (0.07 to 0.23)

0.15 (0.10 to 0.22)

0.03 (0.02 to 0.04)

0.03 (0.01 to 0.06)

0.04 (0.01 to 0.13)

0.03 (0.02 to 0.04)

0.10 (0.07 to 0.15)

0.14 (0.12 to 0.17)

0.18 (0.14 to 0.22)

0.07 (0.04 to 0.10)

0.03 (0.02 to 0.03)

0.04 (0.03 to 0.06)

0.04 (0.03 to 0.06)

0.08 (0.05 to 0.13)

0.41 (0.26 to 0.57)

0.72 (0.61 to 0.80)

0.19 (0.10 to 0.33)

0.33 (0.24 to 0.44)

0.18 (0.10 to 0.28)

0.45 (0.39 to 0.52)

0.10 (0.07 to 0.14);

(0.00 to 0.35)0 0.803

Study Rate(95% CI)

Rate(95% CI)

1

1

2

2

3

3

3

3

3

3

3

4

4

4

5

5

5

5

2

3

3

3

3

3

4

5

5

Round

33/215

32/161

3/139

9/129

3/533

1/82

13/470

23/635

12/103

10/75

24/158

20/757

5/188

2/52

30/1055

23/225

125/874

71/396

82/2971

30/770

33/757

16/192

15/37

58/81

8/42

26/78

88/194

Events/No in group

Fig 2 | Prevalence of severe acute respiratory syndrome coronavirus 2 in pregnant and recently pregnant women identified by various sampling strategies. meta-analysis includes one study (liao 2020) screened using national Health commission china criteria with no events. symptom based screening includes screening based on symptoms or history of contact with individuals with covid-19. round number represents search strategy updates in the living systematic review

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 6: Clinical manifestations, risk factors, and maternal and

RESEARCH

6 doi: 10.1136/bmj.m3320 | BMJ 2020;370:m3320 | the bmj

clinical manifestations of covid-19 during pregnancy and after deliveryThe most common symptoms reported by pregnant and recently pregnant women with suspected or confirmed covid-19 were fever (40%) and cough (39%); lymphopaenia (35%) and raised C reactive protein levels (49%) were the most common laboratory findings (fig 3). Compared with non-pregnant women of reproductive age with covid-19, pregnant and recently pregnant women with the disease were less likely to manifest symptoms of fever (0.43, 0.22 to 0.85; 5 studies, 80 521 women) and myalgia (0.48, 0.45 to 0.51; 3 studies, 80 409 women) (fig 4). A history of pre-existing diabetes was more often observed in pregnant women with covid-19 than in non-pregnant women with the disease (1.78, 1.03 to 3.05; 3 studies, 91 595 women) (see appendix 6b). Sensitivity analysis restricted to various sampling frames showed lower estimates of fever, cough, and dyspnoea in the universal screening population and higher estimates in the symptom based population (see appendix 7). The rates of clinical manifestations were similar to the overall estimates when the analysis was restricted to only women with RT-PCR confirmed covid-19, unselected populations, and women with any risk (see appendix 7).

Outcomes related to covid-19 in pregnant and recently pregnant womenOverall, 73 pregnant women (26 studies, 11 580 women) with confirmed covid-19 died from any cause (0.1%, 95% confidence interval 0.0% to 0.7%). Severe covid-19 was diagnosed in 13% (6% to 21%; 21 studies, 2271 women) of pregnant and recently pregnant women with suspected or confirmed covid-19; 4% (2% to 7%; 17 studies, 10 901 women) of the pregnant women with covid-19 were admitted to an intensive care unit, 3% (1% to 5%; 13 studies, 10 713 women) required invasive ventilation, and 0.4% (0.1% to 0.9%; 9 studies, 1935 women) required extracorporeal membrane oxygenation (fig 3). Appendix 8 provides the rates of complications by sampling strategy. Compared with non-pregnant women of reproductive age with covid-19, the odds of admission to the intensive care unit (1.62, 95% confidence interval 1.33 to 1.96) and need for invasive ventilation (1.88, 1.36 to 2.60) were higher in pregnant and recently pregnant women (four studies, 91 606 women) (table 1). Maternal risk factors associated with severe covid-19 were increasing age (1.78, 1.25 to 2.55; 4 studies, 1058 women), high body mass index (2.38, 1.67 to 3.39; 3 studies, 877 women), chronic hypertension (2.0, 1.14 to 3.48; 2 studies, 858 women), and pre-existing diabetes (2.51, 1.31 to 4.80; 2 studies, 858 women) (fig 5). Pre-existing maternal comorbidity was associated with admission to an intensive care unit (4.21, 1.06 to 16.72; 2 studies, 320 women) and the need for invasive ventilation (4.48, 1.40 to 14.37; 2 studies, 313 women) (table 2).

maternal and perinatal outcomes in pregnant and recently pregnant women with covid-19In pregnant and recently pregnant women with covid-19 the rate of overall preterm birth was 17% (95% confidence interval 13% to 21%; 30 studies, 1872 women) and of spontaneous preterm birth was 6% (3% to 9%; 10 studies, 870 women) (fig 3). In pregnant and recently pregnant women with covid-19 compared with pregnant and recently pregnant women without the disease, the odds of any preterm birth (3.0, 95% confidence interval 1.15 to 7.85; 2 studies, 339 women) were higher, but no differences were observed in other maternal outcomes (table 1). Eighteen stillbirths (27 studies; 2837 offspring) and six neonatal deaths (26 studies; 1728 neonates) occurred among pregnant and recently pregnant women with covid-19, resulting in negligible risks (fig 3). Overall, 25% (95% confidence interval 14% to 37%; 17 studies, 1348 women) of neonates born to women with covid-19 were admitted to the neonatal unit (fig 3), with a higher risk of admission (odds ratio 3.13, 95% confidence interval 2.05 to 4.78; 1 study, 1121 neonates) than those born to mothers without the disease in one study with historical controls. No differences were observed for other perinatal outcomes. Appendix 9 provides the rates of covid-19 related and pregnancy related outcomes for the individual studies.

discussionIn this living systematic review, we found that one in 10 pregnant or recently pregnant women who are attending or admitted to hospital for any reason are diagnosed as having suspected or confirmed covid-19, although the rates vary by sampling strategy. The covid-19 related symptoms of fever and myalgia manifest less often in pregnant and recently pregnant women than in non-pregnant women of reproductive age. Whereas testing for SARS-CoV-2 in non-pregnant women is based on symptoms or contact history, testing in pregnant women is usually done when they are in hospital for reasons that might not be related to covid-19. Pregnant or recently pregnant women with covid-19 seem to be at increased risk of requiring admission to an intensive care unit or invasive ventilation. Increased maternal age, high body mass index, and pre-existing comorbidities might be associated with severe disease. Pregnant women with covid-19 are at increased risk of delivering preterm and their babies being admitted to the neonatal unit. But overall rates of spontaneous preterm births are not high. Stillbirth and neonatal death rates are low in women with suspected or confirmed covid-19. All comparative findings are based on small numbers of studies, despite the large sample sizes. Substantial heterogeneity was observed in the estimates for rates of clinical manifestations and outcomes, which varied by sampling frames, participant selection, and risk status of the participants.

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 7: Clinical manifestations, risk factors, and maternal and

RESEARCH

the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 7

strengths and limitations of this reviewIn this unprecedented pandemic situation, where evidence is rapidly produced and published in various formats, our living systematic review underpinned by robust methods and continually updated at regular intervals is relevant for several reasons. Firstly, it

addresses important research questions relevant to clinical decision making and policies. Secondly, uncertainties remain for key outcomes that require further evidence. Thirdly, the rapid turnover of evidence in various formats requires assessments of study quality and regular updating of the findings.

Clinical manifestations

Symptoms

Fever

Cough

Dyspnoea

Myalgia

Ageusia

Diarrhoea

Laboratory findings

Raised white cell count

Lymphopaenia

Thrombocytopaenia

Abnormal liver function test results

Raised procalcitonin level

Raised C reactive protein level

Radiological findings

Ground glass appearance

Any abnormality on computed tomography

Maternal and perinatal outcomes

Covid related outcomes

All cause mortality

Admission to intensive care unit

Severe covid-19

Invasive ventilation

ECMO

Oxygen, cannula

ARDS

Pneumonia

Cardiac, liver, renal failure

Pregnancy related outcomes

Preterm birth <37 weeks

Spontaneous preterm birth

PPROM <37 weeks

Caesarean section

Vaginal delivery

Postpartum haemorrhage

Offspring outcomes

Stillbirth

Neonatal death

Admission to neonatal unit

Neonatal sepsis

Abnormal Apgar score

Fetal distress

(0.11-0.73)

(0.03-0.81)

(0.00-0.62)

(0.00-0.25)

(0.03-0.28)

(0.00-0.18)

(0.03-0.52)

(0.09-0.90)

(0.01-0.35)

(0.00-0.29)

(0.00-0.97)

(0.23-0.71)

(0.09-1.00)

(0.02-1.00)

(0.00-0.07)

(0.00-0.13)

(0.00-1.00)

(0.00-0.09)

(0.00-0.01)

(0.02-1.00)

(0.00-0.51)

(0.00-1.00)

(0.00-0.13)

(0.00-0.59)

(0.02-0.31)

(0.03-0.17)

(0.33-1.00)

(0.00-0.67)

(0.01-0.09)

(0.00-0.02)

(0.00-0.01)

(0.00-1.00)

(0.03-0.06)

(0.00-0.06)

(0.04-0.15)

Study Proportion(95% CI)

Range

97.4 (0.00)

96.8 (0.00)

96.2 (0.00)

90.7 (0.00)

93.6 (0.00)

65.5 (0.00)

92.3 (0.00)

85.6 (0.00)

85.3 (0.00)

74.1 (0.00)

96.6 (0.00)

86.2 (0.00)

96.5 (0.00)

98.4 (0.00)

80.2 (0.00)

93.6 (0.00)

95.5 (0.00)

93.5 (0.00)

0.0 (0.93)

97.1 (0.00)

98.7 (0.00)

97.9 (0.00)

10.6 (0.35)

71.5 (0.00)

55.0 (0.02)

0.0 (0.66)

91.3 (0.00)

91.4 (0.00)

45.6 (0.14)

0.0 (1.00)

0.0 (1.00)

94.9 (0.00)

Not estimable

0.0 (0.64)

0.0 (0.74)

I2 (%)(P value)

0.40 (0.31 to 0.49)

0.39 (0.31 to 0.47)

0.19 (0.13 to 0.26)

0.10 (0.05 to 0.17)

0.15 (0.00 to 0.41)

0.07 (0.05 to 0.09)

0.27 (0.09 to 0.51)

0.35 (0.26 to 0.45)

0.08 (0.02 to 0.18)

0.11 (0.05 to 0.18)

0.21 (0.00 to 0.59)

0.49 (0.36 to 0.63)

0.69 (0.41 to 0.91)

0.65 (0.46 to 0.82)

0.00 (0.00 to 0.01)

0.04 (0.02 to 0.07)

0.13 (0.06 to 0.21)

0.03 (0.01 to 0.05)

0.00 (0.00 to 0.01)

0.30 (0.14 to 0.48)

0.09 (0.00 to 0.33)

0.49 (0.35 to 0.63)

0.00 (0.00 to 0.01)

0.17 (0.13 to 0.21)

0.06 (0.03 to 0.09)

0.05 (0.03 to 0.08)

0.65 (0.57 to 0.73)

0.35 (0.27 to 0.43)

0.03 (0.00 to 0.08)

0.00 (0.00 to 0.00)

0.00 (0.00 to 0.00)

0.25 (0.14 to 0.37)

0.04 (0.00 to 0.12)

0.01 (0.00 to 0.02)

0.08 (0.05 to 0.12)

Proportion(95% CI)

29

28

22

9

3

17

6

15

7

9

5

7

10

20

26

17

21

13

9

13

6

23

7

30

10

8

28

27

5

27

26

17

2

14

7

Studies

2733/8328

3432/8317

1928/8159

1411/6078

24/310

659/7525

50/251

262/780

36/428

51/491

60/261

174/426

246/387

599/1968

73/11 580

323/10 901

417/2271

155/10 713

16/1935

243/1281

270/1006

729/2577

7/737

386/1872

56/870

28/436

1060/1933

856/1916

13/250

18/2837

6/1728

368/1348

2/51

11/500

25/293

Events/No in group

0 0.2 0.4 0.6 0.8 1.0

Fig 3 | rates of clinical manifestations of coronavirus disease (covid-19) in pregnant women and recently pregnant women with suspected or confirmed covid-19 and associated maternal and perinatal outcomes. ecmO=extracorporeal membrane oxygenation; arDs=acute respiratory distress syndrome; PPrOm=preterm premature rupture of membranes

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 8: Clinical manifestations, risk factors, and maternal and

RESEARCH

8 doi: 10.1136/bmj.m3320 | BMJ 2020;370:m3320 | the bmj

Finally, our living systematic review will produce a strong evidence base for living guidelines on covid-19 and pregnancy.

We undertook a comprehensive search and coordi-nated our efforts with key organisations and research groups, such as WHO, the Cochrane Centre, and EPPI-Centre. To minimise risk of bias we restricted our meta-analysis to cohort studies, and we reported the quality of the included studies. By contacting the authors and obtaining reports not published in PubMed, we minimised the risk of missing relevant studies. Our systematic review has a large sample size and it is continuously increasing. Our living meta-analyses framework will enable us to rapidly update the findings as new data emerge. We undertook extensive work to ensure that duplicate data are not included. Our various

comparative analyses allowed us to comprehensively assess the association between pregnancy and covid-19 related outcomes, covid-19 and pregnancy outcomes, risk factors for SARS-CoV-2 infection, and complications. Our review helps to understand the variations in estimates through sensitivity analyses by sampling strategies, population characteristics, and risk factors, and it provides confidence in the rates of reported outcomes.

Our systematic review also has limitations. The primary studies used varied sampling frames to identify women with covid-19, comprised women with suspected and confirmed covid-19, and primarily reported on pregnant women who required visits to hospital, including for childbirth, thereby affecting the generalisability of the estimates. Although our

Any symptom

Cheng 2020

Wei 2020

Wang 2020

Ellington 2020

Subtotal: I2=80.3%

Fever

Liu 2020

Yin 2020

Cheng 2020

Wang 2020

Ellington 2020

Subtotal: I2=73.9%

Cough

Liu 2020

Yin 2020

Cheng 2020

Wang 2020

Ellington 2020

Subtotal: I2=67.6%

Dyspnoea

Liu 2020

Yin 2020

Cheng 2020

Wang 2020

Ellington 2020

Subtotal: I2=36.0%

Myalgia

Yin 2020

Cheng 2020

Ellington 2020

Subtotal: I2=73.9%

0.16 (0.05 to 0.54)

0.62 (0.08 to 4.92)

0.03 (0.00 to 0.56)

1.07 (0.91 to 1.26)

0.33 (0.08 to 1.41)

0.22 (0.06 to 0.85)

0.20 (0.06 to 0.66)

0.59 (0.26 to 1.37)

0.29 (0.11 to 0.77)

0.87 (0.82 to 0.93)

0.43 (0.22 to 0.85)

0.55 (0.15 to 2.05)

1.11 (0.42 to 2.93)

0.55 (0.24 to 1.27)

0.20 (0.06 to 0.62)

1.10 (1.04 to 1.17)

0.67 (0.37 to 1.23)

0.90 (0.05 to 15.47)

1.00 (0.33 to 3.03)

0.32 (0.11 to 0.92)

0.39 (0.04 to 3.72)

1.12 (1.05 to 1.20)

0.82 (0.47 to 1.43)

0.52 (0.12 to 2.27)

0.30 (0.04 to 2.50)

0.48 (0.45 to 0.51)

0.48 (0.45 to 0.51)

Symptoms Odds ratio(95% CI)

22/31

15/17

22/30

5199/5355

5258/5433

8/21

17/31

15/31

11/30

1190/5355

1241/5468

6/21

15/31

14/31

5/30

1799/5355

1839/5468

1/21

8/31

5/31

1/27

1045/5355

1060/5465

3/31

1/31

1323/8207

1327/8269

No of pregnantwomen with covid-19/

No in group

75/80

24/26

42/42

72 549/74 877

72 690/75 025

14/19

30/35

49/80

28/42

18 474/74 877

18 595/75 053

8/19

16/35

48/80

21/42

23 554/74 877

23 647/75 053

1/19

9/35

30/80

4/45

13 292/74 877

13 336/75 056

6/35

8/80

20 726/72 025

20 740/72 140

No of non-pregnantwomen with covid-19/

No in group

0.01

Note: Weights are from random effects analysis

0.25 0.5 2 101 100

Odds ratio(95% CI)

Fig 4 | clinical manifestations of coronavirus disease (covid-19) in pregnant and recently pregnant women compared with non-pregnant women of reproductive age with covid-19

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 9: Clinical manifestations, risk factors, and maternal and

RESEARCH

the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 9

sensitivity analyses aimed to tackle some of these problems, the numbers and sample sizes of the individual studies were too small to identify differences between the subgroups. The timing of assessment of the clinical manifestations of disease was generally not available. The definitions of symptoms, tests, and outcomes were heterogeneous. Furthermore, poor reporting of the criteria for caesarean section, admissions to the neonatal unit, and the causes of preterm birth, made it difficult to disentangle iatrogenic effect from the true impact of the disease. There is a paucity of comparative data to assess the risk of severe disease in pregnant women compared with non-pregnant women in similarly aged groups, and to compare pregnancy outcomes in women with and without covid-19. Not many studies reported outcomes by trimester for symptom onset, making it difficult to assess the rates of miscarriage and postpartum complications. For some outcomes, the findings were influenced by a single large study.26 Many studies had to be excluded as we could not rule out potential overlap in the study populations.

comparison with existing evidenceAlongside the spread of the pandemic, a shift has occurred in the types of studies published, with initial studies involving pregnant women from epidemic regions in China, followed by reports of large regional and national datasets from the US, UK, Netherlands, Spain, and, more recently, Latin American countries. The study design has also changed from initial small case series and case reports to large observational data, with recent studies also providing comparative data. The prevalence of covid-19 varied widely between studies, particularly when sampling was done based on symptoms or history of contact, highlighting the variations in criteria for

testing. Moreover, the findings only relate to those women attending hospital for any reason. The true prevalence of covid-19 in pregnancy is likely to be lower when all pregnant women are included.

In the recent cohort study of all individuals admitted with covid-19 in the UK, the cluster of respiratory symptoms of cough, fever, and breathlessness were observed in more than two thirds of individuals,77 similar to reported rates in the US and China.78-80 But in our review, fewer pregnant and recently pregnant women with covid-19 manifested these symptoms than the non-pregnant population, indicating possi-ble high rates of asymptomatic presentation in this population. This is likely because of the strategy of universal screening for covid-19 in pregnancy and the low thresholds for testing than in non-pregnancy. Despite the possibility of the above strategies detecting pregnant women with mild disease, we observed an increase in admissions to the intensive care unit and need for invasive ventilation compared with non-pregnant women of reproductive age with covid-19. The findings were mainly influenced by the recent large Centers for Disease Control and Prevention report from the US.26 Pregnancy status was not ascertained in a large proportion of women of reproductive age in the CDC report that could affect the estimates. Furthermore, the outcomes for which the data were missing were considered to be absent in the report, thereby incurring bias. The pooled estimates for severe covid-19 and admission to an intensive care unit were, however, still relatively high in the non-comparative data, indicative of a potential high risk in pregnancy. This is supported by the recent analysis in a Swedish study suggesting a high risk of admission to an intensive care unit and invasive ventilation in pregnant women than non-pregnant women.81

table 1 | Outcomes in pregnant and recently pregnant women with coronavirus disease 2019 (covid-19)

Outcomes no of studieswomen (no with event/no in group (%))

Odds ratio (95% ci) i2 (%)Pregnant women with covid-19 comparison groupcomparison group: non-pregnant women of reproductive age with covid-19All cause mortality 4 16/8282 (0.2) 208/83 327 (0.2) 0.81 (0.49 to 1.33) 0ICU admission 4 121/8276 (1.5) 758/83 330 (0.9) 1.62 (1.33 to 1.96) 0Invasive ventilation 4 43/8276 (0.5) 226/83 330 (0.3) 1.88 (1.36 to 2.60) 0ECMO 1 0/31 (0) 0/80 (0) 2.56 (0.05 to 131.60) NEOxygen through nasal cannula 2 8/48 (16.7) 49/106 (46.2) 0.21 (0.04 to 1.13) 65.7ARDS 1 0/17 (0) 0/26 (0) 1.51 (0.03 to 79.93) NEMajor organ failure 1 0/17 (0) 0/26 (0) 1.51 (0.03 to 79.93) NEcomparison group: pregnant women without covid-19Maternal outcomes: All cause mortality 1* 5/427 (1.2) 0/694 (0) 18.08 (1.00 to 327.83) NE ICU admission 1* 40/427 (9.4) 1/694 (0.1) 71.63 (9.81 to 523.06) NE Preterm birth <37 weeks 2 7/44 (15.9) 18/295 (6.1) 3.01 (1.16 to 7.85) 0.9 Caesarean section 3* 184/491 (37.5) 577/1676 (34.4) 2.02 (0.67 to 6.10) 87.5Perinatal outcomes: Stillbirth 1* 3/427 (0.7) 2/694 (0.3) 2.45 (0.41 to 14.71) NE Neonatal death 1* 2/427 (0.5) 1/694 (0.1) 3.26 (0.30 to 36.07) NE Admission to neonatal unit 1* 64/427 (15.0) 37/694 (5.3) 3.13 (2.05 to 4.79) NE Abnormal Apgar score at 5 minutes 1 0/30 (0) 12/740 (1.6) 0.96 (0.06 to 16.51) NE Fetal distress 1 3/34 (8.8) 12/242 (5.0) 1.86 (0.50 to 6.94) NEICU=intensive care unit; ECMO=extracorporeal membrane oxygenation; ARDS=acute respiratory distress syndrome; NE=not estimable.The denominator is number of pregnancies for all outcomes.*Historical comparative cohort in UK Obstetric Surveillance System study.

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 10: Clinical manifestations, risk factors, and maternal and

RESEARCH

10 doi: 10.1136/bmj.m3320 | BMJ 2020;370:m3320 | the bmj

Age*

Kayem 2020

Martinez-Perez 2020

Khoury 2020

Chen 2020 (continuous age)

Subtotal: I2=9%

Body mass index

Kayem 2020

Martinez-Perez 2020

Khoury 2020

Wu 2020

Subtotal: I2=0%

Multiparity

Chen 2020

Savasi 2020

Martinez-Perez 2020

Subtotal: I2=0%

Third trimester

Yan 2020

Andrikopoulou 2020

Subtotal: I2=0%

Non-white

Savasi 2020

Khoury 2020

Subtotal: I2=73%

Any comorbidity

Savasi 2020

Martinez-Perez 2020

Subtotal: I2=0%

Chronic hypertension

Kayem 2020

Khoury 2020

Subtotal: I2=0%

Pre-existing diabetes

Kayem 2020

Khoury 2020

Subtotal: I2=12%

Pre-eclampsia

Yan 2020

Martinez-Perez 2020

Subtotal: I2=0%

Gestational diabetes

Andrikopoulou 2020

Kayem 2020

Martinez-Perez 2020

Yan 2020

Subtotal: I2=0%

2.24 (1.50 to 3.35)

1.00 (0.13 to 7.46)

1.19 (0.65 to 2.18)

1.87 (0.55 to 6.42)

1.78 (1.25 to 2.55)

2.39 (1.56 to 3.66)

1.11 (0.11 to 11.35)

2.51 (1.31 to 4.81)

Excluded

2.38 (1.67 to 3.39)

1.39 (0.35 to 5.47)

0.82 (0.25 to 2.66)

1.42 (0.14 to 14.29)

1.07 (0.46 to 2.46)

0.64 (0.07 to 5.76)

0.59 (0.26 to 1.32)

0.59 (0.28 to 1.27)

1.88 (0.57 to 6.17)

0.45 (0.19 to 1.06)

0.86 (0.21 to 3.50)

1.88 (0.57 to 6.17)

0.71 (0.07 to 7.14)

1.53 (0.53 to 4.41)

2.51 (0.95 to 6.62)

1.78 (0.90 to 3.51)

2.00 (1.14 to 3.48)

3.98 (1.37 to 11.57)

1.98 (0.96 to 4.08)

2.51 (1.31 to 4.80)

5.00 (0.46 to 54.51)

8.33 (0.66 to 105.71)

6.35 (1.11 to 36.22)

0.60 (0.07 to 5.13)

1.23 (0.69 to 2.21)

5.74 (0.20 to 161.79)

Excluded

1.23 (0.70 to 2.14)

Risk factors Odds ratio(95% CI)

59/128

2/4

22/75

n/9

83/216

46/128

1/4

43/62

0/0

90/194

5/9

8/14

3/4

16/27

7/8

22/34

29/42

6/14

54/65

60/79

6/14

1/4

7/18

7/128

18/75

25/203

7/128

16/75

23/203

1/8

1/4

2/12

1/34

17/128

0/4

0/0

18/166

No of pregnant womenwith risk factor and severe

covid-19/No in group

135/489

39/78

43/166

n/109

219/842

93/489

18/78

55/116

0/13

166/696

46/97

39/63

53/78

138/238

99/108

94/124

193/232

18/63

143/156

161/219

18/63

25/78

43/141

11/489

25/166

36/655

7/489

20/166

27/655

3/108

3/78

6/186

6/124

54/489

1/78

9/116

70/807

No of pregnant womenwith risk factor without

severe covid-19/No in group

Note: Weights are from random effects analysis

0.01 0.25 0.5 2 101 100

Odds ratio(95% CI)

Fig 5 | risk factors associated with severe coronavirus disease 2019 (covid-19) in pregnant and recently pregnant women. symptom based screening: savasi v, Kayem g; nHcc (national Health commission china). criteria based screening: chen, wu, yan. all other studies used universal screening. cut-off for age is 35 years or more, and for body mass index is 30 or more. *includes one study with continuous measurement of risk factor

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 11: Clinical manifestations, risk factors, and maternal and

RESEARCH

the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 11

Similar to the general population, high body mass index and pre-existing comorbidity seemed to be risk factors for severity of covid-19 in pregnancy, including admission to an intensive care unit and invasive ventilation.77 Complications related to covid-19 did not seem to be increased in women presenting in the third trimester or in multiparous women—but existing sample sizes are not large. Both chronic hypertension and pre-existing diabetes were associated with maternal death in pregnant women with covid-19, which are known risk factors in the general population. But it is not known if covid-19 was the direct cause of death for these women, and the numbers of studies

are small. We observed an increase in rates of preterm birth in pregnant women with covid-19 compared with those without the disease. These preterm births could be medically indicated, as the overall rates of spontaneous preterm births in pregnant women with covid-19 was broadly similar to those observed in the pre-pandemic period. Although more than 60% of pregnant women underwent caesarean section in the non-comparative studies, we did not find a statistically significant difference in comparative studies of pregnant women with and without covid-19. The precision of the estimates is expected to improve with the publication of more data in the future. The overall

table 2 | maternal characteristics associated with severe coronavirus disease 2019 (covid-19) and all cause death in pregnant and recently pregnant women with a diagnosis of covid-19

maternal risk factors and outcomes

no of studies

total no of women

Pregnant women (no with risk factor/no in group (%))

Odds ratio (95% ci) i2 (%)with outcome without outcomeAge ≥35 years: Severe disease 4 1058 216* 842* 1.78 (1.25 to 2.55) 9 ICU admission 2 260 8/87 (9.2) 8/173 (4.6) 2.44 (0.43 to 14.01) 63 Invasive ventilation 1 178 3/65 (4.6) 2/113 (1.8) 2.69 (0.44 to 16.51) NE Maternal death 1 288 20/154 (13.0) 16/134 (11.9) 1.10 (0.55 to 2.22) NEMultiparity: Severe disease 3 265 16/154 (10.4) 11/111 (9.9) 1.07 (0.46 to 2.46) 0 ICU admission 1 42 4/22 (18.2) 4/20 (20.0) 0.89 (0.19 to 4.15) NEBody mass index ≥30: Severe disease 3 877 90/256 (35.2) 104/621 (16.7) 2.38 (1.67 to 3.39) 0 ICU admission 1 142 3/22 (13.6) 4/120 (3.3) 4.58 (0.95 to 22.09) NE Invasive ventilation 1 135 5/21 (23.8) 6/114 (5.3) 5.63 (1.54 to 20.59) NE Maternal death 2 596 6/62 (9.7) 37/534 (6.9) 2.57 (0.97 to 6.82) 0Non-white ethnicity: Severe disease 2 298 60/221 (27.1) 19/77 (24.7) 0.86 (0.21 to 3.50) 73 ICU admission 1 42 5/20 (25.0) 3/22 (13.6) 2.11 (0.43 to 10.28) NE Maternal death 2 596 31/220 (14.1) 12/376 (3.2) 2.40 (0.94 to 6.11) 0Any comorbidity: Severe disease 2 159 7/50 (14.0) 11/109 (10.1) 1.53 (0.53 to 4.41) 0 ICU admission 2 320 4/37 (10.8) 11/283 (3.9) 4.21 (1.06 to 16.72) 0 Invasive ventilation 2 313 6/36 (16.7) 10/277 (3.6) 4.48 (1.40 to 14.37) 0Chronic hypertension: Severe disease 2 858 25/61 (41.0) 178/797 (22.3) 2.0 (1.14 to 3.48) 0 ICU admission 1 141 2/5 (40.0) 5/136 (3.7) 17.47 (2.37 to 129.02) NE Invasive ventilation 1 134 4/5 (80.0) 7/129 (5.4) 69.71 (6.85 to 709.34) NE Maternal death 2 596 5/29 (17.2) 38/567 (6.7) 3.38 (1.17 to 9.75) 0Pre-existing diabetes: Severe disease 2 858 23/50 (46.0) 180/808 (22.3) 2.51 (1.31 to 4.80) 12 ICU admission 2 181 1/7 (14.3) 14/174 (8.0) 2.88 (0.44 to 18.96) 0 Invasive ventilation 1 132 1/6 (16.7) 9/126 (7.1) 2.60 (0.27 to 24.71) NE Maternal death 2 596 10/52 (19.2) 33/544 (6.1) 6.63 (0.27 to 161.45) 91Asthma: Severe disease 3 857 17/61 (27.9) 149/796 (18.7) 1.86 (0.88 to 3.93) 22 Maternal death 2 596 3/22 (13.6) 40/574 (7.0) 2.04 (0.61 to 6.85) 0Smoking: Severe disease 3 776 5/23 (21.7) 141/753 (18.7) 1.67 (0.64 to 4.40) 0 ICU admission 1 42 1/2 (50.0) 7/40 (17.5) 4.71 (0.26 to 84.77) NE Maternal death 1 308 0/10 (0) 7/298 (2.3) 1.85 (0.10 to 34.60) NEGestation ≥28 weeks: Severe disease 2 274 29/222 (13.1) 13/52 (25.0) 0.59 (0.28 to 1.27) 0 Maternal death 1 273 22/190 (11.6) 12/83 (14.5) 0.78 (0.36 to 1.65) NEGestational diabetes: Severe disease 4 973 18/88 (20.5) 148/885 (16.7) 1.23 (0.70 to 2.14) 0Pre-eclampsia: Severe disease 2 198 2/8 (25.0) 10/190 (5.3) 6.36 (1.12 to 36.22) 0 ICU admission 1 42 6/6 (100.0) 2/36 (5.6) 179.40 (7.69 to 4186.05) NEICU=intensive care unit; NE=not estimable.*Includes one or more studies with continuous measurement of risk factor.

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 12: Clinical manifestations, risk factors, and maternal and

RESEARCH

12 doi: 10.1136/bmj.m3320 | BMJ 2020;370:m3320 | the bmj

rates of stillbirths and neonatal deaths do not seem to be higher than the background rates. The indications for admissions to the neonatal unit, observed in about a quarter of neonates delivered to mothers with covid-19, have not been reported. Local policies on observation and quarantine of infants with exposure to SARS-CoV-2 might have influenced these rates.

relevance for clinical practice and researchBased on existing data, healthcare professionals should be aware that pregnant and recently pregnant women with covid-19 might manifest fewer symptoms than the general population, with the overall pattern similar to that of the general population. Emerging comparative data indicate the potential for an increase in the rates of admission to intensive care units and invasive ventilation in pregnant women compared with non-pregnant women. Mothers with pre-existing comorbidities will need to be considered as a high risk group for covid-19, along with those who are obese and of greater maternal age. Clinicians will need to balance the need for regular multidisciplinary antenatal care to manage women with pre-existing comorbidities against unnecessary exposure to the virus, through virtual clinic appointments when possible. Pregnant women with covid-19 before term gestation might need to be managed in a unit with facilities to care for preterm neonates.

Further data are needed to assess robustly if preg-nancy related maternal and neonatal complications are increased in women with covid-19 than those without the disease. Similarly, the association between other risk factors such as ethnicity and pregnancy specific risk factors such as pre-eclampsia and gestational diabetes on both covid-19 related and pregnancy related outcomes needs evaluation. Pre-eclampsia was reported to be associated with severe covid-19 in small studies, but it requires further assessment as the clinical presentation of severe pre-eclampsia could mimic worsening covid-19.82 Robust collection of maternal data by trimester of exposure, including the periconception period, is required to determine the effects of covid-19 on early pregnancy outcomes, fetal growth, and risk of stillbirth.

Systematic reviews are considered to be the highest quality evidence informing guidelines, and poor quality reviews will have a direct impact on clinical care. Despite the urgent need for evidence on the impact of covid-19 in pregnant women, systematic reviews and meta-analyses still need to adhere to the reporting guidelines on search criteria, quality assessment, and analysis. This is particularly impor-tant as large numbers of non-peer reviewed scientific papers and reports are currently available in the public domain in multiple versions. Primary studies need to explicitly state if duplicate data have been included to avoid double counting of participants in evidence synthesis. Individual participant data meta-analysis of the emerging cohorts is critical to assess both differential presentation and outcomes by underlying

risk factors, and to determine the differential effects of interventions to reduce the rates of complications. With the establishment of several national and global prospective cohorts, we expect the sample size of our meta-analysis to increase further in the coming months. Our living systematic review and meta-analysis with its regular search and analyses updates is ideally placed to assess the impact of new findings on the rapidly growing evidence base.

autHOr aFFiliatiOns1Institute of Applied Health Research, University of Birmingham, Birmingham, UK2WHO Collaborating Centre for Global Women’s Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK3Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain4CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain5UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland6Birmingham Medical School, University of Birmingham, Birmingham, UK7Division of Birth Cohort Study, Guangzhou Women and Children’s Medical Centre, Guangzhou Medical University, Guangzhou, China8Department of Woman and Child Health Care, Guangzhou Women and Children’s Medical Centre, Guangzhou Medical University, Guangzhou, China9Department of Obstetrics and Gynaecology, Guangzhou Women and Children’s Medical Centre, Guangzhou Medical University, Guangzhou, China10Netherlands Satellite of the Cochrane Gynaecology and Fertility Group, Amsterdam University Medical Centre, Amsterdam, Netherlands11Department of Obstetrics and Gynaecology, Amsterdam University Medical Centre, Amsterdam, Netherlands12Blizard Institute, Queen Mary University of London, London, UK13Barts Health NHS Trust, London, UK14St George’s, University of London, London, UK15Elizabeth Glaser Paediatric AIDS Foundation, Washington DC, USA16Women’s Health Research Unit, Queen Mary University of London, London, UK17Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK

Other members of the PregCOV-19 Living Systematic Review Consortium are Uma Ram, Ajith S Nair, Pura Rayco-Solon, and Hector Pardo-Hernandez. The Cochrane Gynaecology and Fertility Group thank Marijke Strikwerda and Bethany Clark for help with searches and data extraction. The PregCOV-19 Living Systematic Review Group would also like to thank Katie’s Team for its contribution towards the development and reporting of this work, James Thomas from the EPPI-Centre for helping with search updates, and the PregCOV-19 Living Systematic Review steering committee members, Pisake Lumbiganon, Carolina Carvalho Ribeiro do Valle, Clare Whitehead, David Lissauer, Joao Paulo Souza, and Marian Knight, who provided guidance throughout.Contributors: JA and ES are joint first authors. ST, MB, and JA conceptualised the study. MY, SC, LD, TK, ACL, AD, DZ, RB, SL, XQ, and MYuan selected the studies. JA, ES, MY, LD, DZ, XQ, and MYuan extracted the data. JZ conducted the analyses. All coauthors contributed to the writing of the manuscript and approved the final version. ST, JA, ES, and JZ are the guarantors. The corresponding author attests that all listed authors meet authorship criteria and that no others meeting the criteria have been omittedFunding: The project was partially funded by the World Health Organization.Competing interests: All authors have completed the ICMJE uniform disclosure form at www.icmje.org/coi_disclosure.pdf and declare:

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 13: Clinical manifestations, risk factors, and maternal and

RESEARCH

the bmj | BMJ 2020;370:m3320 | doi: 10.1136/bmj.m3320 13

partial funding by the World Health Organization; no financial relationships with any organisations that might have an interest in the submitted work in the previous three years; no other relationships or activities that could appear to have influenced the submitted work.Ethical approval: Not required.Data sharing: No additional data available.The corresponding author (ST) affirms that the manuscript is an honest, accurate, and transparent account of the study being reported; that no important aspects of the study have been omitted; and that any discrepancies from the study as planned have been disclosed.Dissemination to participants and related patient and public communities: The PregCov-19 LSR Group will disseminate the findings through a dedicated website (www.birmingham.ac.uk/research/who-collaborating-centre/pregcov/index.aspx) and social media.Provenance and peer review: Not commissioned; externally peer reviewed.This is an Open Access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/.

1  Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395:497-506. doi:10.1016/S0140-6736(20)30183-5 

2  World Health Organization (WHO). Coronavirus disease. (COVID-19) Pandemic, https://www.who.int/emergencies/diseases/novel-coronavirus-2019 (accessed 7 May 2020)

3  Cabinet Office. Guidance. Staying alert and safe (social distancing). Coronavirus (COVID-19) Guidance and support. Updated 22 May 2020 https://www.gov.uk/government/publications/staying-alert-and-safe-social-distancing/staying-alert-and-safe-social-distancing (accessed 24 May 2020).

4  RCOG. (COVID-19) Infection in Pregnancy, https://www.rcog.org.uk/en/guidelines-research-services/guidelines/coronavirus-pregnancy/

5  Zaigham M, Andersson O. Maternal and perinatal outcomes with COVID-19: A systematic review of 108 pregnancies. Acta Obstet Gynecol Scand 2020;99:823-9. doi:10.1111/aogs.13867 

6  Parazzini F, Bortolus R, Mauri PA, Favilli A, Gerli S, Ferrazzi E. Delivery in pregnant women infected with SARS-CoV-2: A fast review. Int J Gynaecol Obstet 2020;150:41-6. doi:10.1002/ijgo.13166 

7  Elshafeey F, Magdi R, Hindi N, et al. A systematic scoping review of COVID-19 during pregnancy and childbirth. Int J Gynaecol Obstet 2020;150:47-52. doi:10.1002/ijgo.13182 

8  Di Mascio D, Khalil A, Saccone G, et al. Outcome of coronavirus spectrum infections (SARS, MERS, COVID-19) during pregnancy: a systematic review and meta-analysis. Am J Obstet Gynecol MFM 2020;2:100107.

9  Della Gatta AN, Rizzo R, Pilu G, Simonazzi G. Coronavirus disease 2019 during pregnancy: a systematic review of reported cases. Am J Obstet Gynecol 2020;223:36-41. doi:10.1016/j.ajog.2020.04.013 

10  Cheruiyot I, Henry BM, Lippi G. Is there evidence of intra-uterine vertical transmission potential of COVID-19 infection in samples tested by quantitative RT-PCR?Eur J Obstet Gynecol Reprod Biol 2020;249:100-1. doi:10.1016/j.ejogrb.2020.04.034 

11  Gajbhiye R, Modi D, Mahale S. Pregnancy outcomes, Newborn complications and Maternal-Fetal Transmission of SARS-CoV-2 in women with COVID-19: A systematic review of 441 cases. medRxiv [Preprint]. 2020. doi.org/10.1101/2020.04.11.20062356

12  Murad MH, Sultan S, Haffar S, Bazerbachi F. Methodological quality and synthesis of case series and case reports. BMJ Evid Based Med 2018;23:60-3. doi:10.1136/bmjebm-2017-110853 

13  Breslin N, Baptiste C, Gyamfi-Bannerman C, et al. Coronavirus disease 2019 infection among asymptomatic and symptomatic pregnant women: Two weeks of confirmed presentations to an affiliated pair of New York City hospitals. Am J Obstet Gynecol MFM 2020;2:100118.

14  Vintzileos WS, Muscat J, Hoffmann E, et al. Screening all pregnant women admitted to labor and delivery for the virus responsible for coronavirus disease 2019. Am J Obstet Gynecol 2020;223:284-6.

15  Xu L, Yang Q, Shi H, et al. Clinical presentations and outcomes of SARS-CoV-2 infected pneumonia in pregnant women and health status of their neonates. Sci Bull (Beijing) 2020;65:1537-42.

16  Blitz MJ, Grunebaum A, Tekbali A, et al. Intensive care unit admissions for pregnant and nonpregnant women with coronavirus disease 2019. Am J Obstet Gynecol 2020;223:290-1.

17  Released by National Health Commission & National Administration of Traditional Chinese Medicine on 3 March 2020. Trial Version 7. Chin Med J (Engl) 2020;133:1087-95.

18  Allotey J, Bonet M, Zamora J, et al. COVID-19 in pregnant women: a Living Systematic Review on prevalence, presentation, prognosis and treatment. PROSPERO 2020 CRD42020178076. https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42020178076.

19  Living Overview of the Evidence. (LOVE) Platform. app.iloveevidence.com. (accessed 8 July 2020)

20  World Health Organization. Global surveillance for COVID-19 caused by human infection with COVID-19 virus: interim guidance, 20 March 2020. World Health Organization. https://apps.who.int/iris/handle/10665/331506. 2020‎.

21  Dekkers OM, Egger M, Altman DG, Vandenbroucke JP. Distinguishing case series from cohort studies. Ann Intern Med 2012;156:37-40. doi:10.7326/0003-4819-156-1-201201030-00006 

22  Wells G. Proceedings or the Third Symposium on Systematic Reviews beyond the Basics. SBOD. Improving Quality and Impact; The Newcastle–Ottawa Scale (NOS) for Assessing the Quality of non-randomised Studies in Meta-analysis. July 3-5 2000, Oxford.

23  Hoy D, Brooks P, Woolf A, et al. Assessing risk of bias in prevalence studies: modification of an existing tool and evidence of interrater agreement. J Clin Epidemiol 2012;65:934-9. doi:10.1016/j.jclinepi.2011.11.014 

24  Chinn S. A simple method for converting an odds ratio to effect size for use in meta-analysis. Stat Med 2000;19:3127-31. doi:10.1002/1097-0258(20001130)19:22<3127::AID-SIM784>3.0.CO;2-M 

25  Cheng B, Jiang T, Zhang L, et al. Clinical Characteristics of Pregnant Women with Coronavirus Disease 2019 in Wuhan, China. SSRN 2020; https://ssrn.com/abstract=3555240

26  Ellington S, Strid P, Tong VT, et al. Characteristics of Women of Reproductive Age with Laboratory-Confirmed SARS-CoV-2 Infection by Pregnancy Status - United States, January 22-June 7, 2020. MMWR Morb Mortal Wkly Rep 2020;69:769-75. doi:10.15585/mmwr.mm6925a1 

27  Liu F, Liu H, Li J, Hou L, Lan W, Wang D. Clinico-Radiological Features and Outcomes in Pregnant Women with COVID-19: Compared with Age-Matched Non-Pregnant Women. SSRN 2020. https://ssrn.com/abstract=3556647

28  Mohr-Sasson A, Chayo J, Bart Y, et al. Laboratory characteristics of pregnant compared to non-pregnant women infected with SARS-CoV-2. Arch Gynecol Obstet 2020; published online 22 June. doi:10.1007/s00404-020-05655-7 

29  Qiancheng X, Jian S, Lingling P, et al, sixth batch of Anhui medical team aiding Wuhan for COVID-19. Coronavirus disease 2019 in pregnancy. Int J Infect Dis 2020;95:376-83. doi:10.1016/j.ijid.2020.04.065 

30  Wang Z, Wang Z, Xiong G. Clinical characteristics and laboratory results of pregnant women with COVID-19 in Wuhan, China. Int J Gynaecol Obstet 2020. doi:10.1002/ijgo.13265 

31  Wei L, Gao X, Chen S, et al. Clinical Characteristics and Outcomes between Pregnant and Non-Pregnant Women with Coronavirus Disease 2019: A Retrospective Cohort Study. SSRN2020. https://ssrn.com/abstract=3569858

32  Yin M, Zhang L, Deng G, et al. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection During Pregnancy In China: A Retrospective Cohort Study. medRxiv [Preprint]. 2020. doi.org/10.1101/2020.04.07.20053744.

33  Campbell KH, Tornatore JM, Lawrence KE, et al. Prevalence of SARS-CoV-2 Among Patients Admitted for Childbirth in Southern Connecticut. JAMA 2020. doi:10.1001/jama.2020.8904 

34  Knight M, Bunch K, Vousden N, et al. Characteristics and outcomes of pregnant women hospitalised with confirmed SARS-CoV-2 infection in the UK: a national cohort study using the UK Obstetric Surveillance System (UKOSS). 2020.doi.org/10.1101/2020.05.08.20089268.

35  Li N, Han L, Peng M, et al. Maternal and neonatal outcomes of pregnant women with COVID-19 pneumonia: a case-control study. Clin Infect Dis 2020;ciaa352. doi:10.1093/cid/ciaa352 

36  Liao J, He X, Gong Q, Yang L, Zhou C, Li J. Analysis of vaginal delivery outcomes among pregnant women in Wuhan, China during the COVID-19 pandemic. Int J Gynaecol Obstet 2020;150:53-7. doi:10.1002/ijgo.13188 

37  Khalil A, Hill R, Ladhani S, Pattisson K, O’Brien P. Severe acute respiratory syndrome coronavirus 2 in pregnancy: symptomatic pregnant women are only the tip of the iceberg. Am J Obstet Gynecol 2020;223:296-7. doi:10.1016/j.ajog.2020.05.005

38  Khan S, Jun L, Nawsherwan, et al. Association of COVID-19 with pregnancy outcomes in health-care workers and general women. Clin Microbiol Infect 2020;26:788-90. doi:10.1016/j.cmi.2020.03.034 

39  Nie R, Wang S-s, Yang Q, et al. Clinical features and the maternal and neonatal outcomes of pregnant women with coronavirus disease 2019. medRxiv [Preprint]. 2020. doi.org/10.1101/2020.03.22.20041061.

40  Yan J, Guo J, Fan C, et al. Coronavirus disease 2019 in pregnant women: a report based on 116 cases. Am J Obstet Gynecol 2020;223:P111.E1-111.E14. doi:10.1016/j.ajog.2020.04.014

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 14: Clinical manifestations, risk factors, and maternal and

RESEARCH

No commercial reuse: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe

41  Zeng L, Xia S, Yuan W, et al. Neonatal Early-Onset Infection With SARS-CoV-2 in 33 Neonates Born to Mothers With COVID-19 in Wuhan, China. JAMA Pediatr 2020. doi:10.1001/jamapediatrics.2020.0878 

42  Liu F, Lan W-s, Gan Q. Clinical and CT features of coronavirus disease in pregnant women. Radiol Practice 2020;35:417-20.

43  Lokken EM, Walker CL, Delaney S, et al. Clinical Characteristics of 46 Pregnant Women with a SARS-CoV-2 Infection in Washington State. Am J Obstet Gynecol 2020. doi:10.1016/j.ajog.2020.05.031

44  Dória M, Peixinho C, Laranjo M, Mesquita Varejão A, Silva PT. Covid-19 during pregnancy: A case series from an universally tested population from the north of Portugal. Eur J Obstet Gynecol Reprod Biol 2020;250:261-2. doi:10.1016/j.ejogrb.2020.05.029 

45  Yang H, Hu B, Zhan S, Yang LY, Xiong G. Effects of SARS-CoV-2 infection on pregnant women and their infants: A retrospective study in Wuhan, China. Arch Pathol Lab Med 2020; published online 18 May.

46  Pereira A, Cruz-Melguizo S, Adrien M, Fuentes L, Marin E, Perez-Medina T. Clinical course of coronavirus disease-2019 in pregnancy. Acta Obstet Gynecol Scand 2020;99:839-47. doi:10.1111/aogs.13921 

47  Fox NS, Melka S. COVID-19 in Pregnant Women: Case Series from One Large New York City Obstetrical Practice. Am J Perinatol 2020;37:1002-4. doi:10.1055/s-0040-1712529 

48  Savasi VM, Parisi F, Patanè L, et al. Clinical Findings and Disease Severity in Hospitalized Pregnant Women With Coronavirus Disease 2019 (COVID-19). Obstet Gynecol 2020;136:252-8. doi:10.1097/AOG.0000000000003979 

49  Zeng Y, Lin L, Yan Q, et al. Update on clinical outcomes of women with COVID-19 during pregnancy. Int J Gynaecol Obstet 2020;150:264-6. doi:10.1002/ijgo.13236 

50  Liu P, Zheng J, Yang P, et al. The immunologic status of newborns born to SARS-CoV-2-infected mothers in Wuhan, China. J Allergy Clin Immunol 2020;146:101-9.

51  Ceulemans D, Thijs I, Schreurs A, et al. Screening for COVID-19 at childbirth: is it effective?Ultrasound Obstetr Gynecol 2020;56:113-4.

52  Duffy CR, Hart JM, Modest AM, et al. Lymphopenia and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection Among Hospitalized Obstetric Patients. Obstet Gynecol 2020;136:229-31. doi:10.1097/AOG.0000000000003984 

53  Andrikopoulou M, Madden N, Wen T, et al. Symptoms and Critical Illness Among Obstetric Patients With Coronavirus Disease 2019 (COVID-19) Infection. Obstet Gynecol 2020;136:291-9. doi:10.1097/AOG.0000000000003996 

54  Servei Català de la Salut. Àrea de Sistemes d’Informació. Informació visualitzada a les 06: 11h del divendres 29 de maig de 2020.

55  Kayem G, Lecarpentier E, Deruelle P, et al. A snapshot of the Covid-19 pandemic among pregnant women in France. J Gynecol Obstet Hum Reprod 2020;49:101826. doi:10.1016/j.jogoh.2020.101826 

56  Knight M, Bunch K, Vousden N, et al, UK Obstetric Surveillance System SARS-CoV-2 Infection in Pregnancy Collaborative Group. Characteristics and outcomes of pregnant women admitted to hospital with confirmed SARS-CoV-2 infection in UK: national population based cohort study. BMJ 2020;369:m2107. doi:10.1136/bmj.m2107 

57  Martínez-Perez O, Vouga M, Cruz Melguizo S, et al. Association Between Mode of Delivery Among Pregnant Women With COVID-19 and Maternal and Neonatal Outcomes in Spain. JAMA 2020. doi:10.1001/jama.2020.10125 

58  Profile of pregnant and children and adolescents with COVID-19. Brazilian Ministry of Health Covid-19 Bulletin. No 17. Epidemic week 21. 17-23 May. https://www.saude.gov.br/images/pdf/2020/May/29/2020-05-25---BEE17---Boletim-do-COE.pdf

59  Yuan L, Zhang J, Dong X. Clinical characteristics and prognosis analysis of COVID-19 infection in 28 pregnancy women. Xinjiang Yike Daxue Xuebao 2020;43.

60  Griffin I, Benarba F, Peters C, et al. The Impact of COVID-19 Infection on Labor and Delivery, Newborn Nursery, and Neonatal Intensive Care Unit: Prospective Observational Data from a Single Hospital System. Am J Perinatol 2020;37:1022-30. doi:10.1055/s-0040-1713416 

61  Sentilhes L, De Marcillac F, Jouffrieau C, et al. COVID-19 in pregnancy was associated with maternal morbidity and preterm birth. Am J Obstet Gynecol 2020.

62  Cosma S, Carosso A, Cusato J, et al. COVID-19 and first trimester spontaneous abortion: a case-control study of 225 pregnant patients. medRxiv [Preprint]. 2020. doi.org/10.1101/2020.06.19.20135749

63  Crovetto F, Crispi F, Llurba E, Figueras F, Gomez-Roig MD, Gratacos E. Seroprevalence and clinical spectrum of SARS-Cov-2 infection in the first versus third trimester of pregnancy. medRxiv [Preprint]. 2020 doi.org/10.1101/2020.06.17.20134098

64  Khoury R, Bernstein PS, Debolt C, et al. Characteristics and Outcomes of 241 Births to Women With Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection at Five New York City Medical Centers. Obstet Gynecol 2020;136:273-82. doi:10.1097/AOG.0000000000004025 

65  Maraschini A, Corsi E, Salvatore MA, Donati S. Coronavirus and birth in Italy: results of a national population-based cohort study. medRxiv [Preprint]. 2020. doi.org/10.1101/2020.06.11.20128652

66  NVOG: Nederlandse Vereniging voor Obstetrie en Gynaecologie. Update registratie COVID-19 positieve zwangeren in NethOSS Geplaatst op 19 June 2020.

67  Pierce-Williams RAM, Burd J, Felder L, et al. Clinical course of severe and critical COVID-19 in hospitalized pregnancies: a US cohort study. Am J Obstet Gynecol MFM 2020:100134.

68  Dong Y, Mo X, Hu Y, Tong S. Epidemiological and Transmission Patterns of Pregnant Women with 2019 Coronavirus Disease in China. SSRN2020. doi:10.2139/ssrn.3551330

69  London V, McLaren RJr, Atallah F, et al. The Relationship between Status at Presentation and Outcomes among Pregnant Women with COVID-19. Am J Perinatol 2020;37:991-4. doi:10.1055/s-0040-1712164 

70  Qadri F, Mariona F.Pregnancy affected by SARS-CoV-2 infection: a flash report from Michigan. J Matern Fetal Neonatal Med 2020;20:1-3.

71  Lumbreras-Marquez MI, Campos-Zamora M, Lizaola-Diaz de Leon H, Farber MK. Maternal mortality from COVID-19 in Mexico. Int J Gynaecol Obstet 2020;150:266-7. doi:10.1002/ijgo.13250 

72  Romagano MP, Guerrero K, Spillane N, et al. Perinatal outcomes in critically ill pregnant women with coronavirus disease 2019. Am J Obstetr Gynecol MFM 2020.

73  Giannini A, Mantovani A, Vezzoli C, Franchini D, Finazzi P. Lung ultrasound for pregnant women admitted to ICU for Covid-19 pneumonia. Minerva Anestesiol 2020.

74  Blitz MJ, Rochelson B, Minkoff H, et al. Maternal mortality among women with coronavirus disease 2019 admitted to the intensive care unit. Am J Obstet Gynecol 2020;S0002-9378(20)30636-0.

75  Perlman J, Oxford C, Chang C, Salvatore C, Di Pace J. Delivery Room Preparedness and Early Neonatal Outcomes During COVID-19 Pandemic in New York City. Pediatrics 2020;146:e20201567. doi:10.1542/peds.2020-1567 

76  Cohen J, Vignaux O, Jacquemard F. Covid-19 in pregnant women: General data from a French National Survey. Eur J Obstet Gynecol Reprod Biol 2020;251:267-8. doi:10.1016/j.ejogrb.2020.06.002 

77  Docherty AB, Harrison EM, Green CA, et al, ISARIC4C investigators. Features of 20 133 UK patients in hospital with covid-19 using the ISARIC WHO Clinical Characterisation Protocol: prospective observational cohort study. BMJ 2020;369:m1985. doi:10.1136/bmj.m1985 

78  Guan WJ, Ni ZY, Hu Y, et al, China Medical Treatment Expert Group for Covid-19. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 2020;382:1708-20. doi:10.1056/NEJMoa2002032 

79  Richardson S, Hirsch JS, Narasimhan M, et al, and the Northwell COVID-19 Research Consortium. Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area. JAMA 2020. doi:10.1001/jama.2020.6775 

80  Garg S, Kim L, Whitaker M, et al. Hospitalization Rates and Characteristics of Patients Hospitalized with Laboratory-Confirmed Coronavirus Disease 2019 - COVID-NET, 14 States, March 1-30, 2020. MMWR Morb Mortal Wkly Rep 2020;69:458-64. doi:10.15585/mmwr.mm6915e3 

81  Collin J, Byström E, Carnahan A, Ahrne M. Public Health Agency of Sweden’s Brief Report: Pregnant and postpartum women with severe acute respiratory syndrome coronavirus 2 infection in intensive care in Sweden. Acta Obstet Gynecol Scand 2020;99:819-22. doi:10.1111/aogs.13901 

82  Mendoza M, Garcia-Ruiz I, Maiz N, et al. Pre-eclampsia-like syndrome induced by severe COVID-19: a prospective observational study. BJOG 2020; published online 1 June.

Supplementary information: appendices 1-9

copyright. on 15 S

eptember 2020 at B

ibliotheque Faculte M

edecine Geneve. P

rotected byhttp://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3320 on 1 S

eptember 2020. D

ownloaded from

Page 15: Clinical manifestations, risk factors, and maternal and

Case Report

Efficient Maternal to Neonate Transfer of NeutralizingAntibodies after SARS-CoV-2 Vaccination with BNT162b2:A Case-Report and Discussion of the Literature

Jonathan Douxfils 1,2,* , Constant Gillot 2 , Émilie De Gottal 3, Stéphanie Vandervinne 4, Jean-Louis Bayart 5,Jean-Michel Dogné 2 and Julien Favresse 2,6

�����������������

Citation: Douxfils, J.; Gillot, C.; De

Gottal, É.; Vandervinne, S.; Bayart,

J.-L.; Dogné, J.-M.; Favresse, J.

Efficient Maternal to Neonate

Transfer of Neutralizing Antibodies

after SARS-CoV-2 Vaccination with

BNT162b2: A Case-Report and

Discussion of the Literature. Vaccines

2021, 9, 907. https://doi.org/

10.3390/vaccines9080907

Academic Editor: Laurent Dacheux

Received: 23 July 2021

Accepted: 13 August 2021

Published: 15 August 2021

Publisher’s Note: MDPI stays neutral

with regard to jurisdictional claims in

published maps and institutional affil-

iations.

Copyright: © 2021 by the authors.

Licensee MDPI, Basel, Switzerland.

This article is an open access article

distributed under the terms and

conditions of the Creative Commons

Attribution (CC BY) license (https://

creativecommons.org/licenses/by/

4.0/).

1 Qualiblood s.a., 5000 Namur, Belgium2 Namur Thrombosis and Hemostasis Center, Namur Research Institute for Life Sciences,

Department of Pharmacy, Faculty of Medicine, University of Namur, 5000 Namur, Belgium;[email protected] (C.G.); [email protected] (J.-M.D.); [email protected] (J.F.)

3 Département de Gynécologie, Centre Hospitalier Régional de Huy, 4500 Liège, Belgium;[email protected]

4 Laboratoire de Biologie Clinique, Centre Hospitalier Régional Huy, 4500 Liège, Belgium; [email protected] Department of Laboratory Medicine, Clinique Saint-Pierre Ottignies, 1340 Ottignies, Belgium;

[email protected] Department of Laboratory Medicine, Clinique Saint-Luc Bouge, 5004 Namur, Belgium* Correspondence: [email protected]; Tel.: +32-81-72-42-91

Abstract: This case reports on the successful maternal to fetal transfer of neutralizing antibodies aftervaccination with BNT162b2 in a pregnant woman at 25 weeks of gestation. The levels of neutralizingantibodies were approximately 5-fold higher in the umbilical cord than in the maternal blood whilethe level of total antibodies showed only a 2-fold increase. This suggest that the antibodies thatcrossed the syncytiotrophoblast cell barrier have specific characteristics that correlate to functionalneutralizing capacity. Although pregnant and lactating women have been excluded from clinicaltrials for several reasons including ethical concerns about fetal exposure, accumulating evidence hasnow revealed that these vaccines are safe and efficient for both the fetus and the woman. Vaccinationagainst COVID-19 in pregnancy is vital to control disease burden and to decrease morbidity inthe ante-, peri- and post-natal periods. Inclusion of pregnant women in research programs for thedevelopment of SARS-CoV-2 vaccines should be mandatory to provide this population with theequitable benefits of vaccine research.

Keywords: neonates; infant; COVID-19; vaccine; antibodies; pregnancy; immunity

1. Introduction

Before the vaccine rollout, multiple cohort studies documented that pregnantwoman were more susceptible to severe COVID-19 compared to age-matched non-pregnant women [1–3]. This is in line with the initial expectations, which stated thataccording to immunologic and cardiopulmonary adaptations occurring during preg-nancy, the risk of severe illness from respiratory infections typically increases [4]. Asystematic review of 60 studies on SARS-CoV-2 in pregnancy reported that severe illnessoccurred in up to 18% of pregnant patients and critical disease complicated up to 5%of the cases, comparable to rates observed in the general population [5]. Nevertheless,despite recommendations from public health advocates for pregnant women includingthe Center for Disease Control and Prevention (CDC), the American College of Obstetri-cians and Gynecologists (ACOG), and the American Academy of Pediatrics, pregnant orbreastfeeding women have been excluded from clinical trials during the development ofexisting COVID-19 vaccines [6–12]. This causes large gaps in understanding the safetyand efficacy of these vaccines in this population, which can be definitively detrimental to

Vaccines 2021, 9, 907. https://doi.org/10.3390/vaccines9080907 https://www.mdpi.com/journal/vaccines

Page 16: Clinical manifestations, risk factors, and maternal and

Vaccines 2021, 9, 907 2 of 8

vaccine acceptance. Although data are emerging documenting the efficacy and the safetyof these vaccines [13] and although guidance from these data recommend vaccination inpregnant and breastfeeding women, [14–16] getting vaccinated during pregnancy mainlyremains a personal choice. Thus, the acceptance of COVID-19 vaccination in pregnancyis an important concern. According to a survey study, the COVID-19 vaccine acceptancerate during pregnancy was 58.4%, a rate which is consistent with the acceptance rate ofother recommended vaccines in pregnancy but that is not sufficient to avoid deleteriouscases of neonate infection due to maternal to fetal transmission or infection during theearly stages of life [17,18].

Both neonates and pregnant women are particularly susceptible to respiratory in-fections, including influenza and respiratory syncytial virus (RSV) [19–21], and recentdata demonstrate that a greater proportion of neonates and infants have severe or criticalillness upon SARS-CoV-2 infection compared to older children [22–24]. Rare cases ofvertical transmission have been reported in neonates [5], but histopathological changesin the placenta have suggested that the inflammatory nature of SARS-CoV-2 infectionduring pregnancy could cause adverse obstetric and neonatal events [25,26]. The risks ofSARS-CoV-2 infection during pregnancy are preterm delivery, preeclampsia, emergencycesarian delivery, and neonatal complications including respiratory distress or pneumonia,disseminated intravascular coagulation, asphyxia, and perinatal deaths [27,28]. In addition,placental injury may increase the risk of long-term adverse neurodevelopmental, as is thecase for other conditions that affect the placenta through the release of proinflammatorycytokines such as interleukin-6, which leads to the activation of T helper cell 17 [29].

Neonates rely on the transfer of maternal immunoglobulin G (IgG) across the placentafor protection against pathogens, and vaccination aims to mimic the presence of pathogensto stimulate the maternal immune response. Data from the literature show that maternalto neonatal transfer of anti-SARS-CoV-2 antibodies is efficient although the maternal tofetal transfer ratio of IgG antibodies may differ depending on gestational age and thetype of immunization, i.e., disease- or vaccine-induced [30–33]. There is also a significantimprovement in the transfer of spike specific IgG into the umbilical cord with the time fromsecond dose, suggesting that time from vaccination may be an important determinant ofthe transfer rates of specific IgG subpopulations following immunization in pregnancy [32].Despite reassuring data, the neutralizing capacity of these transferred antibodies was notassessed [30,32,33].

This case-report assessed the neutralizing capacity of umbilical cord blood comparedto maternal blood in addition to conventional serological assay in a woman who received tatwo-dose regimen of BNT162b2 during the antenatal period. To appreciate the magnitudeof the antibody response of this dyad, a comparison with an age-matched cohort will alsobe done.

2. Case Description and Methods

A 28-year-old pregnant woman received her first dose of BNT162b2 mRNA COVID-19vaccine (Pfizer-BioNTech, Mainz, Germany) at 25 weeks of gestation. The second wasadministered at 30 weeks. No problem was reported during the antenatal period, and nosevere adverse events were reported after the vaccination. The patient only complainedheadache and tiredness the day after the second dose, which resolved with the adminis-tration of paracetamol twice a day. The delivery was planned to be on the 4th of July butoccurred 8 days earlier, on the 26th of June at the Centre Hospitalier Régional (CHR) of Huy(Liège, Belgium), 90 days after the administration of the first vaccine dose. No deliverycomplications were reported, and all of the parameters of the newborn were normal. At4 weeks, the newborn was still doing well. On the day of delivery, umbilical cord bloodwas collected as well as maternal blood to permit comparison of the level of SARS-CoV-2neutralizing antibodies.

To compare the results obtained with the umbilical cord and maternal blood samples,13 non-pregnant women of a similar age (i.e., 25 to 35 years of age) with no history of

Page 17: Clinical manifestations, risk factors, and maternal and

Vaccines 2021, 9, 907 3 of 8

previous SARS-CoV-2 infection (i.e., no documented positive RT-PCR and the absence ofanti-nucleocapsid antibodies), were included. These vaccinated controls were recruitedat the Clinique Saint-Luc Bouge (Namur, Belgium) due to their participation in the CRO-VAX HCP study, a study which has already been described in detail elsewhere [34–36].All of the participants provided informed consent prior to specimen and data collection.The study was approved by a central ethical committee (EudraCT registration number:2020-006149-21).

Blood samples were collected in SST™ II advanced tubes (BD Vacutainer, Plymouth,UK) and were processed according to the manufacturer recommendations to obtain serum.Serum samples were then aliquoted and were stored at −20 ◦C until analysis. Antibodiesagainst the SARS-CoV-2 nucleocapsid (anti-NCP; Elecsys Anti-SARS-CoV-2 NCP total qual-itative ECLIA, Roche Diagnostics, Machelen, Belgium) and the receptor binding domain ofthe S1 subunit of the spike protein (anti-S; Elecsys anti-SARS-CoV-2 spike total quantitativeECLIA, Roche Diagnostics) were measured at each time point. Results above 0.8 U/mL(manufacturer’s cut-off) or 0.165 COI (cut-off index as found previously [37]) for anti-S andanti-NCP antibodies, respectively, were considered positives.

Neutralizing capacity was estimated by performing a surrogate virus neutralizationtest (sVNT) [38]. The iFlash-2019-nCoV NAbs assay (performed on an iFlash1800 analyzerfrom Shenzhen YHLO Biotech Co., Ltd. (Shenzhen, China)) is a one-step competitiveparamagnetic particle chemiluminescent immunoassay (CLIA) for the quantitative deter-mination of 2019-nCoV NAbs in human serum and plasma. The assay detects NAbs thatblock the binding of RBD and ACE2. First, NAbs (if present) react with the RBD antigencoated on paramagnetic microparticles to form a complex. Second, the acridinium-ester-labeled ACE2 conjugate is added to competitively bind to the RBD-coated particles thatwere not neutralized by the NAbs (if present) from the sample, and these form anotherreaction mixture. Under a magnetic field, magnetic particles are adsorbed to the wall of thereaction tube, and unbound materials are washed away by the wash buffer. The resultingchemiluminescent reaction is measured in relative light units (RLUs), with an inverse rela-tionship between the amount of NAbs and the RLU value being detected. A result <10.0arbitrary unit (AU)/mL is considered negative, and a result >10.0 AU/mL is consideredpositive (according to the manufacturer’s information). The neutralizing capacity was alsoconfirmed using a pseudovirus neutralization test, as previously described [38]. Samplesare considered to be neutralizing if the dilution factor provides a 50% diminution of therelative infectivity that is superior to 1:20. The agreement of the sVNT assay with the pVNTwas found to be 97.2% [38].

3. Results and Discussion

The total anti-RBD titers of the maternal and the umbilical cord blood were 1120 U/mLand 2349 U/mL compared to 1747 U/mL (geometric mean; 95%CI, 1231–2438 U/mL) in thecontrol group at the identical time point, i.e., 90 days post-vaccination. The NAbs titers were163 and 906 AU/mL for the maternal and the umbilical cord blood, respectively, comparedto 427 AU/mL (geometric mean; 95%CI, 222–820 AU/mL) for the control group at 90 dayspost-vaccination. The maternal to fetal transfer ratio was 2.10 for total anti-RBD antibodiesand 5.56 for NAbs. A summary of the results is presented in Table 1 and Figure 1. Theneutralizing capacity of the maternal and umbilical cord blood was confirmed by the pVNT,with both samples having higher dilution factors than 1:20 to half of the relative infectivity.

Page 18: Clinical manifestations, risk factors, and maternal and

Vaccines 2021, 9, 907 4 of 8

Table 1. Summary of serological results obtained from the maternal and the umbilical cord blood samples. For pVNT, thehighest the dilution factor, the stronger the neutralizing capacity.

Serum Samples Anti-N Titer (U/mL) Anti-S Titer (U/mL) sVNT (AU/mL) pVNT (Dilution Factor)

Maternal <0.165 (negative) 1120 162.9 1/40 (positive)

Umbilical cord blood <0.165 (negative) 2349 906.4 1/60 (positive)

Maternal to fetal transferratio † / 2.10 5.56 1.50 ‡

† Ratio is defined as the results of umbilical cord blood divided by maternal blood. ‡ pVNT is not a calibrated quantitative method, andthus, the ratio is only given for information. Abbreviations: pVNT, pseudovirus neutralization test; N, nucleocapsid; NR, non-reactive; S,spike; sVNT, surrogate virus neutralization test.

Vaccines 2021, 9, x FOR PEER REVIEW 4 of 9

Table 1. Summary of serological results obtained from the maternal and the umbilical cord blood samples. For pVNT, the highest the dilution factor, the stronger the neutralizing capacity.

Serum Samples Anti-N Titer (U/mL) Anti-S Titer (U/mL) sVNT (AU/mL) pVNT (Dilution

Factor) Maternal <0.165 (negative) 1120 162.9 1/40 (positive)

Umbilical cord blood <0.165 (negative) 2349 906.4 1/60 (positive) Maternal to fetal transfer

ratio † / 2.10 5.56 1.50 ‡

† Ratio is defined as the results of umbilical cord blood divided by maternal blood. ‡ pVNT is not a calibrated quantitative method, and thus, the ratio is only given for information. Abbreviations: pVNT, pseudovirus neutralization test; N, nucleocapsid; NR, non-reactive; S, spike; sVNT, surrogate virus neutralization test.

Figure 1. Serological response in controls, maternal and umbilical cord sera. Control sera selected from the CRO-VAX HCP study cohort and included 13 non-pregnant women aged from 25 to 35 (median, 30 years of age; min–max range, 25–35), without history of SARS-CoV-2 infection. Blood was collected at baseline (day 0), day 14, day 28, day 42, day 56, and day 90. Maternal and umbilical cord blood were only collected on day 90 after the vaccination, at the time of delivery. Positivity thresholds for the Roche RBD total Ab and the YHLO NAbs assays were 0.8 U/mL and 10.0 AU/mL, respectively.

Recent investigations suggested that maternal SARS-CoV-2 immunization, either in-duced by the disease or by the vaccination, may provide neonatal protection through the

Figure 1. Serological response in controls, maternal and umbilical cord sera. Control sera selected from the CRO-VAX HCPstudy cohort and included 13 non-pregnant women aged from 25 to 35 (median, 30 years of age; min–max range, 25–35),without history of SARS-CoV-2 infection. Blood was collected at baseline (day 0), day 14, day 28, day 42, day 56, and day 90.Maternal and umbilical cord blood were only collected on day 90 after the vaccination, at the time of delivery. Positivitythresholds for the Roche RBD total Ab and the YHLO NAbs assays were 0.8 U/mL and 10.0 AU/mL, respectively.

Recent investigations suggested that maternal SARS-CoV-2 immunization, either in-duced by the disease or by the vaccination, may provide neonatal protection through thetransplacental transfer of antibodies [30–33]. Of particular importance was the demon-

Page 19: Clinical manifestations, risk factors, and maternal and

Vaccines 2021, 9, 907 5 of 8

stration that antibody transfer is correlated with the time from vaccination to delivery,which may allow future determination of the optimal timing of COVID-19 vaccination inpregnant women [33]. Compared to the non-pregnant controls, pregnant women exhibitquite comparable vaccine-specific antibody titers, [39] although IgG titers have also beenfound to be slightly lower in pregnant women [13].

Zdanowski et al. observed a slightly lower maternal to fetal ratio, i.e., 1.28 ± 0.80 [33].Mittal et al. also report that the mean IgG titer is similar in maternal and infant sera,but the average gestational age at first vaccine dose was 33 weeks [40]. In our case, 13weeks separate the first vaccine dose to delivery, which could then explain the higher ratioobserved. This observation was also made by Beharier et al. and Mittal et al., and the samephenomenon has been also reported after SARS-CoV-2 infection [30,40]. Thus, the datain the literature are consistent and should now permit us to draw hypothesis on when tovaccine pregnant women in order to achieve the best maternal to fetal transfer ratio. A firstBNT162b2 dose administered during the second trimester could permit high umbilical cordneutralizing antibody titers that are even higher than the ones observed in the maternalblood to be obtained (Table 1).

Interestingly, our case also confirms the presence of neutralizing antibodies in theumbilical cord blood (Table 1). Another case demonstrated that the antibody subtypes,which are transferred, possess a neutralizing capacity, although a clear description of theneutralization assay that was used was not provided [41]. It is also interesting to notethat total antibodies and NAbs titers in the maternal blood were at the lower range in ourcontrol cohort (Figure 1). This may be due to individual factors or may be the consequenceof the maternal to fetal transfer of IgG antibodies, reducing the level of antibodies in thematernal blood.

The observations that we made in this case report following vaccination with BNT162b2are not so surprising. For most pathogens, umbilical cord titers of IgG are higher than inmaternal blood due to endosomal transport of IgG across the syncytiotrophoblast cell bar-rier from maternal to fetal circulation [42–44]. Among these IgG antibodies, galactosylatedIgG1 antibodies are transferred preferentially, followed by IgG3, IgG2, and IgG4 [45]. Thisis potentially the result of enhanced binding to both placental FcRN and FCGR3, enablingthe selective transfer of specific antibody subpopulations to most effectively arm neonatesin the setting of pathogen exposure [45,46]. The fact that the ratio for neutralizing anti-bodies is higher than the one of total antibodies may also be explained by this preferentialtransfer of IgG1, as most serological assays used to assess the humoral response towardsSARS-CoV-2 vaccination target either all types of antibodies or all IgG subtypes. Indeed, itis possible that specific Fc glycoforms can be linked to Fab specificity and subsequently to adifferent neutralizing capacity, as reported for influenza vaccination [47]. Thus, if a specificfraction of neutralizing galactosylated IgG subtypes is transferred from the maternal bloodto the fetus, the neutralizing capacity ratio may increase more sharply than the total levelof antibodies since other types of antibodies, which are not neutralizing and do not transfertrough the placenta, can compensate for the decrease of neutralizing antibodies in thematernal blood.

Interestingly, in line with the above explanations, immunization transfer may alsodepend on whether it is acquired after vaccination or previous infection. Indeed, somecases were reported and described inefficient maternal to fetal transfer following COVID-19disease during pregnancy [48,49]. This could have been explained by Rh isoimmunization,which can compete for IgG transfer within the neonatal Fc receptor (FcRN), but, despitehigh levels of anti-D antibodies, protective antibodies for other pathogens (i.e., rubeolaand varicella) successfully crossed the placenta [48]. This observation can be explained,however, by changes to the Fc-glycosylation profile of the SARS-CoV-2 antibodies due tothe inflammatory state generated by SARS-CoV-2 infection [31,50].

This may influence the neutralizing capacity of the antibodies and may impact thetransfer of immunity, especially during the third trimester [31,47,50]. Nonetheless, thisperturbed placental transfer is probably dependent on the trimester, as observed across two

Page 20: Clinical manifestations, risk factors, and maternal and

Vaccines 2021, 9, 907 6 of 8

independent third trimester-infection cohorts where normalized following second trimesterinfection, suggesting that inflammation-induced alterations in SARS-CoV-2-specific-glycanprofiles may resolve over time from infection [31]. It remains to be known if such alterationsin Fc glycosylation are also observed after vaccination, but in any case, our results andthose from other groups [30,32,33] strongly suggest that BNT162b2 administration in thesecond trimester permit the generation of a strong immune response that can be efficientlytransferred to the fetus in order to protect neonates from SARS-CoV-2 infection.

Although these data have been obtained in a small number of subjects, these arequite reassuring, and the present case demonstrates that the antibody subtypes that aretransferred possess high neutralizing capacity.

4. Conclusions

This case reports on the successful maternal to fetal transfer of neutralizing antibodiesafter vaccination with BNT162b2 in a pregnant woman at 25 weeks of gestation. The levelsof neutralizing antibodies were approximately 5-fold higher in the umbilical cord thanin the maternal blood, while the level of total antibodies showed only a 2-fold increase.This suggests that the antibodies that crossed the syncytiotrophoblast cell barrier havespecific characteristics that correlate to functional neutralizing capacity. Although pregnantand lactating woman have been excluded from clinical trials for several reasons includingethical concerns about fetal exposure, accumulating evidence has now revealed that thesevaccines are safe and efficient for both the fetus and the woman. Vaccination againstCOVID-19 in pregnancy is vital to control disease burden and to decrease morbidity in theante-, peri- and post-natal periods. Inclusion of pregnant women in research programs forthe development of SARS-CoV-2 vaccines should be mandatory to provide this populationwith the equitable benefits of vaccine research.

Author Contributions: Conceptualization, J.D.; methodology, É.D.G., J.F., C.G., and S.V.; formalanalysis, J.D., J.F., and C.G.; investigation, J.D., J.F., and C.G.; resources, É.D.G., J.F., C.G., and S.V.;writing—original draft preparation, J.D.; writing—review and editing, J.-L.B., É.D.G., J.-M.D., J.F.,and C.G.; supervision, J.D.; project administration, J.D. All authors have read and agreed to thepublished version of the manuscript.

Funding: This research has been funded by the authors own funds.

Institutional Review Board Statement: The study was conducted according to the guidelines of theDeclaration of Helsinki and was approved by the Ethics Committee of Clinique Saint-Luc Bouge(protocol registration number: 2020-006149-21—approved on the 14th of January 2021).

Informed Consent Statement: Informed consent was obtained from all subjects involved in thestudy. In addition, written informed consent has been obtained from the patient to publish this paper.

Data Availability Statement: The data presented in this study are available upon request from thecorresponding author. The data are not publicly available due to ethical and privacy reasons.

Acknowledgments: The authors would like to acknowledge the mother and the neonate for theirparticipation in this study as well as the subjects from the CRO-Vax study who served as controls.The authors would also like to thank the midwifes at the Centre Hospitalier Régional de Huy forassisting during the delivery.

Conflicts of Interest: The authors declare no conflict of interest in relation with the present study.

References1. Zambrano, L.D.; Ellington, S.; Strid, P.; Galang, R.R.; Oduyebo, T.; Tong, V.T.; Woodworth, K.R.; Nahabedian, J.F., 3rd; Azziz-

Baumgartner, E.; Gilboa, S.M.; et al. Update: Characteristics of Symptomatic Women of Reproductive Age with Laboratory-Confirmed SARS-CoV-2 Infection by Pregnancy Status—United States, January 22–October 3, 2020. MMWR Morb. Mortal. Wkly.Rep. 2020, 69, 1641–1647. [CrossRef]

2. Ellington, S.; Strid, P.; Tong, V.T.; Woodworth, K.; Galang, R.R.; Zambrano, L.D.; Nahabedian, J.; Anderson, K.; Gilboa, S.M.Characteristics of Women of Reproductive Age with Laboratory-Confirmed SARS-CoV-2 Infection by Pregnancy Status–UnitedStates, January 22–June 7, 2020. MMWR Morb. Mortal. Wkly. Rep. 2020, 69, 769–775. [CrossRef]

Page 21: Clinical manifestations, risk factors, and maternal and

Vaccines 2021, 9, 907 7 of 8

3. Riley, L.E. mRNA COVID-19 Vaccines in Pregnant Women. N. Engl. J. Med. 2021, 384, 2342–2343. [CrossRef] [PubMed]4. Schwartz, D.A.; Graham, A.L. Potential Maternal and Infant Outcomes from (Wuhan) Coronavirus 2019-nCoV Infecting Pregnant

Women: Lessons from SARS, MERS, and Other Human Coronavirus Infections. Viruses 2020, 12, 194. [CrossRef] [PubMed]5. Pettirosso, E.; Giles, M.; Cole, S.; Rees, M. COVID-19 and pregnancy: A review of clinical characteristics, obstetric outcomes and

vertical transmission. Aust. New Zealand J. Obstet. Gynaecol. 2020, 60, 640–659. [CrossRef]6. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Perez Marc, G.; Moreira, E.D.;

Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 mRNA COVID-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615.[CrossRef] [PubMed]

7. Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al.Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416. [CrossRef] [PubMed]

8. Sadoff, J.; Gray, G.; Vandebosch, A.; Cardenas, V.; Shukarev, G.; Grinsztejn, B.; Goepfert, P.A.; Truyers, C.; Fennema, H.;Spiessens, B.; et al. Safety and Efficacy of Single-Dose Ad26.COV2.S Vaccine against COVID-19. N. Engl. J. Med. 2021, 384,2187–2201. [CrossRef]

9. Logunov, D.Y.; Dolzhikova, I.V.; Shcheblyakov, D.V.; Tukhvatulin, A.I.; Zubkova, O.V.; Dzharullaeva, A.S.; Kovyrshina, A.V.;Lubenets, N.L.; Grousova, D.M.; Erokhova, A.S.; et al. Safety and efficacy of an rAd26 and rAd5 vector-based heterologousprime-boost COVID-19 vaccine: An interim analysis of a randomised controlled phase 3 trial in Russia. Lancet 2021, 397, 671–681.[CrossRef]

10. Voysey, M.; Clemens, S.A.C.; Madhi, S.A.; Weckx, L.Y.; Folegatti, P.M.; Aley, P.K.; Angus, B.; Baillie, V.L.; Barnabas, S.L.;Bhorat, Q.E.; et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: An interim analysis offour randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021, 397, 99–111. [CrossRef]

11. Maykin, M.M.; Heuser, C.; Feltovich, H. Pregnant people deserve the protection offered by SARS-CoV-2 vaccines. Vaccine 2021,39, 171–172. [CrossRef]

12. Craig, A.M.; Hughes, B.L.; Swamy, G.K. Coronavirus disease 2019 vaccines in pregnancy. Am. J. Obstet. Gynecol. MFM 2021, 3,100295. [CrossRef] [PubMed]

13. Bookstein Peretz, S.; Regev, N.; Novick, L.; Nachshol, M.; Goffer, E.; Ben-David, A.; Asraf, K.; Doolman, R.; Sapir, E.; RegevYochay, G.; et al. Short-term outcome of pregnant women vaccinated by BNT162b2 mRNA COVID-19 vaccine. Ultrasound Obs.Gynecol. 2021. [CrossRef] [PubMed]

14. Donders, G.G.G.; Grinceviciene, S.; Haldre, K.; Lonnee-Hoffmann, R.; Donders, F.; Tsiakalos, A.; Adriaanse, A.; Martinez deOliveira, J.; Ault, K.; Mendling, W.; et al. ISIDOG Consensus Guidelines on COVID-19 Vaccination for Women before, during andafter Pregnancy. J. Clin. Med. 2021, 10, 2902. [CrossRef]

15. Ory, S.; Veiga, A.; Horton, M.; Gianaroli, L. Joint IFFS/ESHRE statement on COVID-19 vaccination for pregnant women andthose considering pregnancy. Hum. Reprod. Open 2021, 2021, hoab016. [CrossRef] [PubMed]

16. Society for Maternal-Fetal Medicine. Society for Maternal-Fetal Medicine (SMFM) Statement: SARS-CoV-2 Vaccination inPregnancy. Available online: https://s3.amazonaws.com/cdn.smfm.org/media/2591/SMFM_Vaccine_Statement_12-1-20_(final).pdf (accessed on 21 July 2021).

17. Razzaghi, H.; Kahn, K.E.; Black, C.L.; Lindley, M.C.; Jatlaoui, T.C.; Fiebelkorn, A.P.; Havers, F.P.; D’Angelo, D.V.; Cheung, A.;Ruther, N.A.; et al. Influenza and Tdap Vaccination Coverage Among Pregnant Women–United States, April 2020. MMWR Morb.Mortal. Wkly. Rep. 2020, 69, 1391–1397. [CrossRef]

18. Levy, A.T.; Singh, S.; Riley, L.E.; Prabhu, M. Acceptance of COVID-19 vaccination in pregnancy: A survey study. Am. J. Obstet.Gynecol. MFM 2021, 3, 100399. [CrossRef]

19. Gerretsen, H.E.; Sande, C.J. Development of respiratory syncytial virus (RSV) vaccines for infants. J. Infect. 2017, 74 (Suppl. 1),S143–S146. [CrossRef]

20. Rasmussen, S.A.; Jamieson, D.J.; Uyeki, T.M. Effects of influenza on pregnant women and infants. Am. J. Obstet. Gynecol. 2012,207, S3–S8. [CrossRef] [PubMed]

21. Zaman, K.; Roy, E.; Arifeen, S.E.; Rahman, M.; Raqib, R.; Wilson, E.; Omer, S.B.; Shahid, N.S.; Breiman, R.F.; Steinhoff, M.C.Effectiveness of maternal influenza immunization in mothers and infants. N. Engl. J. Med. 2008, 359, 1555–1564. [CrossRef]

22. Dong, Y.; Mo, X.; Hu, Y.; Qi, X.; Jiang, F.; Jiang, Z.; Tong, S. Epidemiology of COVID-19 Among Children in China. Pediatrics 2020,145, e20200702. [CrossRef]

23. Mithal, L.B.; Machut, K.Z.; Muller, W.J.; Kociolek, L.K. SARS-CoV-2 Infection in Infants Less than 90 Days Old. J. Pediatr. 2020,224, 150–152. [CrossRef] [PubMed]

24. Kim, L.; Whitaker, M.; O’Halloran, A.; Kambhampati, A.; Chai, S.J.; Reingold, A.; Armistead, I.; Kawasaki, B.; Meek, J.; Yousey-Hindes, K.; et al. Hospitalization Rates and Characteristics of Children Aged <18 Years Hospitalized with Laboratory-ConfirmedCOVID-19—COVID-NET, 14 States, March 1–July 25, 2020. MMWR Morb. Mortal. Wkly. Rep. 2020, 69, 1081–1088. [CrossRef][PubMed]

25. Richtmann, R.; Torloni, M.R.; Oyamada Otani, A.R.; Levi, J.E.; Crema Tobara, M.; de Almeida Silva, C.; Dias, L.;Miglioli-Galvao, L.; Martins Silva, P.; Macoto Kondo, M. Fetal deaths in pregnancies with SARS-CoV-2 infection in Brazil: A caseseries. Case Rep. Womens Health 2020, 27, e00243. [CrossRef] [PubMed]

26. Prochaska, E.; Jang, M.; Burd, I. COVID-19 in pregnancy: Placental and neonatal involvement. Am. J. Reprod. Immunol. 2020, 84,e13306. [CrossRef]

Page 22: Clinical manifestations, risk factors, and maternal and

Vaccines 2021, 9, 907 8 of 8

27. Zimmermann, P.; Curtis, N. COVID-19 in Children, Pregnancy and Neonates: A Review of Epidemiologic and Clinical Features.Pediatr. Infect. Dis. J. 2020, 39, 469–477. [CrossRef]

28. Gurol-Urganci, I.; Jardine, J.E.; Carroll, F.; Draycott, T.; Dunn, G.; Fremeaux, A.; Harris, T.; Hawdon, J.; Morris, E.; Muller, P.; et al.Maternal and perinatal outcomes of pregnant women with SARS-CoV-2 infection at the time of birth in England: National cohortstudy. Am. J. Obstet. Gynecol. 2021. [CrossRef]

29. Estes, M.L.; McAllister, A.K. Maternal immune activation: Implications for neuropsychiatric disorders. Science 2016, 353, 772–777.[CrossRef]

30. Beharier, O.; Plitman Mayo, R.; Raz, T.; Nahum Sacks, K.; Schreiber, L.; Suissa-Cohen, Y.; Chen, R.; Gomez-Tolub, R.; Hadar, E.;Gabbay-Benziv, R.; et al. Efficient maternal to neonatal transfer of antibodies against SARS-CoV-2 and BNT162b2 mRNACOVID-19 vaccine. J. Clin. Investig. 2021, 131. [CrossRef]

31. Atyeo, C.; Pullen, K.M.; Bordt, E.A.; Fischinger, S.; Burke, J.; Michell, A.; Slein, M.D.; Loos, C.; Shook, L.L.; Boatin, A.A.; et al.Compromised SARS-CoV-2-specific placental antibody transfer. Cell 2021, 184, 628–642.e610. [CrossRef]

32. Gray, K.J.; Bordt, E.A.; Atyeo, C.; Deriso, E.; Akinwunmi, B.; Young, N.; Baez, A.M.; Shook, L.L.; Cvrk, D.; James, K.; et al.Coronavirus disease 2019 vaccine response in pregnant and lactating women: A cohort study. Am. J. Obste.t Gynecol 2021.[CrossRef] [PubMed]

33. Zdanowski, W.; Wasniewski, T. Evaluation of SARS-CoV-2 Spike Protein Antibody Titers in Cord Blood after COVID-19Vaccination during Pregnancy in Polish Healthcare Workers: Preliminary Results. Vaccines 2021, 9, 675. [CrossRef] [PubMed]

34. Favresse, J.; Bayart, J.L.; Mullier, F.; Dogne, J.M.; Closset, M.; Douxfils, J. Early antibody response in health-care professionalsafter two doses of SARS-CoV-2 mRNA vaccine (BNT162b2). Clin. Microbiol. Infect. 2021. [CrossRef]

35. Favresse, J.; Bayart, J.L.; Mullier, F.; Elsen, M.; Eucher, C.; Van Eeckhoudt, S.; Roy, T.; Wieers, G.; Laurent, C.; Dogne, J.M.; et al.Antibody titres decline 3-month post-vaccination with BNT162b2. Emerg. Microbes Infect. 2021, 10, 1495–1498. [CrossRef][PubMed]

36. Bayart, J.L.; Morimont, L.; Closset, M.; Wieers, G.; Roy, T.; Gerin, V.; Elsen, M.; Eucher, C.; Van Eeckhoudt, S.; Ausselet, N.; et al.Confounding Factors Influencing the Kinetics and Magnitude of Serological Response Following Administration of BNT162b2.Microorganisms 2021, 9, 1340. [CrossRef]

37. Favresse, J.; Eucher, C.; Elsen, M.; Tre-Hardy, M.; Dogne, J.M.; Douxfils, J. Clinical Performance of the Elecsys Electrochemilumi-nescent Immunoassay for the Detection of SARS-CoV-2 Total Antibodies. Clin. Chem. 2020, 66, 1104–1106. [CrossRef]

38. Favresse, J.; Gillot, C.; Di Chiaro, L.; Eucher, C.; Elsen, M.; Van Eeckhoudt, S.; David, C.; Morimont, L.; Dogné, J.-M.; Douxfils, J.Neutralizing Antibodies in COVID-19 Patients and Vaccine Recipients after Two Doses of BNT162b2. Viruses 2021, 13, 1364.[CrossRef]

39. Atyeo, C.; Deriso, E.A.; Davis, C.; Bordt, E.A.; Deguzman, R.M.; Shook, L.L.; Yonker, L.M.; Fasano, A.; Akinwunmi, B.;Lauffenburger, D.A.; et al. COVID-19 mRNA vaccines drive differential Fc-functional profiles in pregnant, lactating, andnon-pregnant women. bioRxiv 2021. [CrossRef]

40. Mithal, L.B.; Otero, S.; Shanes, E.D.; Goldstein, J.A.; Miller, E.S. Cord blood antibodies following maternal coronavirus disease2019 vaccination during pregnancy. Am. J. Obstet. Gynecol. 2021, 2225, 192–194. [CrossRef]

41. Gill, L.; Jones, C.W. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Antibodies in Neonatal Cord Blood AfterVaccination in Pregnancy. Obstet. Gynecol. 2021, 137, 894–896. [CrossRef]

42. Munoz, F.M.; Bond, N.H.; Maccato, M.; Pinell, P.; Hammill, H.A.; Swamy, G.K.; Walter, E.B.; Jackson, L.A.; Englund, J.A.;Edwards, M.S.; et al. Safety and immunogenicity of tetanus diphtheria and acellular pertussis (Tdap) immunization duringpregnancy in mothers and infants: A randomized clinical trial. JAMA 2014, 311, 1760–1769. [CrossRef] [PubMed]

43. Martinez, D.R.; Fong, Y.; Li, S.H.; Yang, F.; Jennewein, M.F.; Weiner, J.A.; Harrell, E.A.; Mangold, J.F.; Goswami, R.;Seage, G.R., 3rd; et al. Fc Characteristics Mediate Selective Placental Transfer of IgG in HIV-Infected Women. Cell 2019, 178,190–201.e11. [CrossRef]

44. Firan, M.; Bawdon, R.; Radu, C.; Ober, R.J.; Eaken, D.; Antohe, F.; Ghetie, V.; Ward, E.S. The MHC class I-related receptor, FcRn,plays an essential role in the maternofetal transfer of gamma-globulin in humans. Int. Immunol. 2001, 13, 993–1002. [CrossRef]

45. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG subclasses and allotypes: From structure to effector functions. Front. Immunol. 2014, 5,520. [CrossRef] [PubMed]

46. Jennewein, M.F.; Goldfarb, I.; Dolatshahi, S.; Cosgrove, C.; Noelette, F.J.; Krykbaeva, M.; Das, J.; Sarkar, A.; Gorman, M.J.;Fischinger, S.; et al. Fc Glycan-Mediated Regulation of Placental Antibody Transfer. Cell 2019, 178, 202–215.e214. [CrossRef]

47. Wang, T.T.; Maamary, J.; Tan, G.S.; Bournazos, S.; Davis, C.W.; Krammer, F.; Schlesinger, S.J.; Palese, P.; Ahmed, R.; Ravetch, J.V.Anti-HA Glycoforms Drive B Cell Affinity Selection and Determine Influenza Vaccine Efficacy. Cell 2015, 162, 160–169. [CrossRef]

48. Toner, L.E.; Gelber, S.E.; Pena, J.A.; Fox, N.S.; Rebarber, A. A Case Report to Assess Passive Immunity in a COVID PositivePregnant Patient. Am. J. Perinatol. 2020, 37, 1280–1282. [CrossRef]

49. Zeng, H.; Xu, C.; Fan, J.; Tang, Y.; Deng, Q.; Zhang, W.; Long, X. Antibodies in Infants Born to Mothers with COVID-19 Pneumonia.JAMA 2020, 323, 1848–1849. [CrossRef] [PubMed]

50. Chakraborty, S.; Gonzalez, J.; Edwards, K.; Mallajosyula, V.; Buzzanco, A.S.; Sherwood, R.; Buffone, C.; Kathale, N.; Providenza, S.;Xie, M.M.; et al. Proinflammatory IgG Fc structures in patients with severe COVID-19. Nat. Immunol. 2021, 22, 67–73. [CrossRef]

Page 23: Clinical manifestations, risk factors, and maternal and

COVID-19 mRNA Vaccination in Lactation: Assessment of adverse effects and 1

transfer of anti-SARS-CoV2 antibodies from mother to child 2

3

Yarden Golan, Ph.D.1, Mary Prahl, M.D.2,3, Arianna G. Cassidy, M.D.4, Caryl Gay Ph.D. 4

5, Alan H.B. Wu, Ph.D. 6, Unurzul Jigmeddagva, M.D. 7, Christine Y. Lin4, Veronica J. 5

Gonzalez, M.D.4, Emilia Basilio, M.D, M.P.H4, Lakshmi Warrier8, Sirirak Buarpung, 6

D.V.M., Ph.D. 4, Lin Li, M.D., Ph.D. 4, Ifeyinwa V. Asiodu, Ph.D, R.N. 5, Nadav Ahituv, 7

Ph.D.1, Valerie J. Flaherman, M.D., M.P.H.2, Stephanie L. Gaw, M.D., Ph.D. 4, 7 8

9

Author Affiliations 10

1 Department of Bioengineering and Therapeutic Sciences, University of California, San 11

Francisco, CA, 94158 and Institute for Human Genetics, University of California, San 12

Francisco, California, United States of America. 13

2 Department of Pediatrics, University of California, San Francisco, California, United 14

States of America. 15

3Division of Pediatric Infectious Diseases and Global Health, University of California, 16

San Francisco, California, United States of America. 17

4 Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and 18

Reproductive Sciences, University of California San Francisco, California, United States 19

of America. 20

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.

Page 24: Clinical manifestations, risk factors, and maternal and

5 Department of Family Health Care Nursing, University of California, San Francisco, 21

California, United States of America. 22

6 Department of Laboratory Medicine, University of California, San Francisco, California, 23

United States of America. 24

7 Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and 25

Reproductive Sciences, University of California San Francisco, California, United States 26

of America. 27

8 Department of Medicine, University of California, San Francisco, California, United 28

States of America. 29

Corresponding author: Stephanie L. Gaw- Division of Maternal-Fetal Medicine, 30

Department of Obstetrics, Gynecology, and Reproductive Sciences, University of 31

California San Francisco, 513 Parnassus Ave, HSE16, Box 0556, San Francisco, CA 32

94143. Phone: 415.476.0535. Email: [email protected] 33

34

35

Conflict of interest: 36

The authors have declared that no conflict of interest exists. 37

38

39

40

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 25: Clinical manifestations, risk factors, and maternal and

Abstract: 41

Background: Data regarding the rate of adverse effects observed in the lactating 42

mother-infant dyad and their immune response to COVID-19 mRNA vaccination during 43

lactation are needed to inform vaccination guidelines. 44

Methods: From a prospective cohort of 50 lactating individuals who received mRNA-45

based vaccines for COVID-19 (mRNA-1273 and BNT162b2), blood and milk samples 46

were collected prior to first vaccination dose, immediately prior to 2nd dose, and 4-10 47

weeks after 2nd dose. Symptoms in mother and infant were assessed by detailed 48

questionnaires. Anti-SARS-CoV-2 antibody levels in blood and milk were measured by 49

Pylon 3D automated immunoassay and ELISA, and PEGylated proteins in milk were 50

measured by ELISA. Blood samples were collected from a subset of infants whose 51

mothers received the vaccine during lactation or pregnancy (4-15 weeks after mothers’ 52

2nd dose). 53

Results: No severe maternal or infant adverse effects were reported in this cohort. Two 54

mothers and two infants were diagnosed with COVID-19 during the study period. 55

Vaccine-related products, PEGylated proteins, were not found at significant levels in 56

milk after vaccination. After vaccination, levels of anti-SARS-CoV-2 IgG and IgM 57

significantly increased in maternal plasma and there was significant transfer of anti-58

SARS-CoV-2-Receptor Binding Domain (anti-RBD) IgA and IgG antibodies to milk. Milk 59

IgA levels after 2nd dose were negatively associated with infant age. Anti-SARS-CoV-2 60

IgG antibodies were not detected in the plasma of infants whose mothers were 61

vaccinated during lactation. 62

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 26: Clinical manifestations, risk factors, and maternal and

Conclusions: COVID-19 mRNA vaccines generate robust immune responses in 63

plasma and milk of lactating individuals without severe adverse effects reported. 64

65

66

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 27: Clinical manifestations, risk factors, and maternal and

Introduction: 67

An important benefit of human milk is the presence of IgA and IgG antibodies that 68

provide passive immunity to the infant (1, 2). Anti-SARS-CoV-2 antibodies are present 69

in milk from lactating women who were infected with SARS-CoV-2 (3, 4) or who 70

received COVID-19 mRNA vaccines (5–8). Specifically, high titers of anti-SARS-CoV-2 71

IgG were reported after vaccination(6). The function of these antibodies in protection of 72

infants against COVID-19 is not fully understood. Recently, work has demonstrated that 73

murine pups can transfer IgG antibodies from ingested milk to their bloodstream, via 74

enteric Fc receptors (9); however, this phenomenon has not been demonstrated in 75

humans. To better understand the function and distribution if human milk antibodies and 76

infant immunization after maternal vaccination, we examine anti-SARS-CoV-2 antibody 77

levels in infant stool and blood. In addition, information is lacking on the potential 78

adverse effects of COVID-19 vaccination on lactating mothers and their infants, who 79

were excluded from initial clinical trials of mRNA vaccination (10). Many breastfeeding 80

individuals have concerns regarding the potential effects on their infant due to the lack 81

of data, leading to delayed vaccination or early cessation of breastfeeding (11). 82

In this study, we examined blood and milk samples from lactating mothers who received 83

a COVID-19 mRNA vaccine and their infants for the presence of anti- SARS-CoV2 84

antibodies, presence of PEGylated protein vaccine products, and self-reported vaccine-85

related symptoms in order to address the gap of knowledge regarding vaccination 86

efficacy and safety in this population. 87

88

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 28: Clinical manifestations, risk factors, and maternal and

Results: 89

Participant characteristics. During the study period, 50 participants answered all 90

study questionnaires, provided blood and/or milk samples, had an infant up to 18 91

months old and were included in this analysis. Two infants were diagnosed with COVID-92

19 during this study (Table S1, infant of participants 1 and 2). One mother reported that 93

her infant had mild symptoms 1 week after the 2nd dose; this infant’s vaccinated mother 94

had a negative test at the time of the infant’s positive PCR test. A second infant had 95

positive plasma anti-SARS-CoV-2 IgG and IgA, despite the mother receiving the 96

vaccine postpartum and reported no known prior COVID-19 infection. The mother’s 97

plasma was subsequently found to be positive to antibodies against SARS-CoV-2 98

nucleocapsid protein, indicating a likely natural asymptomatic SARS-CoV-2 infection 99

(further details in Table S1). Two mothers were positive for COVID-19 and are 100

presented in Table S1 (participants 2 and 3); they were excluded from further analysis 101

of symptomatology. Cohort characteristics are presented in Table 1. Twenty-seven 102

female participants (mean age 35.7 years (± 3.9)) received the BNT162b2 vaccine 103

(Pfizer, 56%), and 21 received the mRNA-1237 (Moderna, 44%). The mean infant age 104

at mother’s 1st dose was 5 months (±3.9). All mothers continued to feed their infants 105

with milk at the time of the 2nd vaccination, and all except one continued up to the time 106

of follow up sample collection (4-10 weeks after 2nd dose). There were no significant 107

differences in maternal or infant characteristics by vaccine manufacturer. 108

109

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 29: Clinical manifestations, risk factors, and maternal and

Post vaccination symptoms. Self-reported symptoms after each vaccine dose are 110

presented in Table 2. Fever, chills, headache, joint pain, muscle aches or body aches, 111

and fatigue or tiredness were reported by significantly more participants after the 2nd 112

dose than after the 1st dose (Table 2). All 21 participants (100%) who received the 113

mRNA-1237 vaccine reported injection site symptoms, while only 21 (78%) of 27 BNT-114

162b2 recipients reported injection site symptoms (p=0.02) (Table 2). Two mothers 115

reported slightly less milk production in the first 24-72 hours after vaccine doses (Table 116

2). With respect to infant symptoms, 12% of mothers reported at least one symptom 117

after the 1st maternal vaccine dose (primarily gastrointestinal symptoms and sleep 118

changes), and none reported an infant symptom after the 2nd dose (Table 3). In 119

summary, no severe adverse effects for mothers or nursing infants were reported in this 120

cohort after vaccination, and reported symptoms resolved up to 72 hours after 121

vaccination. 122

PEG detection in human milk. Polyethylene glycol (PEG) is present in the lipid 123

nanoparticles of the mRNA-based vaccines, and was reported to cause allergic reaction 124

after vaccination in rare cases (12, 13). To address concerns about vaccine 125

components passing to milk after vaccination, we performed ELISA assays to measure 126

PEG-ylated proteins levels in milk after vaccination from 13 participants. PEG-ylated 127

proteins were measured in milk samples collected before the vaccine, and at various 128

time points post-vaccination (from 24 hours after 1st dose to 2 weeks after 2nd dose). 129

Pre-vaccine PEG-ylated proteins levels did not significantly differ from PEG-ylated 130

proteins levels at any post-vaccine time point in either paired or unpaired comparisons 131

(Figure 1). 132

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 30: Clinical manifestations, risk factors, and maternal and

Anti-SARS-CoV-2 antibody levels in blood and milk samples after vaccination. We 133

analyzed blood and milk samples from lactating individuals for anti-SARS-CoV-2 134

antibodies to measure immune response after vaccination. Maternal blood anti-SARS-135

CoV-2 IgM and IgG antibodies increased significantly after the 1st dose (Figure 2). Anti-136

SARS-CoV-2 IgM levels were not significantly higher 4-10 weeks after the 2nd dose 137

compared to samples collected after dose 1 (on the day of the 2nd dose) (Figure 2A). 138

In contrast, anti-SARS-CoV-2 IgG levels increased significantly after the 2nd dose (P 139

value <0.0001) when compared to samples collected immediately prior to the 2nd dose 140

(Figure 2B). There was no significant difference in blood antibody levels between 141

participants who received the mRNA-1237 compared to the BNT-162b2 vaccine after 142

dose 2. 143

We found significantly higher levels of IgA antibodies specific to SARS-CoV-2 RBD 144

protein in human milk samples collected after the 1st dose of both BNT-162b2 and 145

mRNA-1237 vaccines (Figure 3A and 3B). There was no significant increase in milk 146

anti-RBD IgA after the 2nd vaccination as compared to after dose 1 (Figure 3A and 147

3B). Twelve individuals (25%, BNT-162b2 n=7; mRNA-1237 n=5) did not have 148

detectable levels of anti-RBD IgA after either the 1st or 2nd dose (infants age at 1st 149

dose range 1-11 months). Milk anti-RBD IgG levels increased after the 1st dose of 150

vaccine and increased further after the 2nd dose (Figure 3C and 3D). There were no 151

significant differences in milk anti-RBD IgG levels between women who received BNT-152

162b2 (Figure 3C) and mRNA-1237 (Figure 3D). These findings suggest that mRNA 153

vaccine results in a robust immune response leading to increased anti SARS-CoV-2 154

antibody levels in blood, but also in milk during lactation. 155

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 31: Clinical manifestations, risk factors, and maternal and

Correlations between antibody levels, participant characteristics, and symptoms. 156

In order to better understand the differences in antibody responses between individuals 157

in our cohort, we performed multiple correlation tests to determine whether IgG and IgA 158

antibodies levels correlated with timing of sample collection after vaccination (range 4-159

10 weeks after 2nd dose), infant age at time of vaccination, or maternal BMI (Table S2). 160

Milk IgA (but not IgG) levels declined significantly as the infant age at time of 161

vaccination increased (Figure 4A and 4B). There was no significant correlation 162

between IgG and IgA levels and either the length of time after 2nd dose or maternal BMI 163

(Table S2 and S3). The levels of IgG and IgA antibodies induced in milk were 164

significantly correlated after 1st dose (Figure 4C), but not after the 2nd dose (Figure 165

4D). There was no correlation between the anti SARS-CoV-2 IgG levels in blood and 166

milk after 1st dose (Figure 4E), but there was a positive correlation between levels at 4-167

10 weeks after 2nd dose (Figure 4F). 168

Plasma levels of anti-SARS-CoV-2 IgG are not detectable in infants after maternal 169

vaccination during lactation. Although maternal IgG antibodies have been shown in 170

multiple studies to transfer to the infant in utero, there are little data to suggest that milk-171

derived antibodies are similarly transferred to the infant blood circulation during 172

breastfeeding. To investigate whether passive immunity is conferred to the infant’s 173

blood after maternal vaccination during lactation, we analyzed infant blood samples 174

from mothers who were vaccinated after delivery (n=8) and compared to infant blood 175

samples form mothers who were vaccinated against COVID-19 during pregnancy (n=2). 176

Blood samples were collected from these 10 infants (5 male, 5 female) at 61 days to 1 177

year of age (Table S4). Plasma was tested for the presence of anti-SARS-CoV-2 IgG 178

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 32: Clinical manifestations, risk factors, and maternal and

and IgM and anti-RBD IgA. We evaluated infant blood samples collected at time frame 179

of 4-15 weeks after 2nd dose as this time point corresponded to high anti-SARS-CoV-2 180

IgG levels in mothers’ blood and milk (Figure 2 and 3). No antibodies were detected in 181

the blood of nursing infants born to mothers who were vaccinated postpartum (Figure 182

5), despite high IgG levels in maternal blood and milk. In contrast, infants born to 183

mothers who received both doses of vaccine during pregnancy had detectable plasma 184

anti-SARS-CoV-2 IgG levels at follow-up (infant age range 61-65 days, 125-129 days 185

after maternal dose 1) (Figure 5). None of the follow-up infant blood samples (from the 186

lactation or pregnancy cohorts) had detectable levels of anti-RBD IgA antibodies. These 187

results demonstrate that vaccination during lactation induces anti-SARS-CoV-2 188

antibodies in human milk, but only vaccination during pregnancy and not lactation 189

provides additional transfer of anti-SARS-CoV-2 antibodies to the infant blood. 190

We also investigated whether ingested antibodies could survive passage through the 191

infant gastrointestinal tract by testing infant stool for anti-SARS-CoV-2 antibodies after 192

vaccination (Figure S1) in one mother-infant dyad. We found that IgG (but not IgA) was 193

detected in infant stool samples and were correlated with maternal milk anti-SARS-CoV-194

2 IgG levels. 195

196

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 33: Clinical manifestations, risk factors, and maternal and

Conclusion: 197

Our study provides one of the first detailed reports on patient symptoms and antibody 198

responses of the COVID-19 mRNA vaccines in lactating mothers. We found that the 199

rates of reported symptoms were similar to the CDC report from the V-Safe registry(14) 200

but higher than described in the clinical trials, although we do not have a non-lactating 201

comparison group. Importantly, there were no significant side effects noted in the infants 202

of mothers vaccinated during breastfeeding. Comparing the mRNA vaccines made by 203

current manufacturers, we found that lactating individuals may experience more 204

vaccine-related side effects after mRNA-1237 compared to BNT-162b2 vaccine; 205

however, no differences in immune response were observed between those vaccines. 206

It has recently been shown in a few studies that vaccine mRNA is not present in 207

milk samples after vaccination(15, 16), or is only detected in very low levels in some 208

cases(17), providing reassurance that risks of exposure to the breastfed infant are 209

minimal. This study adds to the evidence that vaccine components are minimally 210

transferred to human milk. We found no significant increase in milk PEG-ylated protein 211

concentrations at various time points after vaccine administration in a subset of samples 212

analyzed in our cohort. We did observe one sample with higher ratio of PEG-ylated 213

proteins 24 hours post vaccination (Figure 1, patient 030). This sample had PEG-ylated 214

protein levels equivalent to 2.8µl/ml vaccine. However, we cannot confirm that the 215

increased PEG in this single sample was from the COVID-19 vaccine, as PEG exposure 216

may also be from other sources, such laxatives or ibuprofen. There was no increase in 217

protein PEG-ylation ratio after 2nd dose in the same individual, and no unusual 218

symptoms were reported in either the mother or her infant. Larger studies are needed to 219

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 34: Clinical manifestations, risk factors, and maternal and

increase our understanding of transfer of PEG into human milk, and potential effects 220

after ingestion by the infant. Although expert consensus states there is minimal or no 221

potential risk for the infant from maternal COVID-19 vaccination(18, 19), the minor 222

symptoms that were reported (sleep changes and gastrointestinal symptoms) should be 223

further investigated in future studies to determine if they are related to vaccination. Our 224

findings also suggest that administration of maternal mRNA-based vaccine during 225

lactation did not lead to a detectable immune response in the infant blood, which further 226

suggests that the vaccine is not transferred to the infant via milk and cannot trigger 227

infant immune responses via that route. 228

We also demonstrate that COVID-19 mRNA vaccination induces significant 229

increases in anti-SARS-CoV-2 IgM and IgG levels in lactating mothers’ blood. 230

Consistent with previous studies that showed IgM levels plateaued 28 days after 231

COVID-19 infection (20), our results also demonstrated that dose 2 did not induce 232

significantly higher levels of IgM than was is observed after dose 1. In contrast, 233

maternal blood IgG levels increased by 6-fold after the 2nd dose (compare to the levels 234

after the 1st dose), highlighting the importance of the 2nd dose to boost the antibody 235

response (21). 236

Due to the lack of data about vaccination during pregnancy, many pregnant 237

individuals were initially denied access to, declined, or were recommended to delay 238

vaccination until after pregnancy. As such, many mothers have waited until after 239

delivery to receive the vaccine. Although mothers vaccinated during lactation 240

transferred antibodies to their infant through milk, which is an important component of 241

mucosal immunity for the baby, there was no passive transfer of antibodies to the infant 242

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 35: Clinical manifestations, risk factors, and maternal and

bloodstream, as occurs if the mother is vaccinated during pregnancy. Correlates of 243

infant immune protection to COVID-19 are not yet well understood, however passive in 244

utero transfer of IgG to the infant is important in the prevention of a number of infections 245

including pertussis and influenza (22–24). Passively-transferred milk-derived IgA likely 246

provides partial mucosal immune protection in infants, as breastfeeding is associated 247

with lower risk of infections associated with mucosal defense, especially against 248

respiratory infections (25–27). 249

Two nursing infants in our cohort were infected with COVID-19 during the study 250

(one a week post maternal 2nd dose, and the second one between 1st and 2nd 251

maternal vaccine), indicating that milk antibodies cannot fully protect against SARS-252

CoV-2 infection, especially at the time before full immune response is achieved in the 253

vaccinated mother, typically 2-3 weeks after the booster dose (Figure 2D). Further 254

studies are needed to determine the degree of protection conferred by IgA and IgG anti-255

SARS-CoV-2 antibodies that are present in milk. Interestingly, we demonstrated for the 256

first time that anti-SARS-CoV-2 RBD IgG antibodies are detectable in stool samples 257

collected 4 and 8 weeks post maternal vaccine (Figure S1), which suggests that milk-258

derived antibodies can persist in the infant gastrointestinal tract and provide protection 259

against viral infection via the digestive system(28). Further studies evaluating the 260

additive benefit of both transplacentally-derived maternal IgG, as well as milk-derived 261

IgA and IgG are needed to determine protection against COVID-19 in early infancy. Our 262

findings demonstrate the importance of determining the optimal timing of vaccine 263

administration to confer maximal protection against COVID-19 in infancy. 264

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 36: Clinical manifestations, risk factors, and maternal and

Twenty-five percent of women in our cohort had no detectable levels of anti-RBD 265

IgA in their milk after vaccination. Similar findings were reported in other studies (5, 6), 266

suggesting that production and transfer efficiency vary between individuals. Our 267

analysis showed a negative correlation between infant age and milk anti-RBD IgA 268

levels, which might explain some of the variation in milk IgA levels observed between 269

different individuals. Ten out of the 12 participants who had no detectable anti-RBD IgA, 270

had infants older than 5.5 months at the time of sample collection. Further studies are 271

needed to investigate the relationship between infant age, breastfeeding exclusivity, 272

milk IgA antibodies, and optimal timing of vaccination during lactation. 273

Strengths of our study include the prospective design and comprehensive 274

symptom reporting by the vaccinated participants. We also report on longitudinal follow-275

up of infant immune responses, which has not been previously described. Furthermore, 276

we included both BNT162b2 and mRNA-1273 vaccines and compared responses 277

between the two vaccine manufacturers. Limitations include the small sample size, and 278

that not all samples were able to be collected from all infant participants. 279

In summary, our study reports that no severe adverse effects were noted in 280

lactating individuals or their breastfeeding infants after COVID-19 mRNA vaccination. 281

We demonstrated that human milk confers passive immunity to the infants, primarily 282

through mucosal immunity in the gastrointestinal tract provided by IgA and IgG in milk. 283

These results are important evidence to aid in counseling lactating individuals on the 284

safety and efficacy of the COVID-19 mRNA vaccines, and the potential benefits to both 285

the mother and infant. 286

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 37: Clinical manifestations, risk factors, and maternal and

Methods: 287

Study approval and study population. The institutional review board of the University of 288

California San Francisco approved the study. Written, informed consent was obtained 289

from all study volunteers in the COVID-19 Vaccine in Pregnancy and Lactation 290

(COVIPAL) cohort study from December 2020 to June 2021. Eligible participants were 291

actively pregnant or lactating, planning to receive any COVID-19 vaccine, and willing to 292

donate blood and/or milk samples. 293

Clinical data collection. Clinical data on vaccine side effects were collected through an 294

online questionnaire that was sent to participants 21 days or more after each vaccine 295

administration. Questionnaires were distributed using REDCap. 296

Sample collection. Maternal blood and milk samples were collected at three time points: 297

1) up to 1 day before the 1st dose (pre-vaccine); 2) on the day of and prior to 298

administration of the 2nd dose (after 1st dose); and between 4-10 weeks after the 2nd 299

dose (after 2nd dose). In some cases, additional milk samples were collected up to 31 300

days before the 1st dose, 24 hours after each dose, and weekly for up to 4 weeks after 301

the 2nd dose. Infant blood was collected by heel stick by trained study staff at 5-15 302

weeks after 2nd maternal vaccination. 303

Milk processing. Fresh human milk samples were self-collected by participants into 304

sterile containers at several time points before, during, and after vaccination. Milk 305

samples were either collected immediately by the study staff or frozen by mothers in 306

their home freezer as soon as possible after pumping. Samples were kept on ice during 307

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 38: Clinical manifestations, risk factors, and maternal and

transport from home to the lab for processing. Milk was aliquoted and stored in -80°C 308

until analyzed. 309

Measurement of SARS-CoV-2 specific IgM and IgG in plasma samples. Whole blood 310

was collected into tubes containing EDTA. Plasma was isolated from whole blood by 311

centrifugation and immediately cryopreserved at -80°C until analysis. Anti-SARS-CoV-2 312

plasma IgM and IgG antibodies were measured using the Pylon 3D automated 313

immunoassay system(29) (ET Healthcare, Palo Alto, CA). In brief, quartz glass probes 314

pre-coated with either affinity-purified goat anti-human IgM (IgM capture) or Protein G 315

(IgG capture) were dipped into diluted plasma samples, washed, and then dipped into 316

the assay reagent containing both biotinylated, recombinant spike protein receptor 317

binding domain (RBD) and nucleocapsid protein (NP). After washing, the probes were 318

incubated with Cy®5-streptavidin (Cy5-SA) polysaccharide conjugate reagent, allowing 319

for cyclic amplification of the fluorescence signal. The background-corrected signal of 320

SARS-CoV-2 specific antibodies was reported as relative fluorescent units (RFU). IgM 321

and IgG measurements greater than 50 RFU were considered positive RFUs. 322

Measurement of IgA and IgG by ELISA assay in milk. After thawing, milk fat was 323

separated by cold centrifugation (10,000g for 10 min, 4°C). Milk supernatant samples 324

were diluted 1:2 in sample diluent buffer and were plated in duplicate on a 96-well plate 325

containing S1 spike protein RBD (Ray-Biotech, GA, USA, IEQ-CoVS1RBD-IgG-1 and 326

IEQ-CoVS1RBD-IgA-1). For IgA assays, samples were also plated in duplicate on a 327

second 96-well plate coated with human albumin to account for non-specific binding. 328

OD values for albumin were subtracted from the OD values for RBD. Each plate 329

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 39: Clinical manifestations, risk factors, and maternal and

contained seven anti-RBD standards and one blank negative control. The mean 330

absorbance of each sample was captured on an ELISA plate reader at 450 nm. 331

Background values (blank negative control) were subtracted from the albumin and RBD 332

plates. Standard controls were used to create a standard curve and determine the level 333

of anti-RBD IgA and IgG in unit/ml. 334

Measurement of Polyethylene Glycol (PEG) in human milk by ELISA. Milk supernatant 335

was diluted 1:8 with the provided sample buffer and analyzed by PEGylated protein 336

ELISA kit (Enzo, Farmingdale, NY, USA). Seven wells of each plate were loaded with 337

serial dilutions (1:2) of mRNA-1273 or BNT162b2 to generate the standard curve for 338

each vaccine (Figure 1A). In addition, vaccines were inoculated into human milk 339

samples at three different concentrations (33µl/ml, 3.3µl/ml and 0.33µl/ml) and were 340

analyzed separately to ensure the ability of the kit to detect the vaccine PEGylated 341

components in milk samples (Figure 1B). Prism 9 (v 9.1.2) was used to interpolate 342

vaccine concentration in the samples based on OD values, using a sigmoidal, four 343

parameters logistic curve. A mean of pre-vaccine milk PEG concentration was 344

calculated based on 13 pre-vaccine samples analyzed. Results are presented as a ratio 345

of sample concentration divided by the mean pre-vaccine milk PEG concentration. 346

Statistics: All data analyses were conducted using Stata statistical software (v14, 347

College Station, TX). Descriptive statistics included frequencies for categorical 348

variables, and means, standard deviations, medians, and ranges for continuous 349

variables. Group differences in categorical variables were analyzed using Fisher’s exact 350

test, and group differences in continuous variables were analyzed using Mann-Whiney 351

U tests. McNemar tests were used to evaluate differences in symptom frequencies after 352

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 40: Clinical manifestations, risk factors, and maternal and

each vaccine dose. Spearman correlations was used to assess the magnitude of 353

associations between continuous variables. Non-parametric tests were used to 354

accommodate non-normal distributions and small group sizes. 355

356

357

358

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 41: Clinical manifestations, risk factors, and maternal and

Author contribution: 359

YG designed the study, conducted experiments, acquired data, analyzed data, and was 360

lead author of the manuscript. MP designed the study, conducted experiments, acquired 361

data, analyzed data, assisted with editing and writing the manuscript. AC recruited 362

participants, acquired data, and assisted with editing and writing the manuscript. CG 363

analyzed data, conducted statistical analysis assisted with editing the manuscript. 364

AHBW providing reagents and acquired data. UJ collected samples and conducted 365

experiments. CYL recruited participants and collected samples. VJG, LW, SB, LL 366

collected and process samples. EB collected and processed samples and assisted with 367

editing the manuscript. IVA assisted with study and questionnaires design and editing 368

and writing the manuscript. NA supervised the study and assisted in writing the 369

manuscript. SLG and VJF designed and supervised the study and assisted in writing the 370

manuscript. 371

372

373

374

375

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 42: Clinical manifestations, risk factors, and maternal and

References: 376

1. Labayo HKM et al. Norovirus-specific immunoglobulin A in breast milk for protection 377

against norovirus-associated diarrhea among infants.. EClinicalMedicine 378

2020;27:100561. 379

2. Sadeharju K et al. Maternal antibodies in breast milk protect the child from 380

enterovirus infections. Pediatrics 2007;119(5):941–946. 381

3. Fox A et al. Robust and Specific Secretory IgA Against SARS-CoV-2 Detected in 382

Human Milk[published online ahead of print: 2020]; doi:10.1016/j.isci.2020.101735 383

4. Demers-Mathieu V et al. Difference in levels of SARS-CoV-2 S1 and S2 subunits- 384

and nucleocapsid protein-reactive SIgM/IgM, IgG and SIgA/IgA antibodies in human 385

milk. J. Perinatol. 2020;1–10. 386

5. Perl SH et al. SARS-CoV-2–Specific Antibodies in Breast Milk After COVID-19 387

Vaccination of Breastfeeding Women. JAMA 2021;(19):2013–2014. 388

6. Fox A, Norris C, Amanat F, Zolla-Pazner S, Powell RL. The vaccine-elicited 389

immunoglobulin profile in milk after COVID-19 mRNA-based vaccination is IgG-390

dominant and lacks secretory antibodies. medRxiv 2021;2021.03.22.21253831. 391

7. KELLY JC et al. Anti-SARS-CoV-2 antibodies induced in breast milk after Pfizer-392

BioNTech/BNT162b2 vaccination. Am. J. Obstet. Gynecol. 2021;0(0). 393

doi:10.1016/j.ajog.2021.03.031 394

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 43: Clinical manifestations, risk factors, and maternal and

8. Gray KJ et al. COVID-19 vaccine response in pregnant and lactating women: a 395

cohort study. Am. J. Obstet. Gynecol. 2021;0(0). doi:10.1016/j.ajog.2021.03.023 396

9. Zheng W et al. Microbiota-targeted maternal antibodies protect neonates from enteric 397

infection. Nature 2020;577(7791):543–548. 398

10. Krause PR, Gruber MF. Emergency Use Authorization of Covid Vaccines — Safety 399

and Efficacy Follow-up Considerations. N. Engl. J. Med. 2020;383(19):e107. 400

11. Hall S. COVID vaccines and breastfeeding: what the data say. Nature 401

2021;594(7864):492–494. 402

12. Sellaturay P, Nasser S, Ewan P. Polyethylene Glycol–Induced Systemic Allergic 403

Reactions (Anaphylaxis). J. Allergy Clin. Immunol. Pract. 2021;9(2):670–675. 404

13. Garvey LH, Nasser S. Anaphylaxis to the first COVID-19 vaccine: is polyethylene 405

glycol (PEG) the culprit?. BJA Br. J. Anaesth. 2021;126(3):e106. 406

14. Shimabukuro TT et al. Preliminary Findings of mRNA Covid-19 Vaccine Safety in 407

Pregnant Persons. N. Engl. J. Med. [published online ahead of print: April 21, 2021]; 408

doi:10.1056/nejmoa2104983 409

15. Mattar CN et al. Title page Addressing anti-syncytin antibody levels, and fertility and 410

breastfeeding concerns, following BNT162B2 COVID-19 mRNA vaccination. medRxiv 411

2021;2021.05.23.21257686. 412

16. Golan Y et al. Evaluation of Messenger RNA From COVID-19 BTN162b2 and 413

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 44: Clinical manifestations, risk factors, and maternal and

mRNA-1273 Vaccines in Human Milk. JAMA Pediatr. [published online ahead of print: 414

July 6, 2021]; doi:10.1001/jamapediatrics.2021.1929 415

17. Ming J et al. BNT162b2 vaccination induces SARS-CoV-2 specific antibody 416

secretion into human milk with minimal transfer of vaccine mRNA. medRxiv 417

2021;2021.04.27.21256151. 418

18. STAFFORD IA, PARCHEM JG, SIBAI BM. The COVID-19 vaccine in pregnancy: 419

risks benefits and recommendations. Am. J. Obstet. Gynecol. [published online ahead 420

of print: January 30, 2021]; doi:10.1016/j.ajog.2021.01.022 421

19. Craig AM, Hughes BL, Swamy GK. Coronavirus disease 2019 vaccines in 422

pregnancy. Am. J. Obstet. Gynecol. MFM 2021;3(2):100295. 423

20. Liu X et al. Patterns of IgG and IgM antibody response in COVID-19 patients. 424

Emerg. Microbes Infect. 2020;9(1):1269–1274. 425

21. Polack FP et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. 426

Engl. J. Med. 2020;383(27):2603–2615. 427

22. Dabrera G et al. A case-control study to estimate the effectiveness of maternal 428

pertussis vaccination in protecting newborn infants in England and Wales, 2012-2013. 429

Clin. Infect. Dis. 2015;60(3):333–337. 430

23. Amirthalingam G et al. Effectiveness of maternal pertussis vaccination in England: 431

An observational study. Lancet 2014;384(9953):1521–1528. 432

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 45: Clinical manifestations, risk factors, and maternal and

24. Puck JM, Glezen WP, Frank AL, Six HR. Protection of infants from infection with 433

influenza A virus by transplacentally acquired antibody. J. Infect. Dis. 1980;142(6):844–434

849. 435

25. Wright AL, Holberg CJ, Martinez FD, Morgan WJ, Taussig LM. Breast feeding and 436

lower respiratory tract illness in the first year of life. Group Health Medical Associates.. 437

BMJ 1989;299(6705):946–949. 438

26. Tarrant M, Kwok M, Lam T, Epidemiology GL-, 2010 undefined. Breast-feeding and 439

childhood hospitalizations for infections. JSTOR 440

27. Oddy WH et al. Breast feeding and respiratory morbidity in infancy: A birth cohort 441

study. Arch. Dis. Child. 2003;88(3):224–228. 442

28. Ma C, Cong Y, Zhang H. COVID-19 and the Digestive System. Am. J. 443

Gastroenterol. 2020;115(7):1003–1006. 444

29. Lynch KL et al. Magnitude and Kinetics of Anti-Severe Acute Respiratory Syndrome 445

Coronavirus 2 Antibody Responses and Their Relationship to Disease Severity. Clin. 446

Infect. Dis. 2021;72(2):301–308. 447

448

449

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 46: Clinical manifestations, risk factors, and maternal and

Funding: 450

These studies were supported by the Marino Family Foundation (to M.P), the National 451

Institutes of Health (NIAID K23AI127886 to M.P. and NIAID K08AI141728 to S.L.G.), 452

the Krzyzewski Family (to S.L.G), the Weizmann Institute of Science -National 453

Postdoctoral Award Program for Advancing Women in Science (to Y.G.), the 454

International Society for Research In Human Milk and Lactation (ISRHML) Trainee 455

Bridge Fund (to Y.G.), and of the Human Frontier Science Program (to Y.G.). 456

457

458

459

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 47: Clinical manifestations, risk factors, and maternal and

Figures: 460

461

Figure 1: Detection of vaccine PEG in human milk samples. A) different vaccine 462

concentrations were used to generate a standard curve of OD (Y axis) vs. vaccine 463 concentration (X axis). Vaccine concentrations in each sample were interpolated based 464

on Sigmoidal, four-parameter logistic (4PL) curve. B) mRNA-1273 and BNT-162b2 465 vaccines were inoculated separately from pre-vaccine milk samples and were used to 466

ensure the assay’s sensitivity to detect the vaccine PEG components in milk samples. 467 C) Values shown in heat map are the ratio of calculated PEG concentration in each 468

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 48: Clinical manifestations, risk factors, and maternal and

sample to average PEG concentration in all pre-vaccine milk samples. No significant 469 differences were observed between samples collected at any of the post vaccine (PV) 470

time points and the pre-vaccine samples (paired and unpaired two-tailed t-tests). Y axes 471 represent time of sample collection, as hours (h) or weeks (w) Post vaccine 1 (PV 1), or 472

Post vaccine 2 (PV 2). 473

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 49: Clinical manifestations, risk factors, and maternal and

474

Figure 2: Elevated levels of plasma anti-SARS-CoV2 antibodies in COVID-19 475

mRNA vaccinated lactating individuals. Anti-SARS-CoV2 IgM (A) and IgG (B) levels 476

in plasma of lactating individuals post COVID-19 mRNA vaccine (RFU- relative 477

fluorescent units, positive cut-off >50 RFU). After 1st dose samples were collected on 478

the day of the second vaccine, and after 2nd dose samples were collect 4-10 weeks 479

post 2nd dose. Asterisks represent p-values: *= p-value <0.05, **= p-value <0.01, ***= 480

<0.001, ****= <0.0001 as determined by unpaired Mann-Whitney test. 481

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 50: Clinical manifestations, risk factors, and maternal and

482

Figure 3: Elevated levels of milk anti-SARS-CoV2 IgA antibodies in COVID-19 483 mRNA vaccinated lactating individuals. Milk samples from individuals receiving BNT-484

162b2 (n=27) (A) and mRNA-1273 (n=21) (B) anti- SARS-CoV2 vaccines were 485 analyzed for anti-SARS-CoV2 IgA antibodies using ELISA at various time points as 486

indicated on the X axis. After 1st dose samples were collected on the day of the second 487 vaccine, and after 2nd dose samples were collect 4-10 weeks post 2nd dose. Milk anti-488

SARS-CoV2 IgG levels were measured using ELISA in milk samples from individuals 489 receiving BNT-162b2 (n=27) (C) or mRNA-1273 (n=21) (D). Asterisks represent p-490 values: *= p-value <0.05, **= p-value <0.01, ***= <0.001 as determined by unpaired 491

Mann-Whitney test. 492

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 51: Clinical manifestations, risk factors, and maternal and

493

Figure 4: Correlations between milk antibodies, blood antibodies and infant age. 494

Two-tailed Spearman correlation was used to correlate milk IgA (A) and IgG (B) levels 495

(Y axis) and infant age (X axis) 4-10 weeks after the 2nd dose administration (n=30). In 496

addition, two-tailed Spearman correlation was used to correlate milk IgG (Y axis) and 497

milk IgA levels (X axis) on the day of 2nd dose administration(C), 21-28 days after 1st 498

dose (n=35) and and 4-10 weeks after 2nd dose (D). We also tested correlation 499

between milk (Y axis) and maternal plasma (X axis) IgG levels at day of 2nd dose (E) 500

and 4-10 weeks after the 2nd dose administration (F) (n=30). 501

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 52: Clinical manifestations, risk factors, and maternal and

502

Figure 5: Transfer of anti-SARS-CoV2 IgG from mother to infant. IgG levels were 503

measured in blood samples of infants and mothers 61 days to 1 year postpartum, 61-504

129 days after 1st maternal vaccine administration. Maternal and infant samples were 505

collected in the same week (except in one case in which the maternal sample was 506

collected 18 days prior to the infant sample). X axis represents the time at which the 507

mother received the 1st dose: during pregnancy (pink), or after delivery (red). (RFU- 508

relative fluorescent units, dashed line represents positive cut-off >50 RFU). Sample 509

characteristics and individual antibodies levels are present in Table S4. 510

511

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 53: Clinical manifestations, risk factors, and maternal and

Tables: 512

Table 1. Sample characteristics overall and by vaccine manufacturer 513

Sample Characteristics Full Cohort (n=48, 100%)

BNT162b2 (n=27, 56%)

mRNA-1237 (n=21, 44%)

Maternal characteristics

Maternal age, years

Median (min, max) 35 (27, 46) 35 (30, 45) 35 (27, 46)

Race/ethnicity, % (n)

Asian 31% (15) 30% (8) 33% (7)

Black or African American 2% (1) 4% (1) 0% (0)

White/Caucasian 59% (28) 55% (15) 62% (13)

Other (Arab) 2% (1) 0% (0) 5% (1)

More than 1 race/ethnicity (White+Latina/Asian/Middle Eastern)

6% (3) 11% (3) 0% (0)

Highest level of education completed

Some college 2% (1) 0% (0) 5% (1)

College graduate 17% (8) 19% (5) 14% (3)

Advanced degree 81% (39) 81% (22) 81% (17)

Work in health care?

Yes, providing direct patient care 58% (28) 52% (14) 67% (14)

Yes, but not in direct patient care 19% (9) 15% (4) 24% (5)

No 23% (11) 33% (9) 9% (2)

Pre-Pregnancy Body Mass Index

Median (min, max) 23.4 (19.1, 37.5)

23.4 (19.1, 35.9)

22.8 (19.6, 37.5)

Number of children

1 40% (19) 41% (11) 38% (8)

2 46% (22) 41% (11) 52% (11)

3 12% (6) 15% (4) 10% (2)

4 2% (1) 3% (1) 0% (0)

Duration of most recent pregnancy, weeks

Median (min, max) 39.0 (33.9, 41.1)

39.1 (33.9, 41.0)

39.0 (37.4, 41.1)

Infant characteristics

Infant age at maternal 1st dose, months

Median (min, max) 4.7 (0.1, 17.2)

4.8 (0.2, 15.2)

4.6 (0.1, 17.2)

Sex, % (n)

Male 60% (29) 67% (18) 52% (11)

Female 40% (19) 33% (9) 48% (10)

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 54: Clinical manifestations, risk factors, and maternal and

Exclusively breastfeeding (and no solids)

Yes 23% (11) 22% (6) 24% (5)

No 77% (37) 78% (21) 76% (16)

Days after vaccine that symptoms were assessed

Dose 1

Mean (SD) 78.7 (31.8) 78.3 (35.4) 79.2 (27.4)

Median (min, max) 81 (18, 154] 78 (18, 154) 86 (26, 117)

Dose 2

Mean (SD) 59.6 (25.1) 62.6 (28.3) 55.8 (20.2)

Median (min, max) 58.5 (28, 133)

57 (29, 133) 60 (28, 89)

Note: None of the characteristics above differed significantly by vaccine manufacturer. 514

Abbreviations: Standard deviation (SD), minimum (min), maximum (max) 515

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 55: Clinical manifestations, risk factors, and maternal and

Table 2: Symptoms after each vaccine dose 516

Symptoms Full Cohort: After 1st dose After 2nd dose

1st dose

2nd dose

P-valuea

BNT162b2 mRNA-1237

p-valueb

BNT162b2 mRNA-1237

p-valueb

n=48 n=27 n=21 n=27 n=21

Injection site symptoms, % (n)

Any injection site symptoms

88% (42)

88% (42)

>0.99 78% (21) 100% (21)

0.02 78% (21) 100% (21)

.02

Pain 88% (42)

85% (41)

0.71 78% (21) 100% (21)

0.02 78% (21) 95% (20)

0.12

Redness 4% (2)

10% (5)

0.08 0% (0) 10% (2) 0.19 4% (1) 19% (4) 0.15

Swelling 17% (8)

17% (8)

>0.99 7% (2) 29% (6) 0.12 11% (3) 24% (5) 0.27

Itching 4% (2)

4% (2)

>0.99 4% (1) 5% (1) >.99 4% (1) 5% (1) >.99

Rash around injection site

2% (1)

4% (2)

0.32 0% (0) 5% (1) 0.44 0% (0) 10% (2) 0.19

Generalized symptoms, % (n)

Any general symptoms 48% (23)

92% (44)

<0.001 44% (12) 52% (11)

0.77 85% (23) 100% (21)

0.12

Fever 12% (6)

62% (30)

<0.001 19% (5) 5% (1) 0.21 52% (14) 76% (16) 0.13

Chills 8% (4)

48% (23)

<0.001 11% (3) 5% (1) 0.62 37% (10) 62% (13)

0.14

Headache 21% (10)

67% (32)

<0.001 11% (3) 33% (7) 0.08 56% (15) 81% (17)

0.07

Joint pain 8% (4)

31% (15)

0.002 7% (2) 10% (2) >0.99 30% (8) 33% (7) >0.99

Muscle/body aches 21% (10)

69% (33)

<0.001 30% (8) 10% (2) 0.15 59% (16) 81% (17)

0.13

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 56: Clinical manifestations, risk factors, and maternal and

Fatigue or tiredness 44% (21)

81% (39)

<0.001 41% (11) 48% (10)

0.77 67% (18) 100% (21)

0.003

Nausea 4% (2)

12% (6)

0.10 4% (1) 5% (1) >0.99 7% (2) 19% (4) 0.38

Vomiting 0% (0)

0% (0)

--- 0% (0) 0% (0) --- 0% (0) 0% (0) ----

Diarrhea 4% (2)

4% (2)

>0.99 4% (1) 5% (1) >0.99 0% (0) 10% (2) 0.19

Abdominal pain 2% (1)

0% (0)

0.32 0% (0) 5% (1) 0.44 0% (0) 0% (0) ----

Rash not near injection site

0% (0)

0% (0)

--- 0% (0) 0% (0) --- 0% (0) 0% (0) ----

Lump/swelling in breast (same side as injection)

0% (0)

2% (1)

0.32 0% (0) 0% (0) --- 4% (1) 0% (0) >0.99

Lump/swelling in breast (opposite side as injection)

0% (0)

0% (0)

--- 0% (0) 0% (0) --- 0% (0) 0% (0) ----

Mastitis 2% (1)

0% (0)

0.32 0% (0) 5% (1) 0.44 0% (0) 0% (0) ----

Decrease in milk supply 2% (1)

2% (1)

>0.99 0% (0) 5% (1) 0.44 0% (0) 5% (1) 0.44

a McNemar’s test; b Fisher’s Exact test517

518

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 57: Clinical manifestations, risk factors, and maternal and

35

Table 3. Infant symptoms reported after maternal vaccination (write-in only) 519

INFANT SYMPTOMS % (n) Vaccine

After 1st vaccine dose

None / no changes / blank 88% (42)

“My baby seemed a little tired.” 2% (1) BNT162b2

“He started pooping a lot! And it was more sour smelling diarrhea like poops. I don't know if there is any correlation”

2% (1) BNT162b2

“It could have been a fluke, but both my infant and I slept through the night for the first time the night after I received the 1st dose of the vaccine.”

2% (1) BNT162b2

“He had some diaper rash, but likely unrelated” 2% (1) BNT162b2

“Rash on the face / worsening of baby acne” 2% (1) mRNA-1237

“Disrupted sleep, waking at night when he usually doesn't. More fussy than normal.”

2% (1) mRNA-1237

After 2nd vaccine dose

None / no changes / blank 100% (48)

520

521

522

523

524

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 58: Clinical manifestations, risk factors, and maternal and

36

Table S1. Participants testing positive for SARS-CoV-2 during the study period 525

Participants testing positive for SARS-CoV-2

Participant ID: 1 2 3

Mother positive for SARS-CoV-2 No Yes Yes

Infant positive for SARS-CoV-2 Yes Yes No

Other people in the household diagnosed Yes N/A No

Time of diagnosis 1 week after 2nd dose

N/A 10 day before 1st dose

Baby exclusively breastfed No Yes No

Maternal blood IgG (RFU)

On the day of 2nd dose 244 5503 N/A

4 weeks after 2nd dose 2558 5290 N/A

Infant blood IgG (RFU)

4 weeks after 2nd vaccine dose N/A 1928 N/A

Infant blood IgA (U/ml)

4 weeks after 2nd vaccine dose N/A 122 N/A

Milk anti-RBD IgG levels (U/ml)

Pre-vaccine N/A 55 7.3

On the day of 2nd dose 246 2653 323

4 weeks after 2nd dose 375 2834 250

Maternal blood positive for anti SARS-CoV2-N-protein antibodies

N/A Samples collected on day of 2nd dose and 5 weeks after

2nd dose were positive.

N/A

Maternal injection site symptoms

Reported after 1st vaccine dose: Pain Pain, Itching None

Reported after 2nd vaccine dose: Pain None Pain

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 59: Clinical manifestations, risk factors, and maternal and

37

Maternal generalized symptoms

Reported after 1st vaccine dose: None None None

Reported after 2nd vaccine dose: Fever, Chills, Muscle aches or body aches,

Fatigue or tiredness

Fever, Chills, Fatigue or tiredness

Fatigue or tiredness

Baby symptoms

After 1st dose None None None

after 2nd dose: None Less active. Feverish.

None

526

527

528

529

530

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 60: Clinical manifestations, risk factors, and maternal and

38

Table S2. Correlations between antibody levels and timing of samples in relation to childbirth and vaccine 531

Antibodies and sample types being correlated

N Spearman correlations

rho p

Samples collected after 1st dose

IgG in maternal blood and … 24

Time from childbirth to collection

0.38 0.07

Time from 2nd dose to sample -0.11 0.59

IgG in breast milk and … 35

Time from childbirth to sample -.05 0.75

Time from 2nd dose to sample -0.12 0.47

IgA in breast milk and … 38

Time from childbirth to sample -0.18 0.28

Time from 2nd dose to sample 0.12 0.45

Samples collected after dose 2

IgG in maternal blood and … 32

Time from childbirth to sample 0.01 0.92

Time from 2nd dose to sample -0.34 0.05

IgG in breast milk and … 44

Time from childbirth to sample 0.04 0.75

Time from 2nd dose to sample 0.21 0.16

IgA in breast milk and … 43

Time from childbirth to sample -0.35 0.02

Time from 2nd dose to sample -0.17 0.27

532

533

534

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 61: Clinical manifestations, risk factors, and maternal and

39

Table S3. Antibody levels were not significantly correlated with maternal BMI 535

Correlation between maternal BMI and the following antibody levels:

n

Spearman correlations

rho p

After 1st dose

IgG in maternal blood 24 -.071 .74

IgG in milk 35 -.029 .87

IgA in milk 38 -.204 .22

4-10 weeks after 2nd dose

IgG in maternal blood 32 .113 .54

IgG in milk 44 -.101 .51

IgA in milk 43 .115 .46

536

537

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 62: Clinical manifestations, risk factors, and maternal and

40

Table S4. Follow up maternal and infant blood samples characteristics and anti-SARS-CoV2 IgG levels 538

Participant #

Participant Cohort:

Infant sex

Infant age at

sample date

(months):

Infant age at

1st dose (months)

Time since 1st

dose until

infant sample

collection (days)

Infant anti-SARS-

CoV2 IgG antibodies

(RFU):

Maternal anti-

SARS-CoV2 IgG

antibodies

(RFU):

Vaccine type:

1 Lactating Female 0-3 0-3 84 6 2768 mRNA-1237

2 Lactating Male 0-3 0-3 58 2 1791 mRNA-1237

3 Lactating Female 0-3 0-3 64 0 1638 BNT162b2

4 Lactating Female 3-6 0-3 67 4 5028 mRNA-1237

5 Lactating Female 3-6 0-3 61 6 3449 mRNA-1237

6 Lactating Male 6-9 3-6 81 7 3292 mRNA-1237

7 Lactating Male 9-12 6-9 103 2 1574 mRNA-1237

8 Lactating Male >12 9-12 62 3 3418 BNT162b2

9 Pregnant male 0-3 -2 129 812 271 BNT162b2

10 Pregnant female 0-3 -2 125 1604 288 mRNA-1237

Note: negative values in infant age represent days before delivery. RFU>50 considered as positive value. 539

540

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 63: Clinical manifestations, risk factors, and maternal and

41

541

. CC-BY-NC-ND 4.0 International licenseIt is made available under a is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review)

The copyright holder for this preprint this version posted August 3, 2021. ; https://doi.org/10.1101/2021.03.09.21253241doi: medRxiv preprint

Page 64: Clinical manifestations, risk factors, and maternal and

Menstrual changes after covid-19 vaccinationA link is plausible and should be investigated

Victoria Male lecturer in reproductive immunology

Common side effects of covid-19 vaccination listedby the UK’s Medicines and Healthcare ProductsRegulatoryAgency (MHRA) include a sore arm, fever,fatigue, and myalgia.1 Changes to periods andunexpected vaginal bleeding are not listed, butprimary care clinicians and those working inreproductive health are increasingly approached bypeople who have experienced these events shortlyafter vaccination. More than 30 000 reports of theseevents had been made to MHRA’s yellow cardsurveillance scheme for adverse drug reactions by 2September 2021, across all covid-19 vaccines currentlyoffered.1

Most peoplewho report a change to their period aftervaccination find that it returns to normal thefollowing cycle and, importantly, there is no evidencethat covid-19 vaccination adversely affects fertility.In clinical trials, unintended pregnancies occurredat similar rates in vaccinated and unvaccinatedgroups.2 In assisted reproduction clinics, fertilitymeasures and pregnancy rates are similar invaccinated and unvaccinated patients.3 -6

MHRA states that evaluation of yellow card reportsdoesnot support a linkbetweenchanges tomenstrualperiods and covid-19 vaccines since the number ofreports is low relative to both the number of peoplevaccinated and theprevalence ofmenstrual disordersgenerally.7 However, the way in which yellow carddata are collected makes firm conclusions difficult.Approaches better equipped to compare rates ofmenstrual variation in vaccinated versusunvaccinated populations are needed, and the USNational Institutes ofHealthhasmade$1.67m (£1.2m;€1.4m) available to encourage this importantresearch.8

Menstrual changes have been reported after bothmRNA and adenovirus vectored covid-19 vaccines,1suggesting that, if there is a connection, it is likelyto be a result of the immune response to vaccinationrather than a specific vaccine component.Vaccination against human papillomavirus (HPV)has also been associated with menstrual changes.9Indeed, the menstrual cycle can be affected byimmune activation in response to various stimuli,includingviral infection: inone studyofmenstruatingwomen, around a quarter of those infected withSARS-CoV-2 experienced menstrual disruption.10

Biologically plausible mechanisms linking immunestimulation with menstrual changes includeimmunological influences on the hormones drivingthe menstrual cycle11 or effects mediated by immunecells in the lining of the uterus, which are involvedin the cyclical build-up and breakdown of thistissue.12 Research exploring a possible association

between covid-19 vaccines and menstrual changesmay also help understand the mechanism.

Although reported changes to the menstrual cycleafter vaccination are short lived, robust research intothis possible adverse reaction remains critical to theoverall success of the vaccination programme.Vaccine hesitancy among young women is largelydriven by false claims that covid-19 vaccines couldharm their chances of future pregnancy.13 Failing tothoroughly investigate reports of menstrual changesafter vaccination is likely to fuel these fears. If a linkbetween vaccination and menstrual changes isconfirmed, this information will allow people to planfor potentially altered cycles. Clear and trustedinformation is particularly important for those whorely on being able to predict their menstrual cyclesto either achieve or avoid pregnancy.

We are still awaiting definitive evidence, but in theinterim how should clinicians counsel those whohave experienced these effects? Initially, they shouldbe encouraged to report any changes to periods orunexpected vaginal bleeding to the MHRA’s yellowcard scheme. This will provide more complete datato facilitate research into any link and signal topatients that their concerns about vaccine safety aretaken seriously, building trust. In terms ofmanagement, the Royal College of Obstetricians andGynaecologists and the MHRA recommend thatanyone reporting a change in periods persisting overseveral cycles, or new vaginal bleeding after themenopause, should be managed according to theusual clinical guidelines for these conditions.7 14

One important lesson is that the effects of medicalinterventions on menstruation should not be anafterthought in future research. Clinical trials providethe ideal setting in which to differentiate betweenmenstrual changes caused by interventions fromthose that occur anyway, but participants areunlikelyto report changes toperiodsunless specifically asked.Informationaboutmenstrual cycles andother vaginalbleeding should be actively solicited in future clinicaltrials, including trials of covid-19 vaccines.

Competing interests: The BMJ has judged that there are no disqualifying financialties to commercial companies. The author declares the following other interests:research funding from theWellcome Trust and research charity Borne; paymentsto act as an external examiner for the University of Cambridge and the Universityof Leeds; and royalties received for my contribution to Immunology 9th edition(Elsevier). Further details of The BMJ policy on financial interests is here:https://www.bmj.com/sites/default/files/attachments/resources/2016/03/16-current-bmj-education-coi-form.pdf.

Provenance and peer review: Commissioned; not externally peer reviewed.

1 Medicines and Healthcare Products Regulatory Agency. Coronavirusvaccine—weekly summary of yellow card reporting. 2021.https://www.gov.uk/government/publications/coronavirus-covid-19-vaccine-adverse-reactions/coronavirus-vaccine-summary-of-yellow-card-report-ing#annex-1-vaccine-analysis-print

1the bmj | BMJ ${year};374:n2211 | doi: 10.1136/bmj.n2211

EDITORIALS

Department of Metabolism, Digestionand Reproduction, Imperial CollegeLondon, Chelsea and WestminsterHospital Campus, London, UK

[email protected]

Cite this as: BMJ ${year};374:n2211

http://dx.doi.org/10.1136/bmj.n2211

Published:

on 28 October 2021 at U

ni of Zurich. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.n2211 on 15 Septem

ber 2021. Dow

nloaded from

Page 65: Clinical manifestations, risk factors, and maternal and

2 Male V. Are covid-19 vaccines safe in pregnancy?Nat Rev Immunol 2021;21:200-1.doi: 10.1038/s41577-021-00525-y pmid: 33658707

3 Morris RS. SARS-CoV-2 spike protein seropositivity from vaccination or infection does not causesterility. F S Rep 2021. doi: 10.1016/j.xfre.2021.05.010. pmid: 34095871

4 Orvieto R, Noach-Hirsh M, Segev-Zahav A, Haas J, Nahum R, Aizer A. Does mRNA SARS-CoV-2vaccine influence patients’ performance during IVF-ET cycle?Reprod Biol Endocrinol 2021;19:69.doi: 10.1186/s12958-021-00757-6 pmid: 33985514

5 Bentov Y, Beharier O, Moav-Zafrir A, et al. Ovarian follicular function is not altered by SARS-Cov-2infection or BNT162b2 mRNA Covid-19 vaccination. medRxiv 2021:2021.04.09.21255195.[Preprint.] doi: 10.1101/2021.04.09.21255195

6 Safrai M, Rottenstreich A, Herzberg S, Imbar T, Reubinoff B, Ben-Meir A. Stopping themisinformation: BNT162b2 COVID-19 vaccine has no negative effect onwomen’s fertility. medRxiv2021:2021.05.30.21258079 [Preprint.] doi: 10.1101/2021.05.30.21258079

7 Medicines and Healthcare Products Regulatory Agency. COVID-19 vaccines: updates for August2021. https://www.gov.uk/drug-safety-update/covid-19-vaccines-updates-for-august-2021

8 Eunice Kennedy Shriver National Institute of Child Health and Human Development. Item ofinterest: NIH funds studies to assess potential effects of COVID-19 vaccination on menstruation.2021.https://www.nichd.nih.gov/newsroom/news/083021-COVID-19-vaccination-menstruation

9 Suzuki S, Hosono A. No association between HPV vaccine and reported post-vaccinationsymptoms in Japanese young women: Results of the Nagoya study. Papillomavirus Res2018;5:96-103. doi: 10.1016/j.pvr.2018.02.002 pmid: 29481964

10 Li K, Chen G, Hou H, etal. Analysis of sex hormones and menstruation in COVID-19 women ofchild-bearing age. Reprod Biomed Online 2021;42:260-7.doi: 10.1016/j.rbmo.2020.09.020 pmid: 33288478

11 Karagiannis A, Harsoulis F. Gonadal dysfunction in systemic diseases. Eur J Endocrinol2005;152:501-13. doi: 10.1530/eje.1.01886 pmid: 15817904

12 Monin L, Whettlock EM, Male V. Immune responses in the human female reproductive tract.Immunology 2020;160:106-15. doi: 10.1111/imm.13136 pmid: 31630394

13 Speed B. Young women are the unlikely new face of covid-19 vaccine resistance. i News 2021Jan 6. https://inews.co.uk/news/health/coronavirus-latest-experts-debunk-vaccine-fertility-myths-women-819783

14 Royal College of Obstetricians and Gynaecologists. RCOG responds to reports that COVID-19vaccine affects periods. 2021. https://www.rcog.org.uk/en/news/rcog-responds-to-reports-that-covid-19-vaccine-affects-periods/

This article is made freely available for use in accordance with BMJ's website terms and conditions forthe duration of the covid-19 pandemic or until otherwise determined by BMJ. You may use, downloadand print the article for any lawful, non-commercial purpose (including text and data mining) providedthat all copyright notices and trade marks are retained.

the bmj | BMJ ${year};374:n2211 | doi: 10.1136/bmj.n22112

EDITORIALS

on 28 October 2021 at U

ni of Zurich. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.n2211 on 15 Septem

ber 2021. Dow

nloaded from

Page 66: Clinical manifestations, risk factors, and maternal and

Dow

nloadedfrom

https://journals.lww.com

/greenjournalbyBhD

Mf5ePH

Kav1zEoum1tQ

fN4a+kJLhEZgbsIH

o4XMi0hC

ywCX1AW

nYQp/IlQ

rHD3i3D

0OdR

yi7TvSFl4Cf3VC

1y0abggQZXdgG

j2MwlZLeI=

on09/15/2021

Downloadedfromhttps://journals.lww.com/greenjournalbyBhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdgGj2MwlZLeI=on09/15/2021

Research Letter

Severe Acute Respiratory SyndromeCoronavirus 2 (SARS-CoV-2) Vaccinationin PregnancyMeasures of Immunity and Placental Histopathology

Elisheva D. Shanes, MD, Sebastian Otero, BA, Leena B. Mithal, MD, MSCI, Chiedza A. Mupanomunda, BS,Emily S. Miller, MD, MPH, and Jeffery A. Goldstein, MD, PhD

INTRODUCTION

Vaccines against severe acute respiratory syn-drome coronavirus 2 (SARS-CoV-2) have been

approved for emergency use, but, despite elevated riskof severe disease, pregnant women were excluded fromthe clinical trials that led to their authorization.1 Placen-tal findings can indicate potential clinical risk and couldbe an early signal for rare injury seen only after wide-spread use in the pregnant population.2–6

Maternal SARS-CoV-2 infection has been associ-ated with decidual arteriopathy, fetal vascular mal-perfusion, and chronic histiocytic intervillositis.7–9

mRNA vaccines induce an immune response throughactivation of TLR3, which has been linked to decidualarteriopathy, growth restriction, preterm delivery, andfetal loss in mouse models.10–14

Our objective was to evaluate the frequency ofthese key placental lesions in patients who receivedSARS-CoV-2 vaccination in pregnancy.

METHODS

The study methods have been described previouslyand were approved by the Northwestern Univer-

sity institutional review board.7,15 We reportresults from patients who tested negative forSARS-CoV-2 infection on polymerase chain reac-tion who received vaccine (delivering between Jan-uary and April 2021) and unvaccinated women in acontrol group (negative for SARS-CoV-2 infectionon polymerase chain reaction, immunoglobulin G–

and immunoglobulin M–negative, deliveringbetween April 2020 and April 2021) from anongoing coronavirus disease 2019 (COVID-19)cohort study. Antibody testing used the ACCESSSARS-CoV-2 spike protein RBD test.

Statistical testing was performed with unpaired ttests or Fisher exact test for demographics and logisticregression with gestational age as a covariate for pla-cental lesions (Python SciPy 1.6.1). A post hoc powercalculation was performed, demonstrating at least 80%power to identify a 2.5-fold or higher increased risk ofany lesion with a baseline prevalence of 10% or greaterand a threefold or higher increased risk of any lesionwith a baseline prevalence of 7% or greater (Stata 15.0).

RESULTS

We report findings in 84 women who received aSARS-CoV-2 vaccine during pregnancy and 116women in a control group who did not receive avaccine (Table 1). Women with vaccination weremore likely to deliver vaginally. The first inocula-tion was 46624 days before delivery for the 75patients with known vaccination timing. Vaccinatedwomen showed robust antibody responses, whereaswomen in the control group were negative (Fig. 1and Table 1).

Placental examination in women with vaccinationshowed no increased incidence of decidual arterio-pathy, fetal vascular malperfusion, low-grade chronicvillitis, or chronic histiocytic intervillositis compared

From the Feinberg School of Medicine, Northwestern University, and the Annand Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois.

Supported by the Friends of Prentice (funder) and NIBIB K08EB030120 (toJAG), Stanley Manne Research Institute and K23AI139337 (to LBM).

Each author has confirmed compliance with the journal’s requirements forauthorship.

Published online ahead-of-print May 11, 2021.

Corresponding author: Jeffery A. Goldstein, MD, PhD, Olson 2-455, FeinbergSchool of Medicine, Chicago IL; email: [email protected].

Financial DisclosureThe authors did not report any potential conflicts of interest.

© 2021 by the American College of Obstetricians and Gynecologists. Publishedby Wolters Kluwer Health, Inc. All rights reserved.ISSN: 0029-7844/21

© 2021 by the American College of Obstetriciansand Gynecologists. Published by Wolters Kluwer Health, Inc.

Unauthorized reproduction of this article is prohibited.

VOL. 138, NO. 2, AUGUST 2021 OBSTETRICS & GYNECOLOGY 281

Page 67: Clinical manifestations, risk factors, and maternal and

with women in the control group (Table 1). Incidenceof high-grade chronic villitis was higher in the controlgroup than in the vaccinated group.

DISCUSSION

In our cohort of vaccinated pregnant patients, therewas no observed increase in the incidence of findingscharacteristic of SARS-CoV-2 infection in pregnancyand no evidence of vaccine-triggered breakdown inmaternal immunologic tolerance of the fetus.16

Although limited by population differences betweenvaccinated and unvaccinated patients,17,18 these find-ings add to the growing literature supporting thesafety of SARS-CoV-2 vaccination in pregnancy.

REFERENCES

1. Kucirka LM, Norton A, Sheffield JS. Severity of COVID-19 inpregnancy: a review of current evidence. Am J Reprod Immu-nol 2020;84:e13332. doi: 10.1111/aji.13332.

2. Chisholm KM, Heerema-McKenney A. Fetal thrombotic vas-culopathy: significance in liveborn children using proposedsociety for pediatric pathology diagnostic criteria. Am J SurgPathol 2015;39:274–80. doi: 10.1097/PAS.0000000000000334

3. Ernst LM, Bit-Ivan EN, Miller ES, Minturn L, Bigio EH,Weese-Mayer DE. Stillbirth: correlations between brain injuryand placental pathology. Pediatr Dev Pathol 2016;19:237–43.doi: 10.2350/15-06-1658-OA.1

4. Redline RW, Pappin A. Fetal thrombotic vasculopathy: theclinical significance of extensive avascular villi. Hum Pathol1995;26:80–5. doi: 10.1016/0046-8177(95)90118-3

Table 1. Clinical, Immunologic, and Histologic Findings

VaccineGroup (n584)

ControlGroup (n5116) aOR (95% CI)* P

Clinical characteristicsMaternal age (y) 33.763.1 32.564.8 .071st vaccine-to-delivery interval (d) (n575) 45.9624.3 NAGestational age at delivery (wk) 38.562.4 38.461.9 .86Vaginal delivery 66 (79%) 75(65%) .04Anti–SARS-CoV-2 signal/cutoff ratio

(n552 vaccine group, 116 control group)IgG 22.8614.5 0.0460.05 ,.001IgM 4.1613.2 0.1960.12 .001

Placental findingsDecidual arteriopathy 8 (10) 14 (12) 0.75 (0.3–1.9) .55Fetal vascular malperfusion 5 (6) 8 (7) 0.85 (0.27–2.7) .78Low-grade chronic villitis 10 (12) 9 (8) 1.6 (0.62–4.2) .33High-grade chronic villitis 4 (5) 16 (14) 0.31 (0.1–0.97) .04Chronic histiocytic intervillositis† 0 (0) 2 (1.7) — —

aOR, adjusted odds ratio; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; Ig, immunoglobulin.Data are mean6SD or n (%) unless otherwise specified.* Adjusted for gestational age at delivery.† Multivariate modeling cannot be performed for chronic histiocytic intervillositis given the low incidence.

Fig. 1. Maternal anti–severeacute respiratory syndrome co-ronavirus 2 (SARS-CoV-2) spikeprotein immunoglobulin (Ig)M(A) and IgG (B) at delivery. Vac-cinated patients showed frequent(30/52 over cutoff) IgM androbust (50/52) IgG; women in thecontrol group did not (0/116 and0/116).

Shanes. Placental Pathology in SARS-CoV-2 Vaccination. Obstet Gynecol2021.

© 2021 by the American College of Obstetriciansand Gynecologists. Published by Wolters Kluwer Health, Inc.

Unauthorized reproduction of this article is prohibited.

282 Shanes et al Placental Pathology in SARS-CoV-2 Vaccination OBSTETRICS & GYNECOLOGY

Page 68: Clinical manifestations, risk factors, and maternal and

5. Redline RW. Severe fetal placental vascular lesions in terminfants with neurologic impairment. Am J Obstet Gynecol2005;192:452–7. doi: 10.1016/j.ajog.2004.07.030

6. Redline RW, O’Riordan MA. Placental lesions associated withcerebral palsy and neurologic impairment following term birth.Arch Pathol Lab Med 2000;124:1785–91. doi: 10.1043/0003-9985(2000)124,1785:PLAWCP.2.0.CO;2

7. Shanes ED, Mithal LB, Otero S, Azad HA, Miller ES, GoldsteinJA. Placental pathology in COVID-19. Am J Clin Pathol 2020;154:23–32. doi: 10.1093/ajcp/aqaa089

8. Baergen RN, Heller DS. Placental pathology in covid-19 posi-tive mothers: preliminary findings. Pediatr Dev Pathol 2020;23:177–80. doi: 10.1177/1093526620925569

9. Schwartz DA, Baldewijns M, Benachi A, Bugatti M, CollinsRRJ, De Luca D, et al. Chronic histiocytic intervillositis withtrophoblast necrosis is a risk factor associated with placentalinfection from coronavirus disease 2019 (COVID-19) and intra-uterine maternal-fetal severe acute respiratory syndrome coro-navirus 2 (SARS-CoV-2) transmission in live-born and stillborninfants. Arch Pathol Lab Med 2020;145:517–28. doi: 10.5858/arpa.2020-0771-SA

10. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines—a new era in vaccinology. Nat Rev Drug Discov 2018;17:261–79. doi: 10.1038/nrd.2017.243

11. Zhang J, Wei H, Wu D, Tian Z. Toll-like receptor 3 agonistinduces impairment of uterine vascular remodeling and fetallosses in CBA3DBA/2 mice. J Reprod Immunol 2007;74:61–7. doi: 10.1016/j.jri.2006.10.005

12. Baines KJ, Rampersaud AM, Hillier DM, Jeyarajah MJ,Grafham GK, Eastabrook G, et al. Antiviral inflammationduring early pregnancy reduces placental and fetal growthtrajectories. JI 2020;204:694–706. doi: 10.4049/jimmunol.1900888

13. Koga K, Cardenas I, Aldo P, Abrahams VM, Peng B, Fill S,et al. Original article: activation of TLR3 in the trophoblast isassociated with preterm delivery: activation of TLR3 in tropho-blast. Am J Reprod Immunol 2009;61:196–212. doi: 10.1111/j.1600-0897.2008.00682.x

14. Thaxton JE, Nevers T, Lippe EO, Blois SM, Saito S, Sharma S.NKG2D blockade inhibits poly(I:C)-Triggered fetal loss in wildtype but not in IL-10 2/2 mice. JI 2013;190:3639–47. doi: 10.4049/jimmunol.1203488

15. Mithal LB, Otero S, Shanes ED, Goldstein JA, Miller ES. Cordblood antibodies following maternal COVID-19 vaccinationduring pregnancy. Am J Obstet Gynecol 2021. doi: 10.1016/j.ajog.2021.03.035. [Epub ahead of print]

16. Østergaard SD, Schmidt M, Horváth-Puhó E, Thomsen RW,Sørensen HT. Thromboembolism and the Oxford-AstraZenecaCOVID-19 vaccine: side-effect or coincidence? Lancet 2021;397:1441–3. doi: 10.1016/S0140-6736(21)00762-5

17. Quansah R, Gissler M, Jaakkola JJK. Work as a nurse and amidwife and adverse pregnancy outcomes: a Finnish nation-wide population-based study. J Women’s Health 2009;18:2071–6. doi: 10.1089/jwh.2008.1062

18. Park C, Kang MY, Kim D, Park J, Eom H, Kim EA. Adversepregnancy outcomes in healthcare workers: a Korean nation-wide population-based study. Int Arch Occup Environ Health2017;90:501–6. doi: 10.1007/s00420-017-1213-3

(Obstet Gynecol 2021;138:281–283)DOI: 10.1097/AOG.0000000000004457

PEER REVIEW HISTORYReceived April 10, 2021. Received in revised form April 26, 2021.Accepted April 29, 2021. Peer reviews and author correspondenceare available at http://links.lww.com/AOG/C331.

© 2021 by the American College of Obstetriciansand Gynecologists. Published by Wolters Kluwer Health, Inc.

Unauthorized reproduction of this article is prohibited.

VOL. 138, NO. 2, AUGUST 2021 Shanes et al Placental Pathology in SARS-CoV-2 Vaccination 283

Page 69: Clinical manifestations, risk factors, and maternal and

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

n engl j med nejm.org 1

The authors’ affiliations are listed in the Appendix. Address reprint requests to Dr. Shimabukuro at the Immunization Safety Office, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Dis-eases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA 30329, or at tshimabukuro@ cdc . gov.

*The members of the CDC v-safe COVID-19 Pregnancy Registry Team are listed in the Supplementary Appendix, available at NEJM.org.

This article was published on April 21, 2021, at NEJM.org.

DOI: 10.1056/NEJMoa2104983Copyright © 2021 Massachusetts Medical Society.

BACKGROUNDMany pregnant persons in the United States are receiving messenger RNA (mRNA) coronavirus disease 2019 (Covid-19) vaccines, but data are limited on their safety in pregnancy.

METHODSFrom December 14, 2020, to February 28, 2021, we used data from the “v-safe after vaccination health checker” surveillance system, the v-safe pregnancy registry, and the Vaccine Adverse Event Reporting System (VAERS) to characterize the initial safety of mRNA Covid-19 vaccines in pregnant persons.

RESULTSA total of 35,691 v-safe participants 16 to 54 years of age identified as pregnant. Injection-site pain was reported more frequently among pregnant persons than among nonpregnant women, whereas headache, myalgia, chills, and fever were reported less frequently. Among 3958 participants enrolled in the v-safe preg-nancy registry, 827 had a completed pregnancy, of which 115 (13.9%) resulted in a pregnancy loss and 712 (86.1%) resulted in a live birth (mostly among partici-pants with vaccination in the third trimester). Adverse neonatal outcomes included preterm birth (in 9.4%) and small size for gestational age (in 3.2%); no neonatal deaths were reported. Although not directly comparable, calculated proportions of adverse pregnancy and neonatal outcomes in persons vaccinated against Covid-19 who had a completed pregnancy were similar to incidences reported in studies involving pregnant women that were conducted before the Covid-19 pandemic. Among 221 pregnancy-related adverse events reported to the VAERS, the most frequently reported event was spontaneous abortion (46 cases).

CONCLUSIONSPreliminary findings did not show obvious safety signals among pregnant persons who received mRNA Covid-19 vaccines. However, more longitudinal follow-up, including follow-up of large numbers of women vaccinated earlier in pregnancy, is necessary to inform maternal, pregnancy, and infant outcomes.

A BS TR AC T

Preliminary Findings of mRNA Covid-19 Vaccine Safety in Pregnant Persons

Tom T. Shimabukuro, M.D., Shin Y. Kim, M.P.H., Tanya R. Myers, Ph.D., Pedro L. Moro, M.D., Titilope Oduyebo, M.D., Lakshmi Panagiotakopoulos, M.D.,

Paige L. Marquez, M.S.P.H., Christine K. Olson, M.D., Ruiling Liu, Ph.D., Karen T. Chang, Ph.D., Sascha R. Ellington, Ph.D., Veronica K. Burkel, M.P.H.,

Ashley N. Smoots, M.P.H., Caitlin J. Green, M.P.H., Charles Licata, Ph.D., Bicheng C. Zhang, M.S., Meghna Alimchandani, M.D., Adamma Mba-Jonas, M.D., Stacey W. Martin, M.S., Julianne M. Gee, M.P.H., and Dana M. Meaney-Delman, M.D.,

for the CDC v-safe COVID-19 Pregnancy Registry Team*

Original Article

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 70: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 2

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

The first coronavirus disease 2019 (Covid-19) vaccines available in the United States were messenger RNA (mRNA) vac-

cines: BNT162b2 (Pfizer–BioNTech) and mRNA-1273 (Moderna). In December 2020, the vaccines were granted Emergency Use Authorization (EUA) by the Food and Drug Administration (FDA) as a two-dose series, 3 weeks apart for Pfizer–BioNTech and 1 month apart for Moderna, and were rec-ommended for use by the Advisory Committee on Immunization Practices (ACIP).1-4 Pregnant persons were excluded from preauthorization clinical trials, and only limited human data on safety during pregnancy were available at the time of authorization. However, pregnant persons with Covid-19 are at increased risk for severe ill-ness (e.g., resulting in admission to an intensive care unit, extracorporeal membrane oxygenation, or mechanical ventilation) and death, as compared with nonpregnant persons of reproductive age.5 Furthermore, pregnant persons with Covid-19 might be at increased risk for adverse pregnancy outcomes, such as preterm birth, as compared with pregnant persons without Covid-19.6 The Centers for Disease Control and Prevention (CDC) and ACIP, in collaboration with the American College of Obstetricians and Gynecologists and the American Academy of Pediatrics, have issued guidance indicating that Covid-19 vaccines should not be withheld from pregnant persons.7-9

Postauthorization monitoring in pregnant per-sons is necessary to characterize the safety of these new Covid-19 vaccines, which use mRNA, lipid nanoparticles, and state-of-the-art manu-facturing processes. Furthermore, establishing their safety profiles is critical to inform recom-mendations on maternal vaccination against Covid-19. We report preliminary findings of mRNA Covid-19 vaccine safety in pregnant per-sons from three U.S. vaccine safety monitoring systems: the “v-safe after vaccination health checker” surveillance system,10 the v-safe preg-nancy registry,11 and the Vaccine Adverse Event Reporting System (VAERS).12

Me thods

Monitoring Systems and Covered PopulationsV-safe Surveillance System and Pregnancy Registry

V-safe is a new CDC smartphone-based active-surveillance system developed for the Covid-19 vaccination program; enrollment is voluntary.

V-safe sends text messages to participants with weblinks to online surveys that assess for ad-verse reactions and health status during a post-vaccination follow-up period. Follow-up contin-ues 12 months after the final dose of a Covid-19 vaccine. During the first week after vaccination with any dose of a Covid-19 vaccine, participants are prompted to report local and systemic signs and symptoms during daily surveys and rank them as mild, moderate, or severe; surveys at all time points assess for events of adverse health effects. If participants indicate that they required medical care at any time point, they are asked to complete a report to the VAERS through active telephone outreach.

To identify persons who received one or both Covid-19 vaccine doses while pregnant or who be-came pregnant after Covid-19 vaccination, v-safe surveys include pregnancy questions for persons who do not report their sex as male. Persons who identify as pregnant are then contacted by tele-phone and, if they meet inclusion criteria, are offered enrollment in the v-safe pregnancy reg-istry. Eligible persons are those who received vaccination during pregnancy or in the pericon-ception period (30 days before the last men-strual period through 14 days after) and are 18 years of age or older. For persons who choose to enroll, the pregnancy registry telephone-based survey collects detailed information about the participant, including medical and obstetric his-tory, pregnancy complications, birth outcomes, and contact information for obstetric and pedi-atric health care providers to obtain medical records; infants are followed through the first 3 months of life. Details about v-safe and v-safe pregnancy registry methods have been published previously.10,11

VAERSThe VAERS is a national spontaneous-reporting (passive-surveillance) system established in 1990 that is administered by the CDC and the FDA.12 Anyone can submit a report to the VAERS. Health care providers are required to report cer-tain adverse events after vaccination, including pregnancy-related complications resulting in hos-pitalization and congenital anomalies, under the conditions of the EUAs for Covid-19 vaccines1,2; the CDC encourages reporting of any clinically significant maternal and infant adverse events. Signs and symptoms of adverse events are coded with the use of the Medical Dictionary for Regulatory

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 71: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 3

mRNA Covid-19 Vaccine Safety in Pregnant Persons

Activities (MedDRA), version 23.1.13 We used a pregnancy-status question in the VAERS form and a MedDRA code and text-string search to identify reports involving vaccination in pregnant persons.14

Outcomes

V-safe outcomes included participant-reported lo-cal and systemic reactogenicity to the BNT162b2 (Pfizer–BioNTech) vaccine and the mRNA-1273 (Moderna) vaccine on the day after vaccination among all pregnant persons 16 to 54 years of age and among nonpregnant women 16 to 54 years of age as a comparator. For analysis of pregnancy outcomes in the v-safe pregnancy registry, data were restricted to completed pregnancies (i.e., live-born infant, spontaneous abortion, induced abortion, or stillbirth). Participant-reported preg-nancy outcomes included pregnancy loss (spon-taneous abortion and stillbirth) and neonatal outcomes (preterm birth, congenital anomalies, small size for gestational age, and neonatal death) (Table S1 in the Supplementary Appen-dix, available with the full text of this article at NEJM.org). In the VAERS, outcomes included non–pregnancy-specific adverse events and preg-nancy- and neonatal-specific adverse events.

Statistical Analysis

Demographic information and pregnancy charac-teristics are described for both v-safe and VAERS participants. Descriptive analyses were performed with the use of v-safe survey data for persons who identified as pregnant through February 28, 2021 (35,691 persons); persons enrolled in the v-safe pregnancy registry who were vaccinated through February 28, 2021 (3958 persons); and VAERS reports involving pregnant women re-ceived through February 28, 2021 (221 persons). Local and systemic reactogenicity was compared between persons who identified as pregnant and nonpregnant women. Descriptive analyses were conducted with the use of SAS software, version 9.4 (SAS Institute). All activities were reviewed by the CDC and were conducted in accordance with applicable federal law and CDC policy.

R esult s

V-safe Surveillance: Local and Systemic Reactogenicity in Pregnant Persons

From December 14, 2020, to February 28, 2021, a total of 35,691 v-safe participants identified as

pregnant. Age distributions were similar among the participants who received the Pfizer–BioNTech vaccine and those who received the Moderna vac-cine, with the majority of the participants being 25 to 34 years of age (61.9% and 60.6% for each vaccine, respectively) and non-Hispanic White (76.2% and 75.4%, respectively); most partici-pants (85.8% and 87.4%, respectively) reported being pregnant at the time of vaccination (Ta-ble 1). Solicited reports of injection-site pain, fatigue, headache, and myalgia were the most frequent local and systemic reactions after either dose for both vaccines (Table 2) and were re-ported more frequently after dose 2 for both vaccines. Participant-measured temperature at or above 38°C was reported by less than 1% of the participants on day 1 after dose 1 and by 8.0% after dose 2 for both vaccines.

These patterns of reporting, with respect to both most frequently reported solicited reactions and the higher reporting of reactogenicity after dose 2, were similar to patterns observed among nonpregnant women (Fig. 1). Small differences in reporting frequency between pregnant persons and nonpregnant women were observed for spe-cific reactions (injection-site pain was reported more frequently among pregnant persons, and other systemic reactions were reported more frequently among nonpregnant women), but the overall reactogenicity profile was similar. Preg-nant persons did not report having severe reac-tions more frequently than nonpregnant women, except for nausea and vomiting, which were re-ported slightly more frequently only after dose 2 (Table S3).

V-safe Pregnancy Registry: Pregnancy Outcomes and Neonatal Outcomes

As of March 30, 2021, the v-safe pregnancy reg-istry call center attempted to contact 5230 per-sons who were vaccinated through February 28, 2021, and who identified during a v-safe survey as pregnant at or shortly after Covid-19 vaccina-tion. Of these, 912 were unreachable, 86 de-clined to participate, and 274 did not meet inclu-sion criteria (e.g., were never pregnant, were pregnant but received vaccination more than 30 days before the last menstrual period, or did not provide enough information to determine eligi-bility). The registry enrolled 3958 participants with vaccination from December 14, 2020, to February 28, 2021, of whom 3719 (94.0%) identi-fied as health care personnel. Among enrolled

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 72: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 4

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

participants, most were 25 to 44 years of age (98.8%), non-Hispanic White (79.0%), and, at the time of interview, did not report a Covid-19 diag-nosis during pregnancy (97.6%) (Table 3). Re-ceipt of a first dose of vaccine meeting registry-eligibility criteria was reported by 92 participants (2.3%) during the periconception period, by 1132 (28.6%) in the first trimester of pregnancy, by 1714 (43.3%) in the second trimester, and by 1019 (25.7%) in the third trimester (1 partici-pant was missing information to determine the timing of vaccination) (Table 3). Among 1040 participants (91.9%) who received a vaccine in the first trimester and 1700 (99.2%) who re-ceived a vaccine in the second trimester, initial

data had been collected and follow-up scheduled at designated time points approximately 10 to 12 weeks apart; limited follow-up calls had been made at the time of this analysis.

Among 827 participants who had a completed pregnancy, the pregnancy resulted in a live birth in 712 (86.1%), in a spontaneous abortion in 104 (12.6%), in stillbirth in 1 (0.1%), and in other outcomes (induced abortion and ectopic preg-nancy) in 10 (1.2%). A total of 96 of 104 sponta-neous abortions (92.3%) occurred before 13 weeks of gestation (Table 4), and 700 of 712 pregnan-cies that resulted in a live birth (98.3%) were among persons who received their first eligible vaccine dose in the third trimester. Adverse out-

Table 1. Characteristics of Persons Who Identified as Pregnant in the V-safe Surveillance System and Received an mRNA Covid-19 Vaccine.*

CharacteristicPfizer–BioNTech

VaccineModerna Vaccine Total

number (percent)

Total 19,252 (53.9) 16,439 (46.1) 35,691 (100)

Age at first vaccine dose

16–19 yr 23 (0.1) 36 (0.2) 59 (0.2)

20–24 yr 469 (2.4) 525 (3.2) 994 (2.8)

25–34 yr 11,913 (61.9) 9,960 (60.6) 21,873 (61.3)

35–44 yr 6,002 (31.2) 5,011 (30.5) 11,013 (30.9)

45–54 yr 845 (4.4) 907 (5.5) 1,752 (4.9)

Pregnancy status

Pregnant at time of vaccination 16,522 (85.8) 14,365 (87.4) 30,887 (86.5)

Positive pregnancy test after vaccination 2,730 (14.2) 2,074 (12.6) 4,804 (13.5)

Race and ethnic group†

Participants with available data 14,320 13,232 27,552

Non-Hispanic White 10,915 (76.2) 9,982 (75.4) 20,897 (75.8)

Hispanic 1,289 (9.0) 1,364 (10.3) 2,653 (9.6)

Non-Hispanic Asian 972 (6.8) 762 (5.8) 1,734 (6.3)

Non-Hispanic Black 371 (2.6) 338 (2.6) 709 (2.6)

Non-Hispanic multiple races 315 (2.2) 292 (2.2) 607 (2.2)

Non-Hispanic other race 76 (0.5) 56 (0.4) 132 (0.5)

Non-Hispanic American Indian or Alaska Native 40 (0.3) 54 (0.4) 94 (0.3)

Non-Hispanic Native Hawaiian or other Pacific Islander 33 (0.2) 31 (0.2) 64 (0.2)

Unknown race or unknown ethnic group 309 (2.2) 353 (2.7) 662 (2.4)

* Shown are the characteristics of v-safe participants 16 to 54 years of age who identified as pregnant and who received a messenger RNA (mRNA) coronavirus disease 2019 (Covid-19) vaccine — BNT162b2 (Pfizer–BioNTech) or mRNA-1273 (Moderna) — from December 14, 2020, to February 28, 2021. Percentages may not total 100 because of rounding.

† Race and ethnic group were reported by the participants. Questions about race and ethnic group were added to v-safe af-ter launch of the platform; not all pregnancies had recorded race and ethnic group at the time of data analysis. Therefore, data on race and ethnic group were missing for 22.8% of the total number of participants who identified as pregnant (4932 participants receiving the Pfizer–BioNTech vaccine and 3207 receiving the Moderna vaccine).

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 73: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 5

mRNA Covid-19 Vaccine Safety in Pregnant Persons

comes among 724 live-born infants — including 12 sets of multiple gestation — were preterm birth (60 of 636 among those vaccinated before 37 weeks [9.4%]), small size for gestational age (23 of 724 [3.2%]), and major congenital anoma-lies (16 of 724 [2.2%]); no neonatal deaths were reported at the time of interview. Among the participants with completed pregnancies who reported congenital anomalies, none had re-ceived Covid-19 vaccine in the first trimester or periconception period, and no specific pattern of congenital anomalies was observed. Calculated proportions of pregnancy and neonatal outcomes appeared similar to incidences published in the peer-reviewed literature (Table 4).

Adverse-Event Findings on the VAERS

During the analysis period, the VAERS received and processed 221 reports involving Covid-19 vaccination among pregnant persons; 155 (70.1%) involved nonpregnancy-specific adverse events, and 66 (29.9%) involved pregnancy- or neonatal-specific adverse events (Table S4). The most frequently reported pregnancy-related adverse

events were spontaneous abortion (46 cases; 37 in the first trimester, 2 in the second trimester, and 7 in which the trimester was unknown or not reported), followed by stillbirth, premature rupture of membranes, and vaginal bleeding, with 3 reports for each. No congenital anoma-lies were reported to the VAERS, a requirement under the EUAs.

Discussion

This U.S. surveillance review of the safety of mRNA Covid-19 vaccines during pregnancy and the periconception period indicates that some pregnant persons in the United States are choos-ing to be vaccinated against Covid-19 in all tri-mesters of pregnancy. Solicited local and sys-temic reactions that were reported to the v-safe surveillance system were similar among persons who identified as pregnant and nonpregnant women. Although not directly comparable, the proportions of adverse pregnancy and neonatal outcomes (e.g., fetal loss, preterm birth, small size for gestational age, congenital anomalies,

Table 2. Frequency of Local and Systemic Reactions Reported on the Day after mRNA Covid-19 Vaccination in Pregnant Persons.*

Reported Reaction Pfizer–BioNTech Vaccine Moderna Vaccine Total

Dose 1 (N = 9052)

Dose 2 (N = 6638)

Dose 1 (N = 7930)

Dose 2 (N = 5635)

Dose 1 (N = 16,982)

Dose 2 (N = 12,273)

number (percent)

Injection-site pain 7602 (84.0) 5886 (88.7) 7360 (92.8) 5388 (95.6) 14,962 (88.1) 11,274 (91.9)

Fatigue 2406 (26.6) 4231 (63.7) 2616 (33.0) 4541 (80.6) 5,022 (29.6) 8,772 (71.5)

Headache 1497 (16.5) 3138 (47.3) 1581 (19.9) 3662 (65.0) 3,078 (18.1) 6,800 (55.4)

Myalgia 795 (8.8) 2916 (43.9) 1167 (14.7) 3722 (66.1) 1,962 (11.6) 6,638 (54.1)

Chills 254 (2.8) 1747 (26.3) 442 (5.6) 2755 (48.9) 696 (4.1) 4,502 (36.7)

Fever or felt feverish 256 (2.8) 1648 (24.8) 453 (5.7) 2594 (46.0) 709 (4.2) 4,242 (34.6)

Measured temperature ≥38°C 30 (0.3) 315 (4.7) 62 (0.8) 664 (11.8) 92 (0.5) 979 (8.0)

Nausea 492 (5.4) 1356 (20.4) 638 (8.0) 1909 (33.9) 1,130 (6.7) 3,265 (26.6)

Joint pain 209 (2.3) 1267 (19.1) 342 (4.3) 1871 (33.2) 551 (3.2) 3,138 (25.6)

Injection-site swelling 318 (3.5) 411 (6.2) 739 (9.3) 1051 (18.7) 1,057 (6.2) 1,462 (11.9)

Abdominal pain 117 (1.3) 316 (4.8) 160 (2.0) 401 (7.1) 277 (1.6) 717 (5.8)

Injection-site redness 160 (1.8) 169 (2.5) 348 (4.4) 491 (8.7) 508 (3.0) 660 (5.4)

Diarrhea 178 (2.0) 277 (4.2) 189 (2.4) 332 (5.9) 367 (2.2) 609 (5.0)

Vomiting 82 (0.9) 201 (3.0) 77 (1.0) 357 (6.3) 159 (0.9) 558 (4.5)

Injection-site itching 103 (1.1) 109 (1.6) 157 (2.0) 193 (3.4) 260 (1.5) 302 (2.5)

Rash 20 (0.2) 18 (0.3) 22 (0.3) 18 (0.3) 42 (0.2) 36 (0.3)

* Shown are solicited reactions in v-safe participants 16 to 54 years of age who identified as pregnant and who received an mRNA Covid-19 vaccine (BNT162b2 [Pfizer–BioNTech] or mRNA-1273 [Moderna]) from December 14, 2020, to February 28, 2021.

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 74: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 6

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

and neonatal death) among participants with completed pregnancies from the v-safe preg-nancy registry appear to be similar to the pub-lished incidences in pregnant populations stud-ied before the Covid-19 pandemic.15-26 Many participants in the v-safe pregnancy registry were included in the phase 1a (highest) priority group for Covid-19 vaccination owing to their work as health care personnel.27 V-safe participation is voluntary, and registration information is not uniformly available at all vaccination locations, although information about the surveillance sys-tem is included on the EUA fact sheets for health care providers and patients. Thus, comparisons of the proportions of vaccinated women with these outcomes to previously published estimates are limited by likely differences between these populations in age, ethnic group, and other so-cial, demographic, and clinical characteristics that are known to be associated with pregnancy and neonatal outcomes. However, such compari-sons are helpful to provide a crude sense of whether there are any unexpected safety signals in these early data. At the time of this analysis, just 14.7% of persons who identified as pregnant

in the v-safe surveillance system had been con-tacted to offer enrollment in the pregnancy registry.

Other limitations should also be noted. As with all participant-reported surveillance systems, mistakes in completion of v-safe health surveys can result in misclassification of participants as pregnant; as a result, data for local and sys-temic reactions that participants reported to the v-safe platform may include some reports from nonpregnant persons. Participants are not re-quired to complete surveys at the same time every day, and our ability to assess onset or dura-tion of adverse events, such as fever, is limited. The registry data are preliminary, are from a small sample, and describe mostly neonatal out-comes from third-trimester vaccination; the find-ings may change as additional pregnancy out-comes are reported and the sample size increases, which may facilitate detection of rare outcomes. We were unable to evaluate adverse outcomes that might occur in association with exposures earlier in pregnancy, such as congenital anoma-lies, because no pregnant persons who were vac-cinated early in pregnancy have had live births

Figure 1. Most Frequent Local and Systemic Reactions Reported in the V-safe Surveillance System on the Day after mRNA Covid-19 Vaccination.

Shown are solicited reactions in pregnant persons and nonpregnant women 16 to 54 years of age who received a messenger RNA (mRNA) coronavirus disease 2019 (Covid-19) vaccine — BNT162b2 (Pfizer–BioNTech) or mRNA-1273 (Moderna) — from December 14, 2020, to February 28, 2021. The percentage of respondents was calculated among those who completed a day 1 survey, with the top events shown of injection-site pain (pain), fatigue or tiredness (fatigue), headache, muscle or body aches (myalgia), chills, and fever or felt feverish (fever).

Pregnant Nonpregnant

A Pfizer–BioNTech Vaccine, Dose 1 B Pfizer–BioNTech Vaccine, Dose 2

C Moderna Vaccine, Dose 1 D Moderna Vaccine, Dose 2

Perc

enta

ge

100

8090

7060

4030

10

50

20

0Pain Fatigue Headache Myalgia Chills Fever

Perc

enta

ge

100

8090

7060

4030

10

50

20

0

Perc

enta

ge

100

8090

7060

4030

10

50

20

0Pain Fatigue Headache Myalgia Chills Fever

Perc

enta

ge

Pain Fatigue Headache Myalgia Chills Fever

Pain Fatigue Headache Myalgia Chills Fever

100

8090

7060

4030

10

50

20

0

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 75: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 7

mRNA Covid-19 Vaccine Safety in Pregnant Persons

captured in the v-safe pregnancy registry to date; follow-up is ongoing. In addition, the proportion of pregnant persons who reported spontaneous abortion may not reflect true postvaccination proportions because participants might have been vaccinated after the period of greatest risk in the first trimester, and very early pregnancy losses might not be recognized. Whereas some pregnancies with vaccination in the first and early second trimester have been completed, the majority are ongoing, and a direct comparison

of outcomes on the basis of timing of vaccina-tion is needed to define the proportion of spon-taneous abortions in this cohort. Because of sample-size constraints, both pregnancy and neo-natal outcomes were calculated as a proportion instead of a rate.

Our preliminary analysis uses participant-reported data and has limited information on other potential risk factors for adverse pregnancy and neonatal outcomes. The VAERS is subject to the limitations of passive surveillance.12 Despite

Table 3. Characteristics of V-safe Pregnancy Registry Participants.*

CharacteristicPfizer–BioNTech

VaccineModerna Vaccine Total

number (percent)

Total 2136 (54.0) 1822 (46.0) 3958 (100)

Age at first vaccine dose†

20–24 yr 17 (0.8) 19 (1.0) 36 (0.9)

25–34 yr 1335 (62.5) 1238 (67.9) 2573 (65.0)

35–44 yr 777 (36.4) 560 (30.7) 1337 (33.8)

45–54 yr 7 (0.3) 5 (0.3) 12 (0.3)

Race and ethnic group‡

Non-Hispanic White 1663 (77.9) 1463 (80.3) 3126 (79.0)

Hispanic 164 (7.7) 151 (8.3) 315 (8.0)

Non-Hispanic Asian 225 (10.5) 138 (7.6) 363 (9.2)

Non-Hispanic Black 24 (1.1) 26 (1.4) 50 (1.3)

Non-Hispanic multiple races 42 (2.0) 30 (1.6) 72 (1.8)

Non-Hispanic American Indian or Alaskan Native 5 (0.2) 1 (0.1) 6 (0.2)

Non-Hispanic Native Hawaiian or other Pacific Islander 6 (0.3) 3 (0.2) 9 (0.2)

Missing data or participant declined to answer 7 (0.3) 10 (0.5) 17 (0.4)

Timing of first eligible dose

Periconception: within 30 days before last menstrual period 55 (2.6) 37 (2.0) 92 (2.3)

First trimester: <14 wk 615 (28.8) 517 (28.4) 1132 (28.6)

Second trimester: ≥14 and <28 wk 932 (43.6) 782 (42.9) 1714 (43.3)

Third trimester: ≥28 wk 533 (25.0) 486 (26.7) 1019 (25.7)

Missing data 1 (<0.1) 0 1 (<0.1)

Covid-19 infection during pregnancy

No Covid-19 infection 2084 (97.6) 1779 (97.6) 3863 (97.6)

Before vaccination 32 (1.5) 24 (1.3) 56 (1.4)

≤14 days after first eligible dose of vaccination 3 (0.1) 7 (0.4) 10 (0.3)

>14 days after first eligible dose of vaccination 9 (0.4) 3 (0.2) 12 (0.3)

Missing data 8 (0.4) 9 (0.5) 17 (0.4)

* Shown are registry participants who received an mRNA Covid-19 vaccine (BNT162b2 [Pfizer–BioNTech] or mRNA-1273 [Moderna]) from December 14, 2020, to February 28, 2021. Percentages may not total 100 because of rounding.

† The v-safe pregnancy registry is only enrolling pregnant persons 18 years of age or older; at the time of this analysis, no participants were younger than 20 years of age.

‡ Race and ethnic group were reported by the participants.

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 76: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 8

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

EUA mandatory reporting requirements and CDC guidance on VAERS reporting, there is probably substantial underreporting of pregnancy- and neonatal-specific adverse events. We also do not know the total number of Covid-19 vaccine doses administered to pregnant persons, which further limits our ability to estimate rates of reported adverse events from VAERS data. Among pregnancy-specific conditions reported to the VAERS after Covid-19 vaccination, miscarriage was the most common. This is similar to what was observed during the influenza A (H1N1) pandemic in 2009 after the introduction of the 2009 H1N1 inactivated influenza vaccine, where miscarriage was the most common adverse event reported by pregnant persons who received that vaccine.28

In addition to vaccination protecting women against Covid-19 and its complications during pregnancy, emerging evidence has shown trans-placental transfer of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibod-ies after maternal Covid-19 vaccination during

the third trimester, which suggests that mater-nal vaccination might provide some level of pro-tection to the neonate.29-32 However, we do not have data on antibody transfer and level of pro-tection relative to the timing of vaccination. The CDC and the FDA are continuing to monitor and disseminate information about the safety of mRNA and additional types of Covid-19 vaccines in pregnant persons.

Early data from the v-safe surveillance sys-tem, the v-safe pregnancy registry, and the VAERS do not indicate any obvious safety signals with respect to pregnancy or neonatal outcomes associated with Covid-19 vaccination in the third trimester of pregnancy. Continued monitoring is needed to further assess maternal, pregnancy, neonatal, and childhood outcomes associated with maternal Covid-19 vaccination, including in earlier stages of pregnancy and during the pre-conception period. Meanwhile, the present data can help inform decision making about vaccina-tion by pregnant persons and their health care providers.

Table 4. Pregnancy Loss and Neonatal Outcomes in Published Studies and V-safe Pregnancy Registry Participants.

Participant-Reported Outcome Published Incidence* V-safe Pregnancy Registry†

% no./total no. (%)

Pregnancy loss among participants with a completed pregnancy

Spontaneous abortion: <20 wk15-17 10–26 104/827 (12.6)‡

Stillbirth: ≥ 20 wk18-20 <1 1/725 (0.1)§

Neonatal outcome among live-born infants

Preterm birth: <37 wk21,22 8–15 60/636 (9.4)¶

Small size for gestational age23,24‖ 3.5 23/724 (3.2)

Congenital anomalies25** 3 16/724 (2.2)

Neonatal death26†† <1 0/724

* The populations from which these rates are derived are not matched to the current study population for age, race and ethnic group, or other demographic and clinical factors.

† Data on pregnancy loss are based on 827 participants in the v-safe pregnancy registry who received an mRNA Covid-19 vaccine (BNT162b2 [Pfizer–BioNTech] or mRNA-1273 [Moderna]) from December 14, 2020, to February 28, 2021, and who reported a completed pregnancy. A total of 700 participants (84.6%) received their first eligible dose in the third trimester. Data on neonatal outcomes are based on 724 live-born infants, including 12 sets of multiples.

‡ A total of 96 of 104 spontaneous abortions (92.3%) occurred before 13 weeks of gestation.§ The denominator includes live-born infants and stillbirths.¶ The denominator includes only participants vaccinated before 37 weeks of gestation.‖ Small size for gestational age indicates a birthweight below the 10th percentile for gestational age and infant sex ac-

cording to INTERGROWTH-21st growth standards (http://intergrowth21 . ndog . ox . ac . uk). These standards draw from an international sample including both low-income and high-income countries but exclude children with coexisting conditions and malnutrition. They can be used as a standard for healthy children growing under optimal conditions.

** Values include only major congenital anomalies in accordance with the Metropolitan Atlanta Congenital Defects Pro-gram 6-Digit Code Defect List (www . cdc . gov/ ncbddd/ birthdefects/ macdp . html); all pregnancies with major congeni-tal anomalies were exposed to Covid-19 vaccines only in the third trimester of pregnancy (i.e., well after the period of organogenesis).

†† Neonatal death indicates death within the first 28 days after delivery.

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 77: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 9

mRNA Covid-19 Vaccine Safety in Pregnant Persons

The findings and conclusions in this article are those of the authors and do not necessarily represent the official position of the Centers for Disease Control and Prevention (CDC) or the Food and Drug Administration (FDA). Mention of a product or company name is for identification purposes only and does not constitute endorsement by the CDC or the FDA. All authors are U.S. government employees or U.S. government contractors and do not have any material conflicts of interest. Oracle provided in-kind technical support to build and maintain the v-safe after

vaccination health checker infrastructure for data capture and messaging to participants.

Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.

A data sharing statement provided by the authors is available with the full text of this article at NEJM.org.

We thank the v-safe participants, the members of the Oracle v-safe development team for their contributions, and the mem-bers of the CDC COVID-19 Response Team for their support.

AppendixThe authors’ affiliations are as follows: the Immunization Safety Office, Division of Healthcare Quality Promotion (T.T.S., T.R.M., P.L. Moro, L.P., P.L. Marquez, C.K.O., C.L., B.C.Z., J.M.G.), and the Arboviral Diseases Branch, Division of Vector-Borne Diseases (S.W.M.), National Center for Emerging and Zoonotic Infectious Diseases, the Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities (S.Y.K., V.K.B., C.J.G., D.M.M.-D.), the Division of Reproductive Health, National Center for Chronic Disease Prevention and Health Promotion (T.O., K.T.C., S.R.E., A.N.S.), the World Trade Center Health Program, Na-tional Institute for Occupational Safety and Health (R.L.), and the Epidemic Intelligence Service (K.T.C.) — all at the Centers for Disease Control and Prevention, Atlanta; and the Division of Epidemiology, Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD (M.A., A.M.-J.).

References1. Food and Drug Administration. Fact sheet for healthcare providers administer-ing vaccine (vaccination providers): emer-gency use authorization (EUA) of the Pfizer-BioNTech COVID-19 vaccine to prevent coronavirus disease 2019 (COVID-19). 2021 (https://www . fda . gov/ media/ 144413/ download).2. Food and Drug Administration. Fact sheet for healthcare providers administer-ing vaccine (vaccination providers): emer-gency use authorization (EUA) of the Moderna COVID-19 vaccine to prevent coronavirus disease 2019 (COVID-19). 2021 (https://www . fda . gov/ media/ 144637/ download).3. Oliver SE, Gargano JW, Marin M, et al. The Advisory Committee on Immuniza-tion Practices’ interim recommendation for use of Pfizer-BioNTech COVID-19 vac-cine — United States, December 2020. MMWR Morb Mortal Wkly Rep 2020; 69: 1922-4.4. Oliver SE, Gargano JW, Marin M, et al. The Advisory Committee on Immuniza-tion Practices’ interim recommendation for use of Moderna COVID-19 vaccine — United States, December 2020. MMWR Morb Mortal Wkly Rep 2021; 69: 1653-6.5. Zambrano LD, Ellington S, Strid P, et al. Update: characteristics of symptomatic women of reproductive age with laboratory-confirmed SARS-CoV-2 infection by preg-nancy status — United States, January 22–October 3, 2020. MMWR Morb Mortal Wkly Rep 2020; 69: 1641-7.6. Allotey J, Stallings E, Bonet M, et al. Clinical manifestations, risk factors, and maternal and perinatal outcomes of coro-navirus disease 2019 in pregnancy: living systematic review and meta-analysis. BMJ 2020; 370: m3320.7. Centers for Disease Control and Pre-vention. COVID-19 vaccines: interim clin-ical considerations for use of COVID-19 vaccines currently authorized in the United

States. 2021 (https://www . cdc . gov/ vaccines/ covid - 19/ info - by - product/ clinical - considerations . html).8. American College of Obstetricians and Gynecologists. Vaccinating pregnant and lactating patients against COVID-19: practice advisory. December 2020 (https://www . acog . org/ clinical/ clinical - guidance/ practice - advisory/ articles/ 2020/ 12/ vaccinating - pregnant - and - lactating - patients - against - covid - 19).9. American Academy of Pediatrics. In-terim guidance for COVID-19 vaccination in children and adolescents. 2021 (https://services . aap . org/ en/ pages/ 2019 - novel - coronavirus - covid - 19 - infections/ clinical - guidance/ interim - guidance - for - covid - 19 - vaccination - in - children - and - adolescents/ ).10. Centers for Disease Control and Pre-vention. V-safe active surveillance for COVID-19 vaccine safety. January 2021 (https://www . cdc . gov/ vaccinesafety/ pdf/ V - safe - Protocol - 508 . pdf).11. Centers for Disease Control and Pre-vention. V-safe pregnancy surveillance (amendment). (https://www . cdc . gov/ vaccinesafety/ pdf/ vsafe - pregnancy - surveillance - protocol - 508 . pdf).12. Shimabukuro TT, Nguyen M, Martin D, DeStefano F. Safety monitoring in the Vaccine Adverse Event Reporting System (VAERS). Vaccine 2015; 33: 4398-405.13. Medical Dictionary for Regulatory Ac-tivities (MedDRA) home page. (https://www . meddra . org/ ).14. Centers for Disease Control and Pre-vention. Vaccine Adverse Event Reporting System (VAERS): standard operating pro-cedures for COVID-19. January 2021 (https://www . cdc . gov/ vaccinesafety/ pdf/ VAERS - v2 - SOP . pdf).15. Dugas C, Slane VH. Miscarriage. In: StatPearls. Treasure Island, FL: StatPearls Publishing, 2021 (https://www . ncbi . nlm . nih . gov/ books/ NBK532992/ ).16. American College of Obstetricians and

Gynecologists’ Committee on Practice Bulletins — Gynecology. ACOG practice bulletin no. 200: early pregnancy loss. Obstet Gynecol 2018; 132(5): e197-e207.17. Practice Committee of the American Society for Reproductive Medicine. Evalu-ation and treatment of recurrent preg-nancy loss: a committee opinion. Fertil Steril 2012; 98: 1103-11.18. Hoyert DL, Gregory ECW. Cause-of-death data from the fetal death file, 2015-2017. Natl Vital Stat Rep 2020; 69(4): 1-20.19. MacDorman MF, Gregory ECW. Fetal and perinatal mortality: United States, 2013. Natl Vital Stat Rep 2015; 64(8): 1-24.20. Panagiotakopoulos L, McCarthy NL, Tepper NK, et al. Evaluating the associa-tion of stillbirths after maternal vaccina-tion in the Vaccine Safety Datalink. Ob-stet Gynecol 2020; 136: 1086-94.21. Ferré C, Callaghan W, Olson C, Shar-ma A, Barfield W. Effects of maternal age and age-specific preterm birth rates on overall preterm birth rates — United States, 2007 and 2014. MMWR Morb Mor-tal Wkly Rep 2016; 65: 1181-4.22. Centers for Disease Control and Pre-vention. Percentage of births born pre-term by state (https://www . cdc . gov/ nchs/ pressroom/ sosmap/ preterm_births/ preterm . htm).23. Boghossian NS, Geraci M, Edwards EM, Horbar JD. Morbidity and mortality in small for gestational age infants at 22 to 29 weeks’ gestation. Pediatrics 2018; 141(2): e20172533.24. Francis A, Hugh O, Gardosi J. Custom-ized vs INTERGROWTH-21st standards for the assessment of birthweight and stillbirth risk at term. Am J Obstet Gyne-col 2018; 218: Suppl: S692-S699.25. Update on overall prevalence of major birth defects — Atlanta, Georgia, 1978–2005. MMWR Morb Mortal Wkly Rep 2008; 57: 1-5.26. Centers for Disease Control and Pre-

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 78: Clinical manifestations, risk factors, and maternal and

n engl j med nejm.org 10

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

vention. Infant mortality rates by state (https://www . cdc . gov/ nchs/ pressroom/ sosmap/ infant_mortality_rates/ infant_mortality . htm).27. Dooling K, McClung N, Chamberland M, et al. The Advisory Committee on Im-munization Practices’ interim recommen-dation for allocating initial supplies of COVID-19 vaccine — United States, 2020. MMWR Morb Mortal Wkly Rep 2020; 69: 1857-9.28. Moro PL, Broder K, Zheteyeva Y, et al. Adverse events following administration to pregnant women of influenza A (H1N1)

2009 monovalent vaccine reported to the Vaccine Adverse Event Reporting System. Am J Obstet Gynecol 2011; 205(5): 473.e1-473.e9.29. Rottenstreich A, Zarbiv G, Oiknine-Djian E, Zigron R, Wolf DG, Porat S. Ef-ficient maternofetal transplacental trans-fer of anti-SARS-CoV-2 spike antibodies after antenatal SARS-CoV-2 BNT162b2 mRNA vaccination. March 12, 2021 (https://www . medrxiv . org/ content/ 10 . 1101/ 2021 . 03 . 11 . 21253352v1). preprint.30. Gray KJ, Bordt EA, Atyeo C, et al. COVID-19 vaccine response in pregnant

and lactating women: a cohort study. Am J Obstet Gynecol 2021 March 24 (Epub ahead of print).31. Paul G, Chad R. Newborn antibodies to SARS-CoV-2 detected in cord blood af-ter maternal vaccination — a case report. BMC Pediatr 2021; 21: 138.32. Gill L, Jones CW. Severe acute respi-ratory syndrome coronavirus 2 (SARS-CoV-2) antibodies in neonatal cord blood after vaccination in pregnancy. Obstet Gynecol 2021 March 8 (Epub ahead of print).Copyright © 2021 Massachusetts Medical Society.

The New England Journal of Medicine Downloaded from nejm.org on April 25, 2021. For personal use only. No other uses without permission.

Copyright © 2021 Massachusetts Medical Society. All rights reserved.

Page 79: Clinical manifestations, risk factors, and maternal and

Journal Pre-proof

Pregnancy and birth outcomes after SARS-CoV-2 vaccination inpregnancy

Regan N. Theiler , Myra Wick , Ramila Mehta , Amy L. Weaver ,Abinash Virk , Melanie Swift

PII: S2589-9333(21)00162-2DOI: https://doi.org/10.1016/j.ajogmf.2021.100467Reference: AJOGMF 100467

To appear in: American Journal of Obstetrics & Gynecology MFM

Received date: 10 June 2021Revised date: 16 August 2021Accepted date: 16 August 2021

Please cite this article as: Regan N. Theiler , Myra Wick , Ramila Mehta , Amy L. Weaver ,Abinash Virk , Melanie Swift , Pregnancy and birth outcomes after SARS-CoV-2 vacci-nation in pregnancy, American Journal of Obstetrics & Gynecology MFM (2021), doi:https://doi.org/10.1016/j.ajogmf.2021.100467

This is a PDF file of an article that has undergone enhancements after acceptance, such as the additionof a cover page and metadata, and formatting for readability, but it is not yet the definitive version ofrecord. This version will undergo additional copyediting, typesetting and review before it is publishedin its final form, but we are providing this version to give early visibility of the article. Please note that,during the production process, errors may be discovered which could affect the content, and all legaldisclaimers that apply to the journal pertain.

© 2021 Published by Elsevier Inc.

Page 80: Clinical manifestations, risk factors, and maternal and

Pregnancy and birth outcomes after SARS-CoV-2 vaccination

in pregnancy

Regan N. Theiler, MD, PhD*1, Myra Wick, MD, PhD1, Ramila Mehta3, Amy L. Weaver, MS3,

Abinash Virk, MD2, Melanie Swift, MD5

1 Mayo Clinic Department of Obstetrics and Gynecology, Division of Obstetrics

2 Mayo Clinic Department of Medicine, Division of Infectious Disease

3 Mayo Clinic Department of Quantitative Health Sciences

5 Mayo Clinic Division of Preventive, Occupational, and Aerospace Medicine

*Corresponding author:

200 First St SE

Department of Obstetrics and Gynecology

Mayo Clinic

Rochester, MN 55905

[email protected]

Disclosures:

Dr. Theiler has a know-how license and research funding from HeraMED

Dr. Swift has funding from Pfizer via Duke University for the HERO Together vaccine safety

registry.

Dr. Virk reports being an inventor for Mayo Clinic Travel App interaction with Smart Medical Kit

and Medical Kit for Pilgrims.

The other authors have declared no conflicts of interest

Acknowledgments:

This work was funded in part by grant FP00085005-A1-03-S14 from the U.S. National Center for

Translational Sciences. The funder did not participate in the study design, data collection, data

analysis, or writing for publication.

Page 81: Clinical manifestations, risk factors, and maternal and

Condensation:

COVID-19 vaccine administration during pregnancy did not result in adverse pregnancy

outcomes and was associated with a decrease in COVID-19 infections during pregnancy.

Short Title:

Clinical outcomes after COVID-19 vaccination in pregnancy

AJOG at a Glance:

Why was this study conducted?

Limited data exists supporting safety and efficacy of the COVID-19 vaccines for administration

during pregnancy.

What are the key findings?

Sociodemographic differences in COVID-19 vaccine uptake were observed among pregnant

patients. Vaccinated patients did not experience more adverse pregnancy outcomes than non-

vaccinated patients, but they were less likely to experience COVID-19 infection during

pregnancy.

What does this study add to what is already known?

The study demonstrates both safety and efficacy of the vaccines during pregnancy.

KEYWORDS:

PREGNANCY

COVID-19

SARS-C0V-2

VACCINATION

IMMUNITY

mRNA vaccine

Birth

Page 82: Clinical manifestations, risk factors, and maternal and

Adverse outcomes index (AOI)

Gestation

Teratogenicity

Page 83: Clinical manifestations, risk factors, and maternal and

ABSTRACT

Background: SARS-CoV-2 infection during pregnancy is associated with significant maternal

morbidity and increased rates of preterm birth. For this reason, COVID-19 vaccine

administration in pregnancy has been endorsed by multiple professional societies including

ACOG and SMFM despite exclusion of pregnant women from initial clinical trials of vaccine

safety and efficacy. However, to date little data exists regarding outcomes after COVID-19

vaccination of pregnant patients.

Study Design: A comprehensive vaccine registry was combined with a delivery database for an

integrated healthcare system to create a delivery cohort including vaccinated patients. Maternal

sociodemographic data were examined to identify factors associated with COVID-19

vaccination. Pregnancy and birth outcomes were analyzed, including a composite measure of

maternal and neonatal pregnancy complications, the Adverse Outcome Index.

Results: Of 2002 patients in the delivery cohort, 140 (7.0%) received a COVID-19 vaccination

during pregnancy and 212 (10.6%) experienced a COVID-19 infection during pregnancy. The

median gestational age at first vaccination was 32 weeks (range 13 6/7-40 4/7), and patients

vaccinated during pregnancy were less likely than unvaccinated patients to experience COVID-

19 infection prior to delivery (1.4% (2/140) vs. 11.3% (210/1862), P<0.001). No maternal

COVID-19 infections occurred after vaccination during pregnancy.

Factors significantly associated with increased likelihood of vaccination in a multivariable logistic

regression model included older age, higher level of maternal education, being a non-smoker,

use of infertility treatment for the current pregnancy, and lower gravidity. No significant

difference in the composite adverse outcome (5.0% (7/140) vs. 4.9% (91/1862), P=0.95) or

Page 84: Clinical manifestations, risk factors, and maternal and

other maternal or neonatal complications, including thromboembolic events and preterm birth,

was observed in vaccinated compared to unvaccinated patients.

Conclusions: Vaccinated pregnant women in this birth cohort were less likely to experience

COVID-19 infection compared to unvaccinated pregnant patients, and COVID-19 vaccination

during pregnancy was not associated with increased pregnancy or delivery complications. The

cohort was skewed toward late pregnancy vaccination, and thus findings may not be

generalizable to vaccination during early pregnancy.

Page 85: Clinical manifestations, risk factors, and maternal and

Introduction

In late 2020, the United States Food and Drug Administration (FDA) approved two mRNA

vaccines, manufactured by Pfizer-BioNTech (BNT162b2) (Pfizer, Inc; Philadelphia,

Pennsylvania) and Moderna (mRNA-1273) (ModernaTX, Inc; Cambridge, Massachusetts), for

emergency use to prevent COVID-19 illness. Both vaccines were studied in large numbers of

subjects during phase 3 randomized controlled trials, and both were shown to be highly effective

at preventing COVID-19 infection in non-pregnant participants..1, 2 Because none of the trials

undertaken to gain FDA approval included pregnant or lactating women, use of the vaccines

during pregnancy and lactation has been controversial.3 During phase 1A of the vaccine rollout

in the U.S., healthcare workers were the first population with access to vaccination, and thus

many pregnant healthcare workers received the vaccine. During phase 1B, teachers and other

essential workers were vaccinated, adding to the population of reproductive age who were

eligible for vaccination. Starting in December 2020, recommendations from the American

College of Obstetricians and Gynecologists (ACOG), the Society for Maternal-Fetal Medicine

(SMFM), and the World Health Organization (WHO) endorsed availability of COVID-19

vaccination for pregnant women using a shared decision-making model with healthcare

providers3, 4.

COVID-19 disease during pregnancy is known to have severe manifestations in pregnant

women compared to non-pregnant controls, with increased risk for maternal hospitalization, ICU

admission, invasive ventilation, and death 5-7. Other adverse outcomes observed with SARS-

CoV-2 infection during pregnancy include increased rates of preterm birth and increased pre-

eclampsia incidence, both of which have been linked to inflammatory mechanisms 8. Because

of the known increased maternal risk of adverse outcomes with COVID-19 infection and the lack

of proven harm from the available vaccines, many patients have opted for vaccination despite

limited safety and efficacy data for the vaccines in pregnant patients9. The vaccines are thought

Page 86: Clinical manifestations, risk factors, and maternal and

to be effective when administered during pregnancy, as antibody production occurs rapidly after

administration. However, immune alterations that occur in pregnancy may theoretically

decrease the vigor of cell-mediated immune responses to infection. 2, 10 Neutralizing antibody

response is highly reassuring and suggests robust efficacy during pregnancy with possible

benefit to the neonate.

Recently published COVID-19 vaccine surveillance data from the Centers for Disease Control

and Prevention’s (CDC) voluntary V-SAFE registry including 3958 subjects vaccinated during

pregnancy suggests that pregnant women do not have increased rates of adverse vaccine

reactions compared to control patients, and that patients do not report increases in adverse

pregnancy outcomes compared to non-pregnant women11. The V-SAFE data, however, are

limited to patient-reported vaccine reactions and pregnancy events; conclusions are thus

subject to selection bias and lack of validated primary data supporting conclusions. For this

reason, vaccine efficacy should be demonstrated in pregnancy using infectious outcomes as

well. Adding to the available data, we present pregnancy outcomes from a Mayo Clinic Health

System delivery cohort delivering during the first four months of vaccine availability.

Methods

A comprehensive vaccine registry was created, capturing COVID-19 vaccine administrations,

manufacturer, and patient identifying information from Mayo Clinic vaccination sites as well as

other sites across the states of Minnesota and Wisconsin. The vaccination registry was then

linked to the Mayo Clinic delivery registry, which contains detailed maternal and neonatal

outcomes from all births within the Mayo Clinic Health System. The delivery registry data is

derived directly from elements in the electronic medical record and all fields have been validated

manually during development. Creation of the registries and subsequent analysis was

Page 87: Clinical manifestations, risk factors, and maternal and

performed in accordance with human subjects regulations under approval by the Mayo Clinic

Institutional Review Board. These data sources were combined to form a retrospective cohort

of all births during the study timeframe.

Criteria for study inclusion included all patients age 16-55 years old with a delivery event

between December 10, 2020, and April 19, 2021 at a hospital within the Mayo Clinic Health

System. Patients who opted out of use of their medical records for research were excluded from

the study if their delivery occurred in Minnesota. COVID infection during pregnancy, regardless

of temporal relationship to vaccine, was defined as a positive SARS-CoV-2 RT-PCR test

documented in the medical record between the dates of conception and delivery, stratified by

first trimester (2-13 6/7 weeks gestation), second trimester (14 0/7- 27 6/7 weeks gestation) and

third trimester (≥28 weeks gestation) infection.

For purposes of assessing vaccine side effects and pregnancy outcomes, vaccinated

individuals are defined as those receiving any dose of vaccine during pregnancy. For purposes

of assessing vaccine effectiveness, fully vaccinated was defined as >14 days after the final

dose of vaccine.

The composite outcome, the adverse outcome index (AOI) was calculated as a composite of

any of the following events during the delivery hospitalization: a) maternal death during

hospitalization, b) intrapartum neonatal death within 7 days of birth with birthweight ≥ 2500g and

≥37 weeks gestation, c) hypoxic ischemic encephalopathy, d) uterine rupture, e) unplanned

maternal ICU admission, f) return to the operating room within 72 hours of delivery, g)

postpartum hemorrhage with blood transfusion, h) third or fourth degree laceration, i) five

minute Apgar <7 with birthweight ≥2500g and ≥37 weeks gestation, j) admission to the NICU

within 1 day of birth for >1 day with birthweight ≥2500g and ≥37 weeks gestation, or k) neonatal

birth trauma. All qualifying events were verified by chart review. The AOI for a group of patients

was calculated as the number of patients with one or more identified adverse events divided by

Page 88: Clinical manifestations, risk factors, and maternal and

the total number of deliveries, multiped by 100. A woman with multiple gestations was counted

as a single delivery. A modified AOI was also calculated by not considering third- and fourth-

degree perineal laceration as an adverse event. Additional outcomes measured included

thromboembolism or stroke within 4 weeks before or after delivery, gestational hypertensive

disorders diagnosed up to 72 hours after delivery, low and very low birth weight, preterm birth (<

37 weeks gestation), length of postpartum maternal stay after delivery, and stillbirth.

The primary outcome in this study was AOI.12-14 The AOI was 4.9% within the Mayo Clinic

Health System for calendar year 2019. This study was designed with 80% power, using a two-

sided chi-square test with a type I error rate of 0.05, to detect a difference in AOI of 4.9% vs. at

least 11.5% between those without versus with a COVID-19 vaccine during pregnancy, based

on 1862 and 140 patients in the two groups.

Data management and statistical analysis was performed using SAS version 9.4 (SAS institute,

Cary, NC, USA). Comparisons between groups were evaluated using the chi-square test or

Fisher’s exact test for non-ordered categorical variables, the Wilcoxon rank sum test for ordinal

variables, and the two-sample t-test for continuous variables. A multivariable logistic regression

model was fit to identify a set of characteristics independently associated with having received a

COVID-19 vaccine during pregnancy, by including all of the factors identified with p<0.20 based

on univariate analysis. Prior to fitting the multivariable model an additional category was created

for each variable with missing data. A 95% confidence interval (CI) for the difference in the AOI

between two groups was calculated based on exact methods for a binomial parameter. All

calculated p-values were two-sided and p-values less than 0.05 were considered statistically

significant.

Page 89: Clinical manifestations, risk factors, and maternal and

Results

Of 2002 total patients, 140 (7.0%) received at least one dose of a COVID-19 vaccine prior to

delivery, and 212 (10.6%) experienced a COVID-19 infection during pregnancy. Among the

vaccinated patients, one received the Janssen COVID-19 (Ad.26.COV2.S) vaccine (Janssen

Biotech, Inc, a Janssen Pharmaceutical company, Johnson & Johnson; New Brunswick, New

Jersey), 12 received the Moderna vaccine, and 127 received the Pfizer-BioNTech vaccine

(Appendix Table A1). The median estimated gestational age (EGA) at initiation of the

vaccination series was 32 (range 13 6/7-40 4/7) weeks gestation. Completed vaccination was

documented at a median EGA of 35 2/7 weeks (range 17 1/7-44 1/7), with 73.6% of the 140

patients completing vaccination prior to delivery.

Sociodemographic factors (Table 1) positively associated p<0.05) with maternal vaccination

based on univariate analysis included older maternal age at delivery, higher level of maternal

education, and use of infertility treatment for the current pregnancy. Factors negatively

associated (p<0.05) with vaccination included non-white race, Hispanic ethnicity, current

smoking, current illicit drug use, higher gravidity, and higher pre-pregnancy body mass index.

Presence of comorbid conditions including pre-gestational diabetes, chronic hypertension, and

asthma were not significantly associated with vaccination status. The following factors retained

statistical significance in the multivariable logistic regression analysis: older age, higher level of

maternal education, being a non-smoker, use of infertility treatment for the current pregnancy,

and lower gravidity.

Patients vaccinated during pregnancy were less likely than unvaccinated patients to experience

COVID-19 infection prior to delivery (1.4% (2/140) vs. 11.3% (210/1861), P <0.001), with the

two infections occurring in the vaccinated group prior to vaccine administration. In the

unvaccinated group, COVID-19 infections occurred during each trimester of pregnancy, with 26

infections in the first trimester, 84 during the second trimester, and 100 in the third trimester

Page 90: Clinical manifestations, risk factors, and maternal and

(Table 2). The composite pregnancy outcome, AOI, did not differ by maternal vaccination

status, with rates of 5.0% (7/140) vs. 4.9% (91/1862) in the vaccinated and unvaccinated groups

(95% CI for difference in proportions, -3.6% to 3.6%). No maternal or early neonatal deaths

occurred in the cohort. Mode of delivery, gestational age at delivery, neonatal birth weight,

thromboembolic events, and rates of gestational hypertension and pre-eclampsia also did not

significantly differ between groups. Additional comparison between COVID-19 infected, non-

vaccinated patients (n=210) and vaccinated patients without a history of COVID-19 infection

(n=138) did not show any difference among the pregnancy outcomes examined in the cohort,

but the study was not sufficiently powered to detect a difference in these outcomes (Appendix

Table A2). Among the unvaccinated patients, pregnancy and birth outcomes did not

significantly differ between those with (n=210) versus without (n=1652) a COVID-19 infection

during pregnancy (Appendix Table A2).

Discussion

PRINCIPAL FINDINGS: COVID-19 vaccine administration to pregnant patients was associated

with fewer COVID-19 infections during pregnancy and did not result in a detectable increase in

adverse outcomes during pregnancy, delivery, or the delivery hospitalization.

RESULTS: Sociodemographic factors associated with increased vaccination rates in

pregnancy included older age, higher level of maternal education, being a non-smoker, use of

infertility treatment for the current pregnancy, and lower gravidity. No patterns of adverse

maternal or neonatal outcomes were observed in this cohort of 140 pregnant patients who were

vaccinated under the FDA emergency use authorizations for SARS-CoV-2 vaccines, and

significantly fewer vaccinated women experienced COVID-19 infection during the pregnancy

when compared to unvaccinated women.

Page 91: Clinical manifestations, risk factors, and maternal and

CLINICAL IMPLICATIONS: Given the danger of COVID-19 infection during pregnancy and early

studies supporting the safety and effectiveness of vaccination in pregnant patients, specific

efforts should be targeted toward the less vaccinated populations to enhance uptake moving

forward. This study reveals sociodemographic factors associated with vaccine access and/or

uptake in the pregnant population. Vaccination eligibility during the timeframe analyzed was

limited to healthcare workers, the elderly, and teachers or other essential workers. For this

reason, we cannot discern whether sociodemographic differences in vaccination status are due

to vaccine hesitancy or differences in eligibility for vaccination, but we do observe

socioeconomic disparity between those who received vaccination during gestation and those

who did not.

RESEARCH IMPLICATIONS: The current study largely represents outcomes after third

trimester vaccination, but patients who received the vaccine during the late first through second

trimesters and delivered preterm are also represented in this data set. The absence of an

increase in preterm births in the cohort thus suggests that vaccination is unlikely to increase

preterm birth rates, but analysis of outcomes from currently ongoing pregnancies will be needed

to confirm this finding. Additional studies will be needed to examine differences in rare adverse

birth outcomes, as well as outcomes following vaccination during early pregnancy.

STRENGTHS AND LIMITATIONS: Strengths of this study include the inclusion of

comprehensive population-level vaccine registry data in combination with a validated, all-

inclusive delivery database including births at multiple community and teaching hospitals across

two states. As the data were extracted from the primary medical record it is not subject to recall

bias. Given the absence of infection in any vaccinated patients after the first dose administration

(incidence rate ratio of zero and vaccine efficacy of one) with a short observation period, our

ability to estimate vaccine efficacy is limited for this cohort. Other limitations of this analysis

include the small percentage of non-white subjects in this geographic region, the potential for

Page 92: Clinical manifestations, risk factors, and maternal and

confounding due to the observational nature of the study, as well as the data currently available

being biased toward those vaccinated later in gestation and skewed toward the population in the

United States healthcare workforce. Finally, only two COVID-19 infections occurred in the

vaccinated group early in pregnancy, so they may be a group who had lower baseline exposure

compared to the unvaccinated group. Pre-pregnancy COVID-19 infection history was not

available, so previously existing antibody status was unknown and limits the efficacy

conclusions.

CONCLUSIONS: While COVID-19 infection in pregnancy has been associated with significant

adverse maternal and neonatal outcomes in multiple studies, the current findings should give

clinicians confidence that COVID-19 vaccination during pregnancy is protective against

maternal SARS-CoV-2 infection and that no pattern of adverse maternal or birth outcomes is

evident after vaccination during pregnancy. Outreach to under-represented populations of

pregnant patients should be a focus of future education and vaccination efforts.

References

1. BADEN LR, EL SAHLY HM, ESSINK B, et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. New England Journal of Medicine 2021;384:403-16.

2. POLACK FP, THOMAS SJ, KITCHIN N, et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. New England Journal of Medicine 2020;383:2603-15.

3. ADHIKARI EH, SPONG CY. COVID-19 Vaccination in Pregnant and Lactating Women. JAMA 2021;325:1039-40.

4. Vaccinating Pregnant and Lactating Patients Against COVID-19Practice Advisory: American College of Obstetrics and Gynecology, 2021.

5. VILLAR J, ARIFF S, GUNIER RB, et al. Maternal and Neonatal Morbidity and Mortality Among Pregnant Women With and Without COVID-19 Infection: The INTERCOVID Multinational Cohort Study. JAMA pediatrics 2021.

6. ADHIKARI EH, MORENO W, ZOFKIE AC, et al. Pregnancy Outcomes Among Women With and Without Severe Acute Respiratory Syndrome Coronavirus 2 Infection. JAMA network open 2020;3:e2029256-e56.

7. LOKKEN EM, HUEBNER EM, TAYLOR GG, et al. Disease severity, pregnancy outcomes, and maternal deaths among pregnant patients with severe acute respiratory syndrome coronavirus 2 infection in Washington State. American Journal of Obstetrics and Gynecology 2021.

Page 93: Clinical manifestations, risk factors, and maternal and

8. VILLAR J, ARIFF S, GUNIER RB, et al. Maternal and Neonatal Morbidity and Mortality Among Pregnant Women With and Without COVID-19 Infection: The INTERCOVID Multinational Cohort Study. JAMA Pediatr 2021.

9. RASMUSSEN SA, JAMIESON DJ. Pregnancy, Postpartum Care, and COVID-19 Vaccination in 2021. JAMA 2021;325:1099-100.

10. COLLIER A-RY, MCMAHAN K, YU J, et al. Immunogenicity of COVID-19 mRNA Vaccines in Pregnant and Lactating Women. JAMA 2021.

11. SHIMABUKURO TT, KIM SY, MYERS TR, et al. Preliminary Findings of mRNA Covid-19 Vaccine Safety in Pregnant Persons. New England Journal of Medicine 2021:NEJMoa2104983-NEJMoa83.

12. ATALLAH F, BERNSTEIN PS, DIAZ DA, MINKOFF H. The Adverse Outcome Index: Putting Quality into an Outcome Measure. Obstetrics and Gynecology 2018;132:750-53.

13. FOGLIA LM, NIELSEN PE, HEMANN EA, et al. Accuracy of the adverse outcome index: An obstetrical quality measure. Joint Commission Journal on Quality and Patient Safety 2015;41:370-77.

14. TOLCHER MC, TORBENSON VE, WEAVER AL, et al. Impact of a labor and delivery safety bundle on a modified adverse outcomes index Presented at the Central Association of Obstetricians and Gynecologists 82nd annual meeting, Oct. 21-24, 2015, Charleston, SC. American Journal of Obstetrics and Gynecology 2016;214:401.e1-01.e9.

Page 94: Clinical manifestations, risk factors, and maternal and

Table 1. Socio-demographics in Unvaccinated and Vaccinated Patients

Characteristic

COVID Vaccine During Pregnancy Total

(N=2002) P-value†

No (N=1862)

Yes (N=140)

Maternal Age at Delivery (years), Mean (SD)

30.5 (5.2) 31.8 (3.7) 30.1 (5.2) <0.001

Race 0.019

Asian 89 (4.8%) 6 (4.3%) 95 (4.8%)

Black or African American 99 (5.4%) 3 (2.2%) 102 (5.1%)

Others combined 128 (6.9%) 2 (1.4%) 130 (6.6%)

White 1528 (82.9%) 128 (92.1%) 1656 (83.5%)

Unknown/Not Disclosed 18 1 19

Ethnicity 0.022

Hispanic or Latino 173 (9.5%) 5 (3.6%) 178 (9.1%)

Not Hispanic or Latino 1651 (90.5%) 132 (96.4%) 1783 (90.9%)

Unknown/Not Disclosed 38 3 41

Education (years) <0.001

<12 70 (4.7%) 0 (0.0%) 70 (4.3%)

12-16 1218 (81.9%) 70 (53.4%) 1288 (79.6%)

>16 199 (13.4%) 61 (46.6%) 260 (16.1%)

Not documented 375 9 384

Current Smoker 196 (10.5%) 0 (0.0%) 196 (9.8%) <0.001

Illicit Drug Use 56 (3.0%) 0 (0.0%) 56 (2.8%) 0.030

Gravidity 0.007

1 546 (29.3%) 56 (40.0%) 602 (30.1%)

2 519 (27.9%) 34 (24.3%) 553 (27.6%)

3 350 (18.8%) 29 (20.7%) 379 (18.9%)

4+ 447 (24.0%) 21 (15.0%) 468 (23.4%)

Pre-Pregnancy BMI 0.004

<25 573 (39.6%) 70 (56.5%) 643 (41.0%)

25-30 409 (28.3%) 21 (16.9%) 430 (27.4%)

30-35 226 (15.6%) 15 (12.1%) 241 (15.4%)

35-40 139 (9.6%) 14 (11.3%) 153 (9.7%)

40+ 99 (6.8%) 4 (3.2%) 103 (6.6%)

Page 95: Clinical manifestations, risk factors, and maternal and

Not documented 416 16 432

Pre-Gestational Diabetes 11 (0.6%) 2 (1.4%) 13 (0.6%) 0.23

Pre-Gestational Hypertension

64 (3.4%) 6 (4.3%) 70 (3.5%) 0.63

Asthma 206 (11.1%) 15 (10.7%) 221 (11.0%) 0.90

Infertility Treatment 14 (0.8%) 6 (4.3%) 20 (1.0%) 0.002

Multiple Gestation 0.68

Singleton 1840 (98.8%) 138 (98.6%) 1978 (98.8%)

Twins 22 (1.2%) 2 (1.4%) 24 (1.2%)

GBS Test Result 0.51

Negative 1283 (80.8%) 94 (78.3%) 1377 (80.6%)

Positive 305 (19.2%) 26 (21.7%) 331 (19.4%)

Not Tested 274 20 294

Abbreviations: BMI, body mass index; GBS, Group B Streptococcal.

†Comparisons between groups were evaluated using the two-sample t-test for maternal

age, the Wilcoxon rank sum test for maternal education, gravida, and parity, and the

chi-square test or Fisher’s exact test for the remaining categorical

Page 96: Clinical manifestations, risk factors, and maternal and

Table 2. Pregnancy and Birth Outcomes in Unvaccinated and Vaccinated Patients

Outcome

Covid-19 vaccine during pregnancy

Total (N=2002)

P-value†

No (N=1862)

Yes (N=140)

Covid-19 Infection During Pregnancy

0.003

None 1652 (88.7%)

138 (98.6%)

1790 (89.4%)

Trimester 1 26 (1.4%) 0 (0.0%) 26 (1.3%)

Trimester 2 84 (4.5%) 2 (1.4%) 86 (4.3%)

Trimester 3 100 (5.4%) 0 (0.0%) 100 (5.0%)

AOI 91 (4.9%) 7 (5.0%) 98 (4.9%) 0.95

AOI Excluding Laceration 55 (3.0%) 5 (3.6%) 60 (3.0%) 0.61

AOI components

Maternal Death During Hospitalization

0 (0.0%) 0 (0.0%) 0 (0.0%) --

Intrapartum Neonatal Death Within 7 Days of Birth, ≥ 2500 g, ≥37 Weeks Gestation

0 (0.0%) 0 (0.0%) 0 (0.0%) --

Hypoxic, Ischemic Encephalopathy

1 (0.1%) 0 (0.0%) 1 (0.0%) 1.00

Uterine Rupture 1 (0.1%) 0 (0.0%) 1 (0.0%) 1.00

Unplanned Maternal ICU Admission

2 (0.1%) 1 (0.7%) 3 (0.1%) 0.20

Birth Trauma 11 (0.6%) 0 (0.0%) 11 (0.5%) 1.00

Return to Operating Room 6 (0.3%) 1 (0.7%) 7 (0.3%) 0.40

NICU admit within 1 Day of Birth, for > 1 Day, ≥ 2500 g, ≥37 Weeks Gestation

11 (0.6%) 1 (0.7%) 12 (0.6%) 0.58

5-Minute Apgar <7, ≥ 2500 g, ≥37 Weeks Gestation

38 (2.0%) 3 (2.1%) 41 (2.0%) 0.76

Postpartum Hemorrhage With transfusion

5 (0.3%) 1 (0.7%) 6 (0.3%) 0.35

3rd or 4th Degree Laceration

37 (2.0%) 2 (1.4%) 39 (1.9%) 1.00

Mode of Delivery 0.65

Page 97: Clinical manifestations, risk factors, and maternal and

Spontaneous Vaginal 1238 (66.5%)

89 (63.6%)

1327 (66.3%)

Operative Vaginal 69 (3.7%) 7 (5.0%) 76 (3.8%)

Cesarean 555 (29.8%) 44 (31.4%)

599 (29.9%)

Gestational Age at Delivery (weeks)

0.70281

37+ 1703 (91.5%)

127 (90.7%)

1830 (91.4%)

32-36 6/7 134 (7.2%) 10 (7.1%) 144 (7.2%)

24-31 6/7 21 (1.1%) 2 (1.4%) 23 (1.1%)

<24 4 (0.2%) 1 (0.7%) 5 (0.2%)

Length of Stay (days), Median (IQR)‡

2 (1, 2) 2 (1, 2) 2 (1, 2) 0.27

Quantitative Blood Loss > 1000 mL

57 (3.1%) 6 (4.3%) 63 (3.1%) 0.45

Transfusion 241 (12.9%) 25 (17.9%)

266 (13.3%) 0.12

Thromboembolism1 2/1580 (0.1%)

0/129 (0%)

2 (0.1%) 1.00

Stroke1 2/1581 (0.1%)

0/129 (0.0%)

2 (0.0%) 1.00

Eclampsia / Pre-Eclampsia Up to 72 Hours from Delivery

23 (1.2%) 1 (0.7%) 24 (1.2%) 1.00

Gestational Hypertension 225 (12.1%) 19 (13.6%)

244 (12.2%) 0.60

Low Birth Weight (<2500 g) 121 (6.5%) 11 (7.9%) 132 (6.6%) 0.53

Very Low Birth Weight (<1500 g)

21 (1.1%) 3 (2.1%) 24 (1.2%) 0.23

Stillbirth 6 (0.3%) 0 (0.0%) 6 (0.3%) 1.00

Abbreviations: AOI, Adverse Outcome Index; IQR, interquartile range (25th and 75th percentiles)

Page 98: Clinical manifestations, risk factors, and maternal and

1 Thromboembolism and stroke were assessed within 4 weeks before or after delivery. Crude

percentages are reported among the subset who either had the outcome or who had sufficient follow-

up.

†Comparisons between groups were evaluated using the Wilcoxon rank sum test for gestation age and length of stay and the chi-square test or Fisher’s exact test for the categorical variables. ‡ Length of stay from time of delivery to discharge from the hospital.

Page 99: Clinical manifestations, risk factors, and maternal and

COVID-19 vaccination in pregnancy

Journal Pre-proof

COVID-19 vaccination in pregnancy: early experience from a singleinstitution

Megan E. Trostle MD , Meghana A. Limaye MD ,Ms. Valeryia Avtushka , Jennifer L. Lighter MD ,Christina A. Penfield MD MPH , Ashley S. Roman MD MPH

PII: S2589-9333(21)00159-2DOI: https://doi.org/10.1016/j.ajogmf.2021.100464Reference: AJOGMF 100464

To appear in: American Journal of Obstetrics & Gynecology MFM

Received date: 7 May 2021Revised date: 2 August 2021Accepted date: 11 August 2021

Please cite this article as: Megan E. Trostle MD , Meghana A. Limaye MD , Ms. Valeryia Avtushka ,Jennifer L. Lighter MD , Christina A. Penfield MD MPH , Ashley S. Roman MD MPH , COVID-19vaccination in pregnancy: early experience from a single institution, American Journal of Obstetrics& Gynecology MFM (2021), doi: https://doi.org/10.1016/j.ajogmf.2021.100464

This is a PDF file of an article that has undergone enhancements after acceptance, such as the additionof a cover page and metadata, and formatting for readability, but it is not yet the definitive version ofrecord. This version will undergo additional copyediting, typesetting and review before it is publishedin its final form, but we are providing this version to give early visibility of the article. Please note that,during the production process, errors may be discovered which could affect the content, and all legaldisclaimers that apply to the journal pertain.

© 2021 Published by Elsevier Inc.

Page 100: Clinical manifestations, risk factors, and maternal and

COVID-19 vaccination in pregnancy: early experience from a single institution

Short title: COVID-19 vaccination in pregnancy

Megan E. TROSTLE MD,1 Meghana A. LIMAYE MD,

1 Ms. Valeryia AVTUSHKA,

1 Jennifer

L. LIGHTER MD,2 Christina A. PENFIELD MD MPH,

1 Ashley S. ROMAN MD MPH

1

1. Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, New

York University Langone Health, New York, NY

2. Division of Pediatric Infectious Diseases, Department of Infection Prevention and

Control and Pediatrics, New York University Langone Health, New York, NY

Disclosures: The authors report no conflict of interest.

Funding: The authors received no funding for this research.

Corresponding author:

Megan E. Trostle, MD

550 First Avenue

NBV 9N2

New York, NY 10016

[email protected]

Mobile: 201-873-2254

Word count: 723

Page 101: Clinical manifestations, risk factors, and maternal and

Acknowledgments: none

Keywords: COVID-19, infectious disease, maternal morbidity, neonatal health, pregnancy,

public health, vaccination

Objective

Pregnant women are at increased risk for morbidity due to infection with COVID-191.

Vaccination presents an important strategy to mitigate illness in this population. However, there

is a paucity of data on vaccine safety and pregnancy outcomes as pregnant women were

excluded from initial Phase 3 clinical trials. Our objective was to describe maternal, neonatal,

and obstetrical outcomes of women who received an mRNA COVID-19 vaccine during

pregnancy during the first four months of vaccine availability.

Study Design

This was an IRB-approved descriptive study of pregnant women at NYU Langone Health

who received at least one dose of an mRNA COVID-19 vaccine approved by the Food and Drug

Administration (Pfizer/BioNTech or Moderna) from time of FDA Emergency Use Authorization

to April 22, 2021. Eligible women were identified via search of the electronic medical record

(EMR). Vaccine administration was ascertained via immunization records from the New York

State Department of Health. Women were excluded if they were vaccinated prior to conception

or during the postpartum period. Charts were reviewed for maternal demographics and

pregnancy outcomes. Descriptive analyses were performed using R Version 4.0.2 (Boston, MA).

Results

We identified 424 pregnant women who received an mRNA vaccination. 348 (82.1%)

received both doses and 76 (17.9%) received only one dose. Maternal characteristics and

Page 102: Clinical manifestations, risk factors, and maternal and

vaccination information are shown in Table 1. 4.9% of women had a history of confirmed

COVID-19 infection prior to vaccination. After vaccination, no patient in our cohort was

diagnosed with COVID-19 infection. In terms of pregnancy outcomes, 9 had spontaneous

abortions, 3 terminated their pregnancies, and 327 have ongoing pregnancies. 85 delivered

liveborn infants. There were no stillbirths in our population.

Eight of the nine spontaneous abortions occurred in the first trimester at a range of 6-13

weeks gestation. There was one second trimester loss. The rate of spontaneous abortion among

women vaccinated in the first trimester was 6.5%.

The 327 women with ongoing pregnancies have been followed for a median of 4.6 weeks

(range 0-17) following their most recent dose. 113 (34.6%) initiated vaccination in the first

trimester, 178 (54.4%) in the second trimester, and 36 (11.0%) in the third trimester. Following

vaccination, two fetuses (0.6%) developed growth restriction while five (1.5%) were diagnosed

with anomalies.

Outcomes for the 85 women who delivered are shown in Table 2. 18.8% of women were

diagnosed with a hypertensive disorder of pregnancy. The rate of preterm birth was 5.9%. One

preterm delivery was medically indicated while the remaining three were spontaneous. 15.3% of

neonates required admission to the neonatal intensive care unit (NICU). 61.5% of NICU

admissions were due to hypoglycemia or evaluation for sepsis. Other reasons for admission

included prematurity, hypothermia, and transient tachypnea of the newborn. 12.2% of neonates

were small for gestational age (SGA) per World Health Organization standards.

Conclusion

This series describes our experience with women who received an mRNA COVID-19

vaccination while pregnant. In line with other published findings2, we observed no concerning

Page 103: Clinical manifestations, risk factors, and maternal and

trends. There were no stillbirths. Our 6.5% rate of spontaneous abortion is within the expected

rate of 10%3 and our preterm birth rate of 5.9% is below the national average of 9.5%.

4 Our rate

of pregnancy-related hypertensive disorders is higher than our baseline institutional rate of 9.5%,

though this may be due to underlying characteristics of our study population or skewed by small

sample size. Our 12.2% rate of SGA neonates is near the expected value as well, given by

definition 10% of neonates will be SGA at birth. The NICU admission rate is on par with our

institutional rate of 12%. To date, most women in this series have had uncomplicated

pregnancies and have delivered at term.

Strengths of this study include using the EMR to identify subjects and gather data. We

did not rely on self-enrollment and self-report, reducing selection and recall bias. By performing

manual chart review, we obtained detailed and reliable information about individual patients.

One limitation of this study is lack of a matched control group consisting of unvaccinated

pregnant women, so direct conclusions are unable to be drawn about relative risk of

complications. In addition, our cohort is small and may not be generalizable. Finally, many

women included are healthcare workers who had early access to the vaccine.

As more pregnant women become eligible for the COVID-19 vaccine, there is an urgent

need to report on maternal, neonatal, and obstetrical outcomes with COVID-19 vaccine in

pregnancy. The results of this study can be used to counsel and reassure pregnant patients facing

this decision.

Page 104: Clinical manifestations, risk factors, and maternal and

References

1. S Ellington S, Strid P, Tong VT, et al. Characteristics of Women of Reproductive Age with

Laboratory-Confirmed SARS-CoV-2 Infection by Pregnancy Status - United States, January

22-June 7, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(25):769-775. Published 2020 Jun

26.

2. Shimabukuro TT, Kim SY, Myers TR, et al. Preliminary Findings of mRNA Covid-19

Vaccine Safety in Pregnant Persons [published online ahead of print, 2021 Apr 21]. N Engl J

Med. 2021;10.1056/NEJMoa2104983.

3. American College of Obstetricians and Gynecologists' Committee on Practice Bulletins—

Gynecology. ACOG Practice Bulletin No. 200: Early Pregnancy Loss. Obstet Gynecol.

2018;132(5):e197-e207.

4. Ferré C, Callaghan W, Olson C, Sharma A, Barfield W. Effects of Maternal Age and Age-

Specific Preterm Birth Rates on Overall Preterm Birth Rates - United States, 2007 and

2014. MMWR Morb Mortal Wkly Rep. 2016;65(43):1181-1184. Published 2016 Nov 4.

Page 105: Clinical manifestations, risk factors, and maternal and

Table 1: Study population demographics and vaccination characteristics

Study variable Total study population

N=424

Age (years) 35 (6)

Age ≥35 years 220 (51.9)

Race/ethnicity

White

Black

Asian

Hispanic/Latino

Other/not recorded

262 (61.8)

22 (5.2)

57 (13.4)

37 (8.7)

46 (10.8)

Pre-pregnancy body mass index (kg/m2) 23.2 (5.2)

a

Body mass index ≥30 kg/m2 42 (11.3)

a

Pre-pregnancy comorbidities

Chronic hypertension

Pregestational diabetes

Cardiac disease

Respiratory disease

Autoimmune disease

Malignancy (past or present)

28 (6.6)

5 (1.2)

10 (2.4)

38 (9.0)

19 (4.5)

10 (2.4)

Nulliparous 267 (63.0)

Insurance

Private

407 (96.0)

Page 106: Clinical manifestations, risk factors, and maternal and

Public

Unknown/uninsured

16 (3.8)

1 (0.2)

History of COVID infection 21 (4.9)

Vaccine type

Pfizer/BioNTech

Moderna

332 (78.3)

92 (21.7)

Gestational age at first dose (weeks) 21.0 (16.4)

Gestational age at second dose (weeks) 23.9 (17.6)

Trimester at vaccine initiation

First (<14 weeks)

Second (14-27 weeks)

Third (>28 weeks)

124 (29.2)

193 (45.5)

107 (25.2)

Data reported as n (%) or median (IQR)

a. Missing values, n=371

Page 107: Clinical manifestations, risk factors, and maternal and

Table 2: Characteristics and outcomes of delivered women

Study variable Total delivered population

N=85

Vaccine type

Pfizer/BioNTech

Moderna

65 (86.5)

20 (23.5)

Trimester at vaccine initiation

First (<14 weeks)

Second (14-27 weeks)

Third (>28 weeks)

0

14 (16.5)

71 (83.5)

Time from vaccination until delivery (weeks) 2.86 (0.29 - 12.7)

Both vaccine doses completed prior to delivery 68 (80)

Fetal or neonatal demise 0

Gestational age at delivery (weeks) 39.3 (33.0 - 41.7)

Preterm delivery <37 weeks 5 (5.9)

Mode of delivery

Vaginal birth

Cesarean birth

55 (64.7)

30 (35.3)

Obstetrical complications

Pregnancy-related hypertensive disorders

Preterm labor

Preterm prelabor rupture of membranes

Abruption

16 (18.8)

0

2 (2.4)

1 (1.2)

Page 108: Clinical manifestations, risk factors, and maternal and

Placenta previa 1 (1.2)

Neonatal intensive care unit admission 13 (15.3)

Birthweight (grams) 3374 (1910 - 4360)

Small for gestational age 10 (12.2)

Congenital anomalies 2 (1.2)

Data reported as n (%) or median (range)