Menu

An expert resource for medical professionals
Provided FREE as a service to women’s health

The Alliance for
Global Women’s Medicine
A worldwide fellowship of health professionals working together to
promote, advocate for and enhance the Welfare of Women everywhere

An Educational Platform for FIGO

The Global Library of Women’s Medicine
Clinical guidance and resourses

A vast range of expert online resources. A FREE and entirely CHARITABLE site to support women’s healthcare professionals

The Global Academy of Women’s Medicine
Teaching, research and Diplomates Association

This chapter should be cited as follows:
Fell DB, Håberg SE, et al, Glob. libr. women's med.,
ISSN: 1756-2228; DOI 10.3843/GLOWM.418023

The Continuous Textbook of Women’s Medicine SeriesObstetrics Module

Volume 17

Maternal immunization

Volume Editors: Professor Asma Khalil, The Royal College of Obstetricians and Gynaecologists, London, UK; Fetal Medicine Unit, Department of Obstetrics and Gynaecology, St George’s University Hospitals NHS Foundation Trust, London, UK
Professor Flor M Munoz, Baylor College of Medicine, TX, USA
Professor Ajoke Sobanjo-ter Meulen, University of Washington, Seattle, WA, USA

Chapter

Safety of Vaccination During Pregnancy

First published: May 2023

Study Assessment Option

By completing 4 multiple-choice questions (randomly selected) after studying this chapter readers can qualify for Continuing Professional Development awards from FIGO plus a Study Completion Certificate from GLOWM
See end of chapter for details

INTRODUCTION

Assessment of vaccine safety during pregnancy involves several unique considerations, notably, the need to evaluate safety for the vaccine recipient (mother) and also for the fetus/infant,1 and to assess numerous pregnancy-specific outcomes. Although vaccination during pregnancy is not new, much of the research on safety emerged after the global 2009 A/H1N1 influenza pandemic, when immunization campaigns in many countries prioritized pregnant women.2

Since the 2009 A/H1N1 influenza pandemic, the evidence base on the safety of vaccination during pregnancy has evolved further with randomized clinical trials (RCTs)3 and observational studies of seasonal influenza immunization, observational studies of acellular-pertussis-containing vaccines, and most recently, observational studies of COVID-19 vaccination. While these vaccines are now commonly administered during pregnancy, each was initially licensed based on efficacy and safety studies in non-pregnant populations.4 It follows, therefore, that most assessments of vaccine safety during pregnancy have been post-licensure studies. Several new vaccine candidates, intended specifically for use during pregnancy, are at varying stages of clinical development; inclusion of pregnant women in the early stages of their creation marks the start of a new and evolving chapter in evaluation of vaccine safety during pregnancy.

EVALUATION OF VACCINE SAFETY IN PREGNANCY

Approaches for monitoring vaccine safety

Prior to product licensure, vaccines are evaluated for their safety in RCTs. While RCTs are considered the “gold standard” study design for assessing efficacy and safety of interventions and have been instrumental in developing new maternal vaccines, RCTs of vaccines intended for use in the general adult population usually exclude special populations, such as pregnant individuals, necessitating the use of alternate approaches to post-licensure.1 Comprehensive post-licensure vaccine safety assessment involves complementary systems using different approaches and types of data, each with their own strengths and weaknesses.5,6 Typically, this includes both passive (spontaneous reporting; for example, Vaccine Adverse Event Reporting System [VAERS])7 and active (prospective monitoring; for example, Canadian Immunization Monitoring Program ACTive [IMPACT])8 systems, which are important for rapid “signal detection.”6,7,9 Industry-led prospective pregnancy exposure registries may also be established.10 Comparative epidemiologic studies using linked databases capturing whole populations, or subsets of populations (such as those with medical insurance), can be used to follow-up on signals detected by passive/active approaches,7 as well as assess safety of rare and longer-term outcomes.1,5 The expanding availability of routinely collected electronic medical records and healthcare databases since the early 1990s1 has made it possible to conduct observational epidemiologic studies of maternal, fetal, and infant outcomes following vaccination during pregnancy in populations from many countries around the world.

Outcomes

In all populations, pre-licensure vaccine RCTs and post-licensure vaccine safety surveillance systems monitor general adverse events following immunization (e.g., local and systemic reactions, serious adverse events). However, when vaccines are administered during pregnancy, additional pregnancy and birth outcomes must also be evaluated. The Brighton Collaboration’s Global Alignment of Immunization Safety Assessment in Pregnancy (GAIA) initiative has undertaken extensive work to identify core recommended outcomes, with corresponding detailed case definitions, to standardize outcome assessment in vaccine RCTs that include pregnant women (Table 1).11,12

Many of the currently recommended GAIA outcomes are among those most commonly evaluated in observational epidemiological studies (Table 1). A small, but increasing, number of observational studies has assessed longer-term health outcomes in offspring exposed to influenza and pertussis vaccination during pregnancy.13,14 Outcomes assessed have included immune-mediated conditions (e.g., infectious, atopic, autoimmune), neurodevelopmental conditions (e.g., epilepsy, autism spectrum disorders), and all cause morbidity and mortality (Table 1), ascertained for up to 5–6 years of age in the children. While the studies to date have not identified any long-term offspring health concerns following vaccination during pregnancy, the number of studies remains low13,14 and standardization of outcomes and case definitions would enhance the ability to further assess maternal immunization safety.15

1

Outcomes recommended and assessed in pregnancy vaccine safety studies.

Recommended outcomes: Brighton Collaboration Global Alignment of Immunization Safety Assessment in Pregnancy (GAIA)11

  • Hypertensive disorders of pregnancy16
  • Non-reassuring fetal status17
  • Postpartum hemorrhage18
  • Preterm birth19
  • Maternal death20
  • Congenital anomalies21
  • Stillbirth22
  • Neonatal infections23
  • Neonatal death24
  • Antenatal bleeding25
  • Congenital microcephaly26
  • Dysfunctional labor27
  • Failure to thrive28
  • Fetal growth restriction29
  • Gestational diabetes mellitus30
  • Low birth weight31
  • Neonatal encephalopathy32
  • Neonatal respiratory distress33
  • Small for gestational age34
  • Spontaneous abortion and ectopic pregnancy35

Pregnancy and birth outcomes assessed in existing studiesa36

  • Spontaneous abortion
  • Stillbirth
  • Chorioamnionitis
  • Congenital anomalies
  • Microcephaly
  • Neonatal death
  • Neonatal infection
  • Preterm birth
  • Low birth weight
  • Maternal death
  • Small for gestational age

Longer-term offspring outcomes assessed in existing studies37,38,39

  • Infections (common cold, pharyngitis, otitis media, influenza, pneumonia, fever, gastrointestinal infection)
  • Mortality (1 year and 5 year)
  • Infection-related primary care contact
  • Hospitalizations and emergency department visits
  • Autoimmune diseases
  • Neurological diseases
  • Behavioral disorders (autism spectrum disorder, attention deficit hyperactivity disorder)
  • Atopic diseases
  • Neoplasms
  • Sensory disorders
  • Complex chronic conditions
  • Infant growth up to 6 months

a Not an exhaustive list, but those most commonly summarized in systematic reviews.

VACCINES CURRENTLY RECOMMENDED IN PREGNANCY

Influenza vaccines

Pregnancy is a well-established risk factor for severe influenza40 and influenza during pregnancy is associated with adverse pregnancy outcomes.41,42 Influenza vaccination is, therefore, recommended during pregnancy to prevent complications from infection among mothers and their infants.43,44 Inactivated influenza vaccine is the most widely used vaccine during pregnancy globally.45 Many countries recommend inactivated influenza vaccine during any stage of pregnancy, with some restricting recommendations to the second or third trimester or to women with special-risk medical conditions.46,47,48 Live attenuated influenza vaccines are contraindicated in pregnancy.49

Local and systemic reactions

The safety of influenza vaccination during pregnancy has been documented since the 1960s. The first evidence supporting the safety of inactivated influenza vaccination was published in the 1960s and 1970s and indicated no severe reactions and no differences in outcomes between vaccinated and unvaccinated pregnancies.50,51 In the early 2000s, retrospective cohort studies and analysis of passive surveillance data also reported no serious adverse events following inactivated influenza vaccination during pregnancy.52,53 The landmark Mother’s Gift RCT from Bangladesh was published in 2008 – 340 women were randomized to receive either seasonal inactivated influenza vaccine or 23-valent pneumococcal polysaccharide vaccine during their third trimester.54 This trial documented minor local and systemic adverse events during the 7 days post-immunization, but no increased risk of small-for-gestational age (SGA) birth or low birth weight.55

Pregnancy and birth outcomes

The 2009 influenza A/H1N1 pandemic triggered one of the world’s largest mass immunization campaigns. Monovalent adjuvanted and unadjuvanted pandemic vaccines were rapidly developed and disseminated.56 Clinical evidence identified pregnant women as being at higher risk of severe infection,57 and they were prioritized for immunization in many countries.2 Rates of immunization during pregnancy increased dramatically, as did research supporting the safety of maternal immunization. Observational evidence from mass immunization campaigns showed no severe adverse events among pregnant women and no increase in the risk of gestational diabetes, pre-eclampsia, stillbirth, preterm birth, SGA birth, low birth weight, neonatal death, or congenital malformations.58,59,60,61,62,63,64 Clinical trials of pandemic H1N1 vaccine administered during pregnancy found no increase in serious adverse events for mothers or newborns.65,66,67 A systematic review of pandemic vaccines administered during pregnancy indicated that adjuvanted vaccines had more local, but not more serious, adverse events than unadjuvanted vaccines.56

After the 2009 influenza A/H1N1 pandemic, RCTs and observational studies continued to investigate the effects of seasonal inactivated influenza vaccines on perinatal health, largely because of the increasing demand, acceptance, and uptake of influenza vaccination during pregnancy.68,69 Phase 4 RCTs in Mali, Nepal, and South Africa conducted between 2011 and 2013 showed that while immunized pregnant women reported common side effects (injection site reactions, fatigue, headache, malaise, nausea, myalgia, joint pain, rigors),39,70 seasonal inactivated influenza vaccination was not associated with severe maternal or neonatal health outcomes.39 Data from these RCTs showed no association with increased risk of fetal death, preterm birth, low birth weight, or SGA birth.39,46,55,71,72

Active and passive vaccine safety surveillance of recently immunized pregnant women also confirmed the safety of inactivated influenza vaccine during pregnancy,73,74,75 documenting similar rates of adverse events after immunization among non-pregnant women.73 Severe reactions were uncommon among pregnant women;70,76 fewer than 1% reported seeking medical care to treat an adverse event,74,75,77 similar to the prevalence in the general population.78,79 Although incidental exposure to live attenuated influenza vaccines is uncommon (<0.1% of pregnancies),80 several registry-based studies documented no maternal or fetal harm.80,81

Retrospective observational cohort studies have played a major role in the assessment of the safety of inactivated influenza vaccination. The most commonly examined fetal outcomes have been stillbirth, preterm birth, SGA birth, and low birth weight, with studies indicating no increase in risk.36,82,83,84 Fewer analyses have evaluated spontaneous abortion, congenital anomalies, and neonatal death, but again, existing evidence supports no harmful association with inactivated influenza vaccination.36,85 Observational studies that examined the risk of congenital malformations after first-trimester immunization with inactivated influenza vaccines consistently documented no increased risk for adjuvanted or unadjuvanted vaccines.86 Findings on the risk of spontaneous abortion have been inconsistent. Four analyses suggested no association;62,87,88,89 two, a protective association;60,90 and one, a harmful association.91 Given methodological shortcomings of observational studies and limited data from RCTs, safety assessment of early pregnancy exposure to inactivated influenza vaccines is an area of ongoing research.85 Another evolving area is the evaluation of potential non-specific effects of maternal vaccination – a systematic review and meta-analysis of four RCTs of influenza vaccination during pregnancy evaluated this research question. Results showed that although influenza immunization during the second or third trimester of pregnancy reduced influenza infection, immunized women and their infants were more likely to be diagnosed with non-influenza infections.92

Longer-term offspring outcomes

More recently, researchers have used these cohorts and other large administrative databases for longitudinal follow-up of offspring exposed to influenza vaccines in utero. Analyses of the Phase 4 RCTs from Nepal, Mali, and South Africa were updated to include measures of infant growth at 6 months.39 Pooled analysis of these trials showed no association between maternal influenza vaccination and infant growth.39 Pooled analysis of two placebo-controlled clinical trials showed that maternal influenza immunization was not associated with differences in infant all-cause mortality.93 Observational cohort data from the Vaccine Safety Datalink revealed no association with infant hospitalization or mortality.94

Relatively few studies have evaluated offspring outcomes after 12 months. Nonetheless, studies of the incidence of influenza and other childhood infections, primary care utilization, hospital admission, autoimmune diseases, atopic conditions, neurologic diseases, neoplasms, behavioral, neurodevelopmental and sensory disorders, and mortality through 5 years of age showed no evidence of increased risks associated with maternal influenza vaccination with either monovalent pandemic 2009 A/H1N1 vaccines or seasonal influenza vaccines.37,95

Tetanus toxoid vaccines

Maternal and neonatal tetanus predominantly affects the least developed low-income countries. These countries have used tetanus toxoid vaccine during pregnancy since the 1960s,96 and since the mid-1990s, it has been a key component of the global tetanus elimination campaign.97,98

Despite a limited number of epidemiological studies, single antigen tetanus toxoid vaccination during pregnancy is considered safe – mild local reactions such as erythema and injection site pain are common, but severe adverse events are rare.97,99,100 Most safety evidence originates from studies of tetanus toxoid in combination with diphtheria toxoid and pertussis antigens (discussed in the subsequent section). Studies of single antigen tetanus toxoid vaccination include a non-randomized experimental study conducted in New Guinea in the 1960s, which reported no severe adverse events or adverse birth outcomes after up to three doses during pregnancy, compared with no vaccination.96 A hospital-based case-control study of 34,293 cases and 34,477 controls across nine South American countries found tetanus toxoid vaccination in early pregnancy was not associated with congenital malformations.101 Another hospital-based case-control study in Hungary from 1980–1994 compared 21,563 case infants with a congenital anomaly and 35,727 control infants and found no association with tetanus toxoid vaccination in the first three months of pregnancy.102 A 1995 case-control study in a government maternity hospital in Manila, Philippines evaluated risk of spontaneous abortion before 20 weeks’ gestation.103 Cases (n = 100) were women who had a spontaneous abortion and controls (n = 318) were the next three women admitted to hospital immediately after each case with a full-term live birth. No association was observed between spontaneous abortion and receipt of tetanus toxoid vaccine from before pregnancy up to 19 weeks’ gestation. No association was found for those receiving their first dose in early pregnancy, and no difference was related to the number of doses received before or during early pregnancy.103

Pertussis-containing vaccines

In 2011 and 2012, the United States and the United Kingdom, respectively, adopted recommendations that pregnant women be immunized with acellular pertussis-containing vaccines (tetanus-diphtheria-acellular pertussis [Tdap] or diphtheria-tetanus-acellular pertussis [dTpa]) to protect newborns.104,105 Similar recommendations have since been implemented in other high- and middle-income countries.4,106 Several systematic reviews of research published since 2012 reported no association between immunization with pertussis-containing vaccines during pregnancy and serious adverse events in the mother or adverse pregnancy, fetal, or neonatal outcomes in the perinatal period.107,108,109,110,111

Local and systemic reactions

Studies of local and systemic reactions after Tdap immunization during pregnancy have found that the vaccine is generally well tolerated. A prospective observational study in New Zealand followed 793 pregnant women for solicited and unsolicited adverse outcomes after Tdap vaccination during pregnancy; mild injection site reactions were common, but none of the serious adverse events reported were deemed vaccine-related.112 Another study from the United States found increased frequency of moderate/severe injection-site pain in 374 pregnant women, compared with 255 non-pregnant women after Tdap vaccination, but no differences in other moderate/severe reactions or in adverse events requiring medical attention.113 A large matched retrospective cohort study of 53,885 Tdap-exposed and 109,253 unexposed pregnancies in the United States found no increased risk of medically attended allergic reactions, fever, malaise, seizures, altered mental status, or other reactions within 3 days of vaccination, and no differences in the incidence of neurologic events, thrombotic events, new onset proteinuria, gestational diabetes, or cardiac events within 42 days of vaccination. Decreased rates of venous thromboembolic events and thrombocytopenia were observed within 42 days of vaccination among Tdap-vaccinated, compared with unvaccinated, pregnant women.114

Pregnancy and birth outcomes

Overall, studies to-date provide reassuring evidence that Tdap vaccination during pregnancy is not associated with adverse pregnancy or birth outcomes. A systematic review of 14 studies published up to January 10, 2019 (1.4 million pregnant women) found no significant differences in congenital anomalies, pre-eclampsia/eclampsia, low birth weight, neonatal intensive care unit (NICU) admission, neonatal death, or neonatal sepsis between Tdap-vaccinated and unvaccinated women.111 Four studies reported an increase in maternal fever among Tdap-vaccinated women111 – three were RCTs with few events;115,116,117 however, the fourth was a matched retrospective cohort study conducted by the Vaccine Safety Datalink in the United States.114 The study included 53,885 Tdap-vaccinated and 109,253 unvaccinated women; a significantly increased risk of medically attended maternal fever was observed (adjusted incidence rate ratio: 2.16; 95% CI: 1.65–2.83).114

Three cohort studies have reported a small, but significantly increased, risk of chorioamnionitis associated with Tdap vaccination (relative risks ranged from 1.11 to 1.23).118,119,120 Because these studies found no accompanying increases in risk of adverse perinatal outcomes often associated with chorioamnionitis (preterm birth, NICU admission, respiratory distress, neonatal sepsis), the clinical significance of the finding is unclear.118,119,120 It should be noted that this outcome was also assessed by at least four other studies in which no association was observed.121,122,123,124 One study reported an increased risk of postpartum hemorrhage among women who received prenatal Tdap,118 but this association was not observed in several other studies,122,125,126 including a recent population-based retrospective cohort study from Canada, that included 11,519 women vaccinated during pregnancy (postpartum hemorrhage aRR: 1.01, 95% CI: 0.91–1.13; severe postpartum hemorrhage aRR: 0.79, 95% CI: 0.55–1.13).126

At least 12 studies have assessed preterm birth after Tdap vaccination during pregnancy, including three small randomized controlled trials with too few preterm birth outcomes to draw conclusions.111,126,127 In the remaining observational studies, preterm birth was either not associated with vaccination, or was reduced among vaccinated women;111,126,127 the single exception found an increased risk when Tdap was administered before 27 weeks’ gestation, but a lower risk at 27 weeks or later.118 Six studies evaluated stillbirth risk after Tdap vaccination during pregnancy (one RCT115 and five observational studies).123,124,125,126,128 While none reported an increased risk of stillbirth, most of the studies had fewer than 10 events; thus, this is an area that would benefit from future large studies.

Longer-term offspring outcomes

Assessments of longer-term offspring outcomes after Tdap vaccination during pregnancy are more limited than assessments of outcomes proximal to birth. Table 2 summarizes studies that have investigated offspring health beyond 6 months; outcomes evaluated included infectious, atopic, autoimmune, and neurodevelopmental disorders/diseases, as well as all-cause morbidity and mortality.

Two RCTs evaluated growth and development of infants exposed to maternal Tdap immunization. Munoz et al. randomly allocated 33 women to receive a Tdap vaccine and 15 to receive a placebo injection at 30 to 32 weeks’ gestation, followed by crossover vaccination postpartum.116 Outcomes were infant growth (weight, length, and head circumference) at 2, 7, and 13 months and infant development at 13 months assessed using the Bayley-III Scales of Infant and Toddler Development, which measures cognitive, communication, and motor skill development; no statistically significant differences in these measures were found between the two groups.116 Halperin et al. randomly allocated 273 pregnant women to receive either Tdap or tetanus-diphtheria vaccine and compared infant development at 18 months, assessed using the Bayley-III Scales; no developmental differences between groups were detected.129

A small retrospective cohort study by Shakib et al. compared the incidence of complex chronic conditions (composite indicator derived from health administrative data) within the first year among infants exposed and unexposed to Tdap in utero.128 A total of 138 women vaccinated during pregnancy were compared with 552 randomly selected controls; infants exposed to Tdap were no more likely than non-exposed infants to have a complex chronic condition.128 A case-control study from the Vaccine Safety Datalink examined 413,034 singleton, live births between 2004 and 2014 and compared the odds of maternal Tdap vaccination among infants who were hospitalized or died in their first 6 months, with control infants. No association with Tdap vaccination during pregnancy was found for infant hospitalization (adjusted odds ratio [aOR]: 0.94, 95% CI: 0.88–1.01) or infant mortality (aOR: 0.44, 95% CI: 0.17–1.13).94

Few studies of associations between Tdap vaccination during pregnancy and pediatric health outcomes have followed infants beyond 18 months.130,131,132 The outcomes assessed varied widely across studies and included infant growth and development, hospitalization, death, complex chronic conditions, autism spectrum disorder, and attention deficit/hyperactivity disorder, but no study found any increased risk for these outcomes among children born to mothers who received Tdap during pregnancy (Table 2).

2

Summary of findings from studies of longer-term offspring health outcomes after Tdap vaccination during pregnancy.

First author (year of publication)

Location and time period

Study design

Study population and size

Infant outcomes and length of follow-up

Results

Studies with 6–12 months of follow-up

Munoz et al. (2014)116

Houston, Durham, Seattle (USA), 2008–2012

Randomized clinical trial

33 women randomized to receive Tdap and 15 randomized to placebo

  • Outcomes: infant growth at 2, 7, or 13 months (weight, length, and fronto-occipital circumference); development (Bayley III screening test)
  • Follow-up: up to 13 months of age

No difference in infant growth at 2, 7, or 13 months or development measured at 13 months

Shakib et al. (2013)128

Utah (USA), 2005–2009

Retrospective cohort study

138 women vaccinated with Tdap, 552 randomly selected unvaccinated women

  • Outcomes: pediatric complex chronic conditions
  • Follow-up: up to 12 months of age

No significant association between Tdap during pregnancy and pediatric complex chronic conditions within 12 months of age

Sukumaran et al. (2018)94

5 Vaccine Safety Datalink sites in California, Colorado, Wisconsin, and Oregon (USA), 2004–2014

Case-control study

413,034 singleton, live births

  • Outcomes: all-cause hospitalization, hospitalizations from respiratory causes; all-cause mortality
  • Follow-up: up to 6 months of age

No significant association between Tdap during pregnancy and all-cause hospitalization or mortality within 6 months, but significantly lower odds of hospitalizations from respiratory causes

Studies with >12 months of follow-up

Halperin et al. (2018)129

Nova Scotia (Canada), 2007–2014

Randomized clinical trial

135 women allocated to receive Tdap and 138 to Td

  • Outcomes: infant development (Bayley III screening test)
  • Follow-up: at 18 months of age

No difference in development assessed at 18 months

Becerra-Culqui et al. (2018)130

Kaiser Permanente hospitals in Southern California (USA), 2011–2014

Retrospective cohort study

All singleton live births (n = 81,993)

39,077 vaccinated

  • Outcomes: autism spectrum disorder (ASD)
  • Follow-up: 3 to 6.5 years of age

No increased risk of ASD following Tdap vaccination during pregnancy

Becerra-Culqui et al. (2020)131

Kaiser Permanente hospitals in Southern California (USA), 2011–2014

Retrospective cohort study

41,781 women vaccinated with Tdap, 43,826 unvaccinated

  • Outcomes: attention deficit/hyperactivity disorder (ADHD)
  • Follow-up: 3 to <8 years of age

No association between Tdap vaccination during pregnancy and ADHD

Laverty et al. (2021)132

Ontario (Canada), 2012–2017

Retrospective cohort study

12,045 live births exposed to Tdap in utero, 613,598 unexposed

  • Outcomes: immune-related outcomes, pediatric infections, sensory disorders, neoplasm and urgent and inpatient health service utilization
  • Follow-up: up to 6 years of age

No significantly increased risks of adverse childhood outcomes following prenatal Tdap exposure, but inverse associations with upper respiratory infections, gastrointestinal infections, and urgent and inpatient health service use

Tdap, tetanus-diphtheria-acellular pertussis; Td, tetanus-diphtheria.

COVID-19 vaccines

Early in the COVID-19 pandemic, pregnant individuals were determined to be at higher risk of severe disease, with associated adverse pregnancy outcomes.133 Pre-licensure vaccine RCTs excluded pregnant women,134 which created some hesitancy in recommending vaccination for pregnant individuals when the vaccines became available in late 2020. However, in the early months of 2021, mRNA COVID-19 vaccines were recommended during pregnancy in the United States135,136 and Israel.137 As of February 2022, 105 countries now have similar recommendations.138 Systematic reviews continue to confirm a higher risk of maternal and infant morbidity and mortality from SARS-CoV-2 infection during pregnancy,133,139 thus supporting the importance of vaccination.135

Local and systemic reactions

The prevalence of local and systemic reactions post-COVID-19 vaccination has been similar in pregnant and non-pregnant people.140,141,142,143 The most common symptoms include injection site pain, fatigue, headache, and myalgia. The last three symptoms are generally more prevalent after dose 2 than after dose 1 of an mRNA vaccine, and also more prevalent among recipients of mRNA-1273 than of BNT162b2, but otherwise, little difference is apparent by pregnancy status.141,142

Pregnancy and birth outcomes

Descriptive studies of COVID-19 vaccination safety in pregnancy published early in 2021 did not observe increases in the cumulative incidence of preterm birth or other pregnancy outcomes compared with background incidence rates.141,144 As of August 2022, a number of large comparative epidemiological studies have since been published and provide more robust evidence (Table 3). At least two population-based case-control found no increased risk of miscarriage in women vaccinated during their first trimester.145,146 

Three large cohort studies from Scandinavia (Norway and Sweden)147 and Ontario,148,149 Canada that used population-wide birth registry information found no association between COVID-19 vaccination during pregnancy and adverse outcomes, including postpartum hemorrhage, chorioamnionitis, cesarean delivery, preterm birth, SGA birth, stillbirth, NICU admission, or lower Apgar scores. This held for all mRNA vaccine types and all trimesters, although the number of women vaccinated in their first trimester was considerably lower than the number who were vaccinated in the second or third trimester of pregnancy. Two additional large cohort studies of pregnant individuals insured through large health maintenance organizations in the United States (Vaccine Safety Datalink)150 and Israel151 observed no associations between COVID-19 vaccination during pregnancy and risk of preterm birth or SGA birth.

Despite the small number of large, comparative observational studies as of August 2022, findings thus far consistently support the safety of COVID-19 vaccination during pregnancy152,153,154 and complement evidence that COVID-19 vaccination provides effective protection against infection for pregnant women155,156 and their newborns.157,158 Further evaluation is needed for outcomes following first-trimester vaccination and for longer-term offspring outcomes. Initial assessments of both show no indication that vaccination increases the risk of congenital anomalies or indicators of poor health in infants up to 7 months of age (for example, risk of hospitalization or mortality).151 Finally, there is limited safety evidence available on the use of non-mRNA COVID-19 vaccines during pregnancy;152,159 this remains an important evidence gap as of August 2022.

3

Summary of findings from large comparative epidemiological studies of outcomes after COVID-19 vaccination during pregnancy.

First author (year of publication)

Location and time period

Study design

Study population and size

Outcomes

Results

Magnus et al. (2021)146

Norway, February 15–August 15 2021

Case-control study

4,521 women with miscarriages before 14-weeks of gestation (cases), 13,956 confirmed ongoing pregnancies in the first trimester (controls)

  • Outcome: miscarriage before 14 weeks of gestational age

No association between first-trimester miscarriage and vaccination in the previous 3 weeks, and significantly reduced odds for vaccination in the previous 5 weeks

Results were unchanged after stratifying by mRNA vaccine product

Kharbanda et al. (2021)145

8 Vaccine Safety Datalink sites in California, Colorado, Minnesota, Oregon, Washington, Wisconsin (USA), 2020–2021

Case-control study

13,160 women with spontaneous abortion (cases), 92,286 ongoing pregnancies (controls)

  • Outcome: spontaneous abortion before 20 weeks of gestational age

Compared to ongoing pregnancies, no significantly increased odds of vaccination in the previous 28 days

Results unchanged after stratifying by mRNA vaccine product and gestational age of vaccination (6–8 weeks, 9–13 weeks, 14–19 weeks)

Lipkind et al. (2022)150

8 Vaccine Safety Datalink Sites in California, Colorado, Minnesota, Oregon, Washington, Wisconsin (USA), 2020–2021

Retrospective cohort study

46,079 singleton live births; 10,064 with ≥1 COVID-19 vaccine doses in pregnancy, 36,015 unvaccinated

  • Outcomes: preterm birth, SGA birth

Compared to unvaccinated pregnant women, COVID-19 vaccination in pregnancy was not significantly associated with increased risk for preterm birth overall or SGA birth

Results were unchanged after stratifying by mRNA vaccine product and 2nd or 3rd trimester vaccination

Goldshtein et al. (2022)151

Maccabi Healthcare Services (one of four mandated health organizations), Israel, 2021

Retrospective cohort study

24,288 singleton live births; 16,697 exposed to COVID-19 vaccination in utero, 7,591 unexposed

  • Outcomes: preterm birth, SGA birth, congenital malformations, all-cause hospitalizations (>1 day and ≤28 days following birth), jaundice requiring phototherapy, infant death
  • Follow-up: minimum of 1 month, up to a maximum of 7 months

No significant difference or increased risk between prenatal exposure to BNT162b2 vaccination and incidence of preterm birth, SGA birth, all-cause neonatal hospitalizations, postneonatal hospitalizations, congenital anomalies, or infant mortality

Magnus et al. (2022)147

Sweden, Norway, 2021–2022

Retrospective cohort study

157,521 singleton pregnancies; 103,409 in Sweden and 54,112 in Norway

28,506 with ≥1 COVID-19 vaccine doses in pregnancy, 129,015 unvaccinated

  • Outcomes: preterm birth, stillbirth, SGA birth, low newborn 5-minute Apgar score (<7), neonatal intensive care unit admission

No significant association between COVID-19 vaccination during pregnancy and preterm birth, stillbirth, SGA birth, low 5-minute Apgar score, or neonatal intensive care unit admission

Results were unchanged after stratifying by mRNA vaccine product, number of doses received during pregnancy (1 or 2), and 2nd or 3rd trimester vaccination

Fell et al. (2022)148

Ontario (Canada), December 2020– September 2021

Population-based retrospective cohort study

97,590 pregnant individuals; 22,660 with ≥1 COVID-19 vaccine doses in pregnancy, 74,930 unvaccinated during pregnancy (44,815 initiated vaccination post-partum, 30,115 were never vaccinated at any point)

  • Outcomes: post-partum hemorrhage, chorioamnionitis, cesarean delivery (overall and emergency cesarean delivery), admission to neonatal intensive care unit, low newborn 5-minute Apgar score (<7)

COVID-19 vaccination during pregnancy, compared to unvaccinated during pregnancy (each comparison group separately), was not significantly associated with increased risk of post-partum hemorrhage, chorioamnionitis, cesarean delivery, admission to neonatal intensive care, or low 5-minute Apgar score

Results were unchanged after stratifying by mRNA vaccine product, number of doses received during pregnancy (1 or 2), and 2nd or 3rd trimester vaccination

Fell et al. (2022)149

Ontario (Canada), May to December 2021

Population-based retrospective cohort study

85,162 births; 43,099 exposed to ≥1 COVID-19 vaccine doses in pregnancy, 42,063 unvaccinated

Outcomes: preterm birth, SGA birth, stillbirth


No increased risk of preterm birth, SGA birth, or stillbirth associated with COVID-19 mRNA vaccination during pregnancy

 

Results were unchanged after stratifying by mRNA vaccine product,  number of doses received during pregnancy (1 or 2), or trimester of vaccination

SGA, small-for-gestational-age.

VACCINES CONSIDERED IN SPECIFIC CIRCUMSTANCES

Some vaccines are occasionally indicated for unvaccinated or under-vaccinated pregnant women, such as in circumstances when the risk of maternal infection is deemed to be high due to work with a high-risk population, during an outbreak, or during travel to an area with an outbreak or endemic disease.153,160,161 As a consequence of not being routinely recommended, vaccines in this category have limited evidence on safety following exposure during pregnancy. While not a complete list, vaccines in this category with the most safety information are summarized below.

Hepatitis A and B vaccines

Six studies have evaluated the safety of hepatitis A and B vaccines in pregnancy (two retrospective cohort studies from the U.S. Vaccine Safety Datalink162,163 two descriptive reports of passive surveillance data,164,165 one industry-led prospective pregnancy exposure registry descriptive study,166 and one RCT that described pregnancy outcomes among pregnant adolescents who received hepatitis B vaccine as part of the control group167). The two Vaccine Safety Datalink studies assessed over 600,000 pregnancies between 2004 through 2015, of which 1,399 pregnant women received hepatitis B vaccine and 1,140 received hepatitis A vaccine.162,163 Women vaccinated with hepatitis A or B vaccines during pregnancy were compared with women who received other vaccines in pregnancy and with those who were unvaccinated in pregnancy. No significant associations between hepatitis B vaccination during pregnancy and gestational hypertension, gestational diabetes, pre-eclampsia/eclampsia, cesarean delivery, preterm birth, low birth weight, or SGA birth were observed;162 however, an association between hepatitis A vaccination and SGA birth was observed (aOR: 1.32, 95% CI: 1.09–1.60).163 No serious or non-serious adverse events, or adverse pregnancy outcomes were identified from passive reporting,164,165 or from the pregnancy registry166 or RCT.167

Meningococcal vaccines

A recent review summarized studies that have described pregnancy outcomes among women who received a meningococcal vaccine during pregnancy.107 Across the ten studies summarized, including four RCTs (three in which meningococcal vaccine was compared with pneumococcal vaccine and the other in which it was compared with inactivated influenza vaccine), no safety concerns were identified. In the RCT with influenza vaccine as the active treatment group,168 the rate of infant mortality was lower among mothers who received meningococcal vaccine; however, this difference was not statistically significant.93 None of the other studies, including a descriptive cohort study from California169 and two descriptive reports of passive surveillance data from VAERS,170,171 identified any safety concerns.107

Pneumococcal vaccines

A Cochrane Collaboration review, published in 2015,172 identified seven RCTs investigating the use of pneumococcal vaccines in pregnancy.54,173,174,175,176,177,178 While the review’s primary endpoint of interest was efficacy against infant pneumococcal infection, safety data were also evaluated; there were no differences between pneumococcal and control vaccines with respect to injection site tenderness177,175 and no serious adverse events reported.172 More recently, two small RCTs of pneumococcal vaccination during pregnancy have been published – one among Aboriginal mothers in Northern Australia179 and the other among HIV-positive mothers in Brazil.180 The Australian RCT found a higher rate of preterm births among women who received 23-valent pneumococcal polysaccharide vaccine during pregnancy, compared to unvaccinated women.179 While this finding was not statistically significant, the authors suggest that the finding warrants further research and consideration. The other RCT did not identify any safety concerns, nor did a descriptive report of passive surveillance data from VAERS.181

Rabies vaccines

Due to the severe consequences of rabies infection,182 pregnancy is not considered a contraindication to post-exposure prophylaxis with a dose of human rabies immunoglobulin (HRIG) and rabies vaccine. Although there is a theoretical risk of adverse effects from rabies HRIG and vaccination, studies of small cohorts of recently immunized pregnant women (ranging from 6 to 202 pregnant women) have indicated that adverse systemic reactions are similar among pregnant women compared to non-pregnant individuals183 and identified no safety signals in terms of cesarean delivery, spontaneous abortion, stillbirth, congenital malformations, or preterm birth.183,184,185,186 Although the size of existing studies and the range of pregnancy and fetal outcomes evaluated is small, leaving some uncertainty around the safety of rabies HRIG and vaccination during pregnancy, the clinical consensus is that the benefits of immunization of pregnant women far outweigh any theoretical risk of adverse effects from exposure to HRIG and killed rabies virus vaccines.107

Cholera vaccines

Although cholera can have severe consequences for maternal health187 and is known to have harmful effects on the fetus,188,189 the safety of cholera vaccination during pregnancy remains controversial. Three killed whole-cell oral cholera vaccines are currently available and pre-qualified by the World Health Organization for use.187 No specific clinical studies in pregnant women have been conducted, nor have animal studies been completed to evaluate reproductive toxicity and effects on embryo-fetal development; however, since these vaccines are inactivated, there are no theoretical concerns with respect to fetal health. Of five cohort studies that have assessed the safety of cholera vaccination during pregnancy, none identified significant safety risks.188,190,189,191,192,193 A meta-analysis of the 5,584 women included in these cohorts (2,920 vaccinated and 2,664 unvaccinated pregnant women) compared rates of spontaneous abortion, stillbirth, preterm birth, low birth weight, and congenital malformations.194 Results identified no significant increase in risk of these adverse pregnancy outcomes associated with cholera vaccination.194

VACCINES TYPICALLY CONTRAINDICATED IN PREGNANCY

Live vaccines, such as live attenuated viral vaccines, are typically contraindicated during pregnancy because of their ability to replicate in the host, thereby posing a theoretical risk that the virus could cross the placental barrier and either infect, or adversely affect, the fetus.49,195,196,197 As a result, evidence on the safety of this group of vaccines during pregnancy is limited. With the exception of smallpox vaccines, the existing studies on other live virus vaccines are small in size and overwhelmingly pertain to inadvertent exposure to these vaccines, either early in the first trimester, prior to pregnancy recognition, or during the periconceptional period, often in the context of a mass vaccination campaign during an outbreak.195

Smallpox vaccines

On July 23, 2022, the World Health Organization declared the monkeypox outbreak to be a Public Health Emergency of International Concern.198 According to interim guidance released by the World Health Organization on June 14, 2022, mass vaccination is not recommended; however, post-exposure prophylaxis or pre-exposure prophylaxis may be considered for close contacts of monkeypox-infected individuals or groups with high risk of occupational exposure, respectively.199 Pregnant women may be considered for post-exposure or pre-exposure prophylaxis using smallpox vaccine, if an appropriate vaccine is available, following a careful evaluation of risks and benefits, which may differ by type of vaccine product.199Non-replicating (MVA-BN) smallpox vaccines (or minimally-replicating [LC16]  smallpox vaccines) should be used when considering vaccination during pregnancy.199 Although there are limited data on the use of MVA-BN vaccine during pregnancy, no adverse pregnancy outcomes have been reported to-date and there is no theoretical concern since it is a non-replicating vaccine.200         

A systematic review of studies evaluating safety of smallpox vaccination during pregnancy included 37 articles published up to September 2014.201 The review concluded that there was no association identified between smallpox vaccination during pregnancy and spontaneous abortion, preterm birth, or stillbirth. An increased risk of congenital anomalies associated with first-trimester vaccination was observed by the systematic review (pooled OR: 1.34; 95% CI: 1.02-1.77; 5 studies);201 however, in the largest and most recent individual study included in the meta-analysis,202 no association was observed with congenital anomalies overall (adjusted OR: 1.40; 95% CI: 0.94-2.07) or with seven specific types of anomalies.202 The systematic review documented 21 cases of fetal vaccinia among women vaccinated against smallpox during pregnancy—notably, all cases were reported between 1809 and 1985,201 before the availability of non-replicating smallpox vaccine (MVA-BN first became available in 2013).199

Although another review reported statistically significant associations between smallpox vaccination during the first trimester and increased odds of miscarriage (pooled OR: 4.82; 95% CI 2.38-9.77), these findings were observed in two small studies published in 1964 and 1975; both were deemed to be of very low quality.203

Other live virus vaccines

A systematic review of 15 comparative studies published up to November 2019 assessed evidence on the safety of live virus vaccines administered during pregnancy and included the following vaccines: smallpox (eight studies), monovalent rubella (three studies), oral poliovirus (two studies), yellow fever (one study), and dengue (one study).203 No association with miscarriage was found across vaccines (pooled OR: 0.98, 95% CI 0.87–1.10; nine studies), or when assessed by individual vaccine type, with the exception of smallpox vaccine, as discussed above. There were no associations with stillbirth (nine studies), congenital anomalies (12 studies), preterm birth (five studies), or neonatal death (three studies) across vaccines, or when stratified by individual vaccine type.203 The authors of this review also evaluated 23 descriptive studies of outcomes after live virus vaccination during pregnancy and found no documented cases of congenital rubella syndrome among 3918 infants exposed to monovalent rubella vaccine in utero, but one probable case of intrauterine infection after yellow fever vaccination.203

A review of studies published up to December 2018 on the safety of travel vaccines for pregnant women provided narrative summaries of RCTs, descriptive and comparative cohort studies, and case series studies. Live virus vaccines covered by this review included yellow fever, rubella, and measles–mumps–rubella vaccines.107 A total of 732 women were inadvertently exposed to yellow fever vaccine during pregnancy, documented by nine observational studies; although the quality of the studies was judged to be poor, no compelling evidence of safety concerns was reported. A similar conclusion was reached with respect to six studies of rubella vaccine. Little-to-no data are available on the safety of measles and mumps vaccines in pregnant women.107,195

Data are very limited for other live virus vaccines, particularly those that are not targeted toward individuals of reproductive age (e.g., herpes zoster, rotavirus). Analysis of 17 years of data from a pregnancy exposure registry for varicella-zoster virus-containing vaccines documented 928 women vaccinated within 3 months before or during pregnancy. No cases of congenital varicella syndrome or congenital anomalies consistent with congenital varicella syndrome were reported, and the prevalence of major congenital anomalies was similar to background rates.204

Although Japanese encephalitis virus vaccine is a live vaccine and, thus, contraindicated during pregnancy, a study from 2003–2014 among active-duty U.S. military service members documented 513 exposures to Japanese encephalitis virus vaccine during pregnancy (92.4% were vaccinated during the first trimester), within a population of 192,570 pregnancies.205 The investigators evaluated a range of maternal (fever, rash, myalgia, anaphylaxis), pregnancy (spontaneous abortion, pre-eclampsia) and birth outcomes (preterm birth, low birth weight, major congenital anomalies). No statistically significant associations with receipt of one or more doses of Japanese encephalitis vaccine were reported, although the study was underpowered to rule out small risks for most outcomes.205

VACCINES NOT RECOMMENDED IN PREGNANCY

Human papillomavirus (HPV) vaccine is not recommended during pregnancy.206 Nevertheless, as this vaccine is largely targeted to females of early reproductive age, there are a number of studies that have evaluated outcomes following inadvertent pregnancy exposure.167,207,208,209,210,211,212,213,214 One RCT described pregnancy outcomes among 777 pregnant adolescents who received HPV vaccine as part of an RCT in which the comparison group received hepatitis B vaccine; the incidence of pregnancy outcomes between the two groups were similar.167 In a secondary analysis of RCT data from Costa Rica, Panagiotou et al. found no association, overall, with miscarriage for pregnancies conceived within 90 days of HPV vaccination; however, there was an increased risk of miscarriage at 13–20 weeks’ gestation among women who conceived at any point after HPV vaccination.207

Four population-based comparative retrospective cohort studies have been conducted.208,209,210,211 In Denmark, Faber et al. found no increased risk of spontaneous abortion, stillbirth, or 1-year infant mortality among women who received HPV vaccine from 4 weeks before conception up to 22 weeks of gestational age, compared with unvaccinated; there were no differences in findings according to the number of doses received during pregnancy, or whether the vaccination was received before, or during, pregnancy.208 Scheller et al. also used the Danish national registries over a similar time period and found that HPV vaccination during pregnancy was not associated with a significantly higher risk of spontaneous abortion, stillbirth, major birth defect, small size for gestational age, low birth weight, or preterm birth, compared with unvaccinated women.209 Similarly, no associations with adverse pregnancy outcomes were observed in a large study of U.S. military women who received HPV vaccine during pregnancy,210 or in a study from the United Kingdom that assessed outcomes among pregnancies conceived within 30 days before and 45 days after HPV vaccination.211

No other studies, including one descriptive study of passive surveillance data from VAERS212 and three industry-based pregnancy exposure registry studies,213,214,215 have reported any concerning or unexpected patterns in pregnancy and birth outcomes among HPV-vaccinated pregnant women. Despite a generally favorable safety profile, HPV vaccine is not recommended during pregnancy and if a woman becomes pregnant prior to series completion, current recommendations are to delay second and/or third doses until after pregnancy.206

VACCINES UNDER DEVELOPMENT FOR FUTURE USE IN PREGNANCY

A number of new vaccine products that are currently at different stages of development or implementation have potential future implications for pregnancy. This includes vaccines that are (i) specifically indicated for use during pregnancy to passively protect newborns via transplacental transfer of maternal vaccine-derived antibodies (respiratory syncytial virus [RSV], Group B Streptococcus [GBS]); (ii) intended for use in general populations, including pregnant women, in regions with high disease burdens that, in some cases, extol disproportionately increased risks during pregnancy (Ebola, Malaria); or (iii) intended for use in reproductive-age individuals to prevent congenital infections (Zika virus, cytomegalovirus).4

Respiratory syncytial virus (RSV) vaccines

Among RSV vaccine candidates intended for use during pregnancy to confer neonatal protection, the development of recombinant vaccines containing the RSV-F protein is most advanced. Several clinical trials in pregnant individuals have been completed or are underway to explore the safety, immunogenicity, and efficacy of maternal vaccination against infant RSV outcomes. These trials monitor general safety outcomes (solicited and unsolicited adverse events for mothers and infants, including any leading to study withdrawal) and pregnancy-specific outcomes. Of two completed phase 2 and phase 3 studies in pregnant participants, no major safety signals were identified, although short-term reactogenicity and local reactions were more common among vaccine recipients than placebo recipients.216,217 A phase 3 study, which was published in 2020 and included 4,636 pregnant women (3,051 received the vaccine),216 is the largest RCT completed to-date to have evaluated a vaccine specifically developed for use during pregnancy.2

Group B Streptococcus (GBS) vaccines

Three phase 2 studies218,219,220 and one phase 1/2 study221 of a trivalent protein-conjugated capsular polysaccharide GBS vaccine candidate have been completed in pregnant participants; the vaccine was well tolerated, and no safety signals were reported. The focus of clinical studies has recently shifted to GBS vaccine candidates covering more serotypes. An ongoing phase 1/2 study of a hexavalent capsular polysaccharide GBS vaccine in pregnant women will include a range of maternal and infant outcomes up to 12 months (NCT03765073). Two phase 2 studies of a recombinant protein-based GBS vaccine are in progress, one with healthy pregnant participants (NCT05154578), and the other, with pregnant women with and without HIV infection (NCT04596878). The primary outcome of the former is immunogenicity, but the primary outcomes in the latter (NCT04596878) are adverse events and pregnancy- and birth-related outcomes such as gestational age, growth measures and APGAR scores at birth, and developmental outcomes at 6 months.

Ebola vaccines

Even though pregnancy was an exclusion criterion during the clinical development of Ebola vaccines,222 five studies recorded inadvertent exposures in early pregnancy or in new conceptions within 60 days after vaccination.223,224,225,226,227 One study documented a higher frequency of pregnancy loss among vaccinated than unvaccinated participants who became pregnant, but the difference was not statistically significant and may have been due to a reporting bias.223,224 Two phase 3 studies are currently evaluating EBV vaccine (Ad.26.ZEBOV) during pregnancy (NCT04556526 and NCT04152486). Both will assess maternal and infant adverse events, such as maternal and neonatal death, spontaneous abortion, postpartum hemorrhage, congenital malformation, and preterm birth.

Malaria vaccines

A phase 2 placebo-controlled study underway in Mali is exploring the safety and efficacy of PfSPZ malaria vaccine in females of childbearing potential (NCT03989102). Pregnancy is an exclusion criterion, but participants must report interest in becoming pregnant within 2 years. The primary outcomes are local and systemic adverse events within 7 days of vaccination. Although there is a stated intention to follow those who become pregnant until the end of pregnancy and through the infants’ first year of life, specific outcomes are not listed in the summary on clinicaltrials.gov.

Zika virus and cytomegalovirus vaccines

No Zika or cytomegalovirus vaccine studies involving pregnant participants are currently registered, but a phase 2 trial of an mRNA vaccine candidate in healthy adults is underway (NCT04232280). It will evaluate traditional safety endpoints, in order to inform a phase 3 trial in the target population of females of reproductive age. When available, both Zika and cytomegalovirus vaccines would optimally be administered before pregnancy to prevent adverse outcomes associated with early pregnancy infection.161

CHALLENGES FOR VACCINE SAFETY ASSESSMENT DURING PREGNANCY

While in-depth discussion is beyond the scope of this chapter, it is important to highlight several challenges for vaccination safety assessment during pregnancy, for both currently available and future vaccines.

Exclusion of pregnant individuals from pre-licensure vaccine research

A major issue – amplified during the COVID-19 pandemic and recent Ebola outbreaks – is the routine exclusion of pregnant women from pre-licensure vaccine research, resulting in a lack of pregnancy-specific safety data and, ultimately, delayed or limited access to potentially life-saving vaccines.134,197,228 Ethical considerations for including pregnant women in RCTs are complex. While there may be justification for exclusion from the earliest stages of safety assessment, a requirement that developmental and reproductive toxicity studies are completed during early developmental phases of new vaccine candidates can help ensure that pregnant women can be responsibly included, when appropriate, in pre-licensure RCTs to expedite equitable access.134,228

Surveillance systems and data requirements for vaccine safety assessment in pregnancy

Given the typical exclusion of pregnant women from RCTs, it follows that vaccine safety surveillance and observational studies of vaccination during pregnancy using “real-world” health data are critical during all phases of vaccine program implementation in this population. Key resources essential for such assessments include population-level information on background rates of pregnancy-related maternal and infant outcomes; active and passive vaccine surveillance capacity; and, ideally, linkable individual-level sources of information on pregnancy outcomes and vaccination status during pregnancy.

Low- and middle-income country (LMIC) settings

The above-listed resources for vaccine safety monitoring in pregnant individuals may be limited, or even entirely lacking, in low- and middle-income countries (LMICs).229,230,231,232,233 Specifically, many LMICs lack data sources with information on pregnancy and birth outcomes, due to inadequate birth registration systems and lack of central repositories for antenatal care records (most of which are still paper-based and poorly maintained).231,233 This poses important challenges for establishing local background rates of adverse pregnancy outcomes and monitoring safety after vaccine introduction.229,234 Strengthening health information systems and pregnancy registries in settings currently lacking these resources is likely to become even more important in the future, as new vaccines targeting the pregnant population are licensed and implemented, particularly in LMICs with high disease burdens.232

Recommended outcomes and standardized case definitions

Despite Brighton Collaboration’s GAIA initiative to align core outcome definitions for maternal immunization safety studies (Table 1), the types of outcomes and definitions used in vaccination studies are often heterogeneous83 (partly owing to jurisdiction-specific birth registration requirements and/or data systems). As well, gestational age thresholds and local reference standards are often poorly reported. Such heterogeneity impedes evidence synthesis through meta-analysis, which is important for rare outcomes, such as stillbirth and congenital anomalies.11,12 Of particular value to vaccine safety evaluation are networks that can share or combine data across geopolitical boundaries, thereby enabling harmonized study definitions and analytical methodologies in large populations.1 The Vaccine Safety Datalink, which collects vaccination and healthcare data on more than 10 million insured people in the United States per year,235 is one example.

Methodological challenges facing observational studies of vaccination during pregnancy

Comparative observational studies provide complementary evidence to RCTs and, together, they can inform clinicians’ and policymakers’ risk–benefit decisions about vaccination during pregnancy. However, observational epidemiological studies assessing vaccination during pregnancy face some unique methodological challenges, such as achieving adequate control of confounding factors and accounting for complex temporal issues (for example, seasonality, immortal time, and cohort truncation) that can severely distort results particularly for vaccines available only at specific times (seasonal influenza vaccine or during a period of intense vaccine deployment [2009 A/H1N1 pandemic influenza vaccine, COVID-19 vaccine]) and for outcomes that are time dependent (preterm birth, stillbirth).236,237,238

PRACTICE RECOMMENDATIONS

  • Safety assessment of vaccination during pregnancy involves diverse and complementary approaches – adverse event reporting (passive and active) for rapid detection of safety signals, ideally followed by epidemiological studies designed to compare risks of adverse pregnancy and birth outcomes in vaccinated and unvaccinated pregnant individuals.
  • There are robust safety data for several vaccines currently recommended during pregnancy that can be used during clinical counseling of pregnant patients: influenza; monovalent tetanus toxoid; tetanus toxoid, inactivated reduced diphtheria toxoid, and acellular pertussis (Tdap).
    • Influenza: a large number of published studies, including clinical trials, have documented the safety of inactivated influenza vaccination during pregnancy for mother and infant. Studies of longer-term offspring outcomes are fewer in number, but have not documented increases in the risk of adverse childhood health outcomes.
    • Monovalent tetanus toxoid: a limited number of studies of monovalent tetanus toxoid vaccination during pregnancy are available; most safety evidence is for combination vaccines (see below). Nevertheless, tetanus toxoid vaccine is considered safe during pregnancy, and it is widely administered during pregnancy in many low- and middle-income countries. Mild local reactions such as erythema and injection site pain are common, but severe adverse events are rare and there is no evidence of any adverse pregnancy and birth outcomes.
    • Tetanus-diphtheria-acellular pertussis (Tdap): research published since 2012 reported no association between Tdap immunization during pregnancy and serious adverse events in the mother or adverse pregnancy, fetal, or neonatal outcomes in the perinatal period. An increasing number of studies of longer-term offspring outcomes has not documented increases in the risk of adverse childhood health outcomes.
  • COVID-19 vaccination is also currently recommended for pregnant individuals in many countries due to the increased risk of adverse maternal and newborn outcomes associated with SARS-CoV-2 infection during pregnancy.
    • Safety evidence from descriptive and comparative epidemiological studies is rapidly emerging and, to-date (as of August 2022), is reassuring and suggests there are no associations between mRNA COVID-19 vaccination during pregnancy with miscarriage, preterm birth, SGA birth, stillbirth, or other adverse outcomes.
    • Nevertheless, evidence on other important outcomes, including congenital anomalies, remains limited. Studies following periconceptional and 1st trimester vaccination are required, in addition to longer-term follow-up of newborn health outcomes after birth, and evidence on safety of non-mRNA COVID-19 vaccines used during pregnancy. 
  • Additional inactivated vaccines may warrant consideration for unvaccinated or under-vaccinated pregnant women with chronic health conditions or in specific circumstances, such as during an outbreak or travel/prolonged stay in areas with endemic disease; there are relatively limited data on outcomes following exposure to these vaccines during pregnancy.
    • These vaccines include hepatitis A and B, meningococcal conjugate, pneumococcal, rabies, cholera, and anthrax.
    • Care providers should balance patient-specific risks and benefits and consider offering the vaccine to pregnant patients when potential benefits outweigh potential risks.
  • Live virus vaccines, and other live vaccines, are typically contraindicated during pregnancy due to a theoretical risk that the agent could replicate, cross the placental barrier, and harm the fetus; there are very limited data on outcomes following exposure to these vaccines during pregnancy.
    • These live vaccines include BCG vaccine, live attenuated influenza vaccine, measles–mumps–rubella vaccine, varicella vaccine, yellow fever vaccine, zoster vaccine, Japanese encephalitis virus vaccine, oral polio vaccine, typhoid toxoid vaccine, and dengue vaccine.
    • In the context of the global monkeypox outbreak in 2022, the World Health Organization recommends that pregnant women may be considered for post-exposure or pre-exposure prophylaxis using a non-replicating smallpox vaccine, following a careful evaluation of risks and benefits.
    • Despite theoretical concerns about safety, inadvertent vaccination with a live vaccine during the periconceptional period or during early gestation, before pregnancy recognition, is not a reason to terminate pregnancy.
  • HPV vaccine is not recommended during pregnancy and if a woman becomes pregnant prior to series completion, current recommendations are to delay second and/or third doses until after pregnancy.
  • Since recommendations for vaccination during pregnancy differ by country/jurisdiction, care providers should refer to country-specific guidance. Note that in the context of an outbreak, such as COVID-19 or monkeypox, recommendations concerning vaccination during pregnancy may change during the course of the outbreak as additional data become available. 
  • Care providers should be aware of local passive and active vaccine safety surveillance programs and report any suspected vaccine-associated adverse events following vaccination during pregnancy.


CONFLICTS OF INTEREST

The author(s) of this chapter declare that they have no interests that conflict with the contents of the chapter.

REFERENCES

1

Dodd C, Andrews N, Petousis-Harris H, et al. Methodological frontiers in vaccine safety: qualifying available evidence for rare events, use of distributed data networks to monitor vaccine safety issues, and monitoring the safety of pregnancy interventions. BMJ Glob Heal 2021;6(Suppl 2). doi:10.1136/bmjgh-2020-003540

2

Kachikis A, Eckert LO, Englund JA. Chapter 1. The History of Maternal Immunization. 1st edn. In: Leuridan EE, Nunes MC, Jones CE. (eds.). Elsevier, 2019.

3

Omer SB, Richards JL, Madhi SA, et al. Three randomized trials of maternal influenza immunization in Mali, Nepal, and South Africa: Methods and expectations. Vaccine 2015;33(32):3801–12. doi:10.1016/j.vaccine.2015.05.077.

4

Abu-Raya B, Maertens K, Edwards KM, et al. Global Perspectives on Immunization During Pregnancy and Priorities for Future Research and Development: An International Consensus Statement. Front Immunol 2020;11:1282. doi:10.3389/fimmu.2020.01282.

5

Moro PL, Haber P, McNeil MM. Challenges in evaluating post-licensure vaccine safety: observations from the Centers for Disease Control and Prevention. Expert Rev Vaccines 2019;18(10):1091–101. doi:10.1080/14760584.2019.1676154.

6

Products UKM& H, Agency R. Report of the Commission on Human Medicines Expert Working Group on COVID-19 Vaccine Safety Surveillance, 2021. https://www.gov.uk/government/publications/report-of-the-commission-on-human-medicines-expert-working-group-on-covid-19-vaccine-safety-surveillance/report-of-the-commission-on-human-medicines-expert-working-group-on-covid-19-vaccine-safety-surveillance.

7

Moro PL, Li R, Haber P, et al. Surveillance systems and methods for monitoring the post-marketing safety of influenza vaccines at the Centers for Disease Control and Prevention. Expert Opin Drug Saf 2016;15(9):1175–83. doi:10.1080/14740338.2016.1194823.

8

Bettinger JA, Halperin SA, Vaudry W, et al. The Canadian Immunization Monitoring Program, ACTive (IMPACT): Active surveillance for vaccine adverse events and vaccine-preventable diseases. Can Commun Dis Rep 2014;40(Suppl 3):41–4. doi:10.14745/ccdr.v40is3a06.

9

Dolk H, Damase-Michel C, Morris JK, et al. COVID-19 in pregnancy-what study designs can we use to assess the risk of congenital anomalies in relation to COVID-19 disease, treatment and vaccination? Paediatr Perinat Epidemiol 2022. doi:10.1111/ppe.12840.

10

Wyszynski DF. Pregnancy exposure registries: academic opportunities and industry responsibility. Birth Defects Res A Clin Mol Teratol 2009;85(1):93–101. doi:10.1002/bdra.20525.

11

Munoz FM, Eckert LO, Katz MA, et al. Key terms for the assessment of the safety of vaccines in pregnancy: Results of a global consultative process to initiate harmonization of adverse event definitions. Vaccine 2015;33(47):6441–52. doi:10.1016/j.vaccine.2015.07.112.

12

Bonhoeffer J, Kochhar S, Hirschfeld S, et al. Global alignment of immunization safety assessment in pregnancy – The GAIA project. Vaccine 2016;34(49):5993–7. doi:10.1016/j.vaccine.2016.07.006.

13

Munoz FM, Long SS. The Safety of Maternal Influenza Vaccination and Infant Health Outcomes. Pediatrics 2020. doi:10.1542/peds.2020-006916.

14

Hviid A. Vaccine Safety in Pregnancy: Going Beyond the Perinatal Period. Ann Intern Med 2020;173(8):658–9. doi:10.7326/M20-5489.

15

Edwards KM. Ensuring the Safety of Maternal Immunization. Pediatrics 2021;147(5). doi:10.1542/peds.2020-047456.

16

Rouse CE, Eckert LO, Wylie BJ, et al. Hypertensive disorders of pregnancy: Case definitions & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2016;34(49):6069–76. doi:10.1016/j.vaccine.2016.03.038.

17

Gravett C, Eckert LO, Gravett MG, et al. Non-reassuring fetal status: Case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2016;34(49):6084–92. doi:10.1016/j.vaccine.2016.03.043.

18

Kerr R, Eckert LO, Winikoff B, et al. Postpartum haemorrhage: Case definition and guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2016;34(49):6102–9. doi:10.1016/j.vaccine.2016.03.039.

19

Quinn J-A, Munoz FM, Gonik B, et al. Preterm birth: Case definition & guidelines for data collection, analysis, and presentation of immunisation safety data. Vaccine 2016;34(49):6047–56. doi:10.1016/j.vaccine.2016.03.045.

20

Patwardhan M, Eckert LO, Spiegel H, et al. Maternal death: Case definition and guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2016;34(49):6077–83. doi:10.1016/j.vaccine.2016.03.042.

21

DeSilva M, Munoz FM, Mcmillan M, et al. Congenital anomalies: Case definition and guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2016;34(49):6015–26. doi:10.1016/j.vaccine.2016.03.047.

22

Tavares Da Silva F, Gonik B, McMillan M, et al. Stillbirth: Case definition and guidelines for data collection, analysis, and presentation of maternal immunization safety data. Vaccine 2016;34(49):6057–68. doi:10.1016/j.vaccine.2016.03.044.

23

Vergnano S, Buttery J, Cailes B, et al. Neonatal infections: Case definition and guidelines for data collection, analysis, and presentation of immunisation safety data. Vaccine 2016;34(49):6038–46. doi:10.1016/j.vaccine.2016.03.046.

24

Pathirana J, Muñoz FM, Abbing-Karahagopian V, et al. Neonatal death: Case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2016;34(49):6027–37. doi:10.1016/j.vaccine.2016.03.040.

25

Prabhu M, Eckert LO, Belfort M, et al. Antenatal bleeding: Case definition and guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2017;35(48 Pt A):6529–37. doi:10.1016/j.vaccine.2017.01.081.

26

DeSilva M, Munoz FM, Sell E, et al. Congenital microcephaly: Case definition & guidelines for data collection, analysis, and presentation of safety data after maternal immunisation. Vaccine 2017;35(48 Pt A):6472–82. doi:10.1016/j.vaccine.2017.01.044.

27

Boatin AA, Eckert LO, Boulvain M, et al. Dysfunctional labor: Case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2017;35(48 Pt A):6538–45. doi:10.1016/j.vaccine.2017.01.050.

28

Ross E, Munoz FM, Edem B, et al. Failure to thrive: Case definition & guidelines for data collection, analysis, and presentation of maternal immunisation safety data. Vaccine 2017;35(48 Pt A):6483–91. doi:10.1016/j.vaccine.2017.01.051.

29

Easter SR, Eckert LO, Boghossian N, et al. Fetal growth restriction: Case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2017;35(48 Pt A):6546–54. doi:10.1016/j.vaccine.2017.01.042.

30

Kachikis A, Eckert LO, Walker C, et al. Gestational diabetes mellitus: Case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2017;35(48 Pt A):6555–62. doi:10.1016/j.vaccine.2017.01.043.

31

Cutland CL, Lackritz EM, Mallett-Moore T, et al. Low birth weight: Case definition & guidelines for data collection, analysis, and presentation of maternal immunization safety data. Vaccine 2017;35(48 Pt A):6492–500. doi:10.1016/j.vaccine.2017.01.049.

32

Sell E, Munoz FM, Soe A, et al. Neonatal encephalopathy: Case definition & guidelines for data collection, analysis, and presentation of maternal immunisation safety data. Vaccine 2017;35(48 Pt A):6501–5. doi:10.1016/j.vaccine.2017.01.045.

33

Sweet LR, Keech C, Klein NP, et al. Respiratory distress in the neonate: Case definition & guidelines for data collection, analysis, and presentation of maternal immunization safety data. Vaccine 2017;35(48 Pt A):6506–17. doi:10.1016/j.vaccine.2017.01.046.

34

Schlaudecker EP, Munoz FM, Bardají A, et al. Small for gestational age: Case definition & guidelines for data collection, analysis, and presentation of maternal immunisation safety data. Vaccine 2017;35(48 Pt A):6518–28. doi:10.1016/j.vaccine.2017.01.040.

35

Rouse CE, Eckert LO, Babarinsa I, et al. Spontaneous abortion and ectopic pregnancy: Case definition & guidelines for data collection, analysis, and presentation of maternal immunization safety data. Vaccine 2017;35(48 Pt A):6563–74. doi:10.1016/j.vaccine.2017.01.047.

36

Macias Saint-Gerons D, Solà Arnau I, De Mucio B, et al. Adverse events associated with the use of recommended vaccines during pregnancy: An overview of systematic reviews. Vaccine 2020. doi:10.1016/j.vaccine.2020.07.048.

37

Foo DYP, Sarna M, Pereira G, et al. Early Childhood Health Outcomes Following In Utero Exposure to Influenza Vaccines: A Systematic Review. Pediatrics 2020. doi:10.1542/peds.2020-0375.

38

Lasky T, McMahon AW, Hua W, et al. Methodologic approaches in studies using real-world data (RWD) to measure pediatric safety and effectiveness of vaccines administered to pregnant women: A scoping review. Vaccine 2021;39(29):3814–24. doi:10.1016/j.vaccine.2021.05.071.

39

Omer SB, Clark DR, Madhi SA, et al. Efficacy, duration of protection, birth outcomes, and infant growth associated with influenza vaccination in pregnancy: a pooled analysis of three randomised controlled trials. Lancet Respir Med 2020;8(6):597–608. doi:10.1016/S2213-2600(19)30479-5.

40

Mertz D, Lo CK-F, Lytvyn L, et al. Pregnancy as a risk factor for severe influenza infection: an individual participant data meta-analysis. BMC Infect Dis 2019;19(1):683. doi:10.1186/s12879-019-4318-3.

41

Fell DB, Savitz DA, Kramer MS, et al. Maternal influenza and birth outcomes: systematic review of comparative studies. BJOG 2017;124(1):48–59. doi:10.1111/1471-0528.14143.

42

Gunnes N, Gjessing HK, Bakken IJ, et al. Seasonal and pandemic influenza during pregnancy and risk of fetal death: A Norwegian registry-based cohort study. Eur J Epidemiol 2020;35(4):371–9. doi:10.1007/s10654-020-00600-z.

43

World Health Organization. Vaccines against influenza WHO position paper – November 2012. Wkly Epidemiol Rec 2012;47(47):461–76. http://www.ncbi.nlm.nih.gov/pubmed/24340402.

44

Grohskopf LA, Alyanak E, Broder KR, et al. Prevention and Control of Seasonal Influenza with Vaccines: Recommendations of the Advisory Committee on Immunization Practices – United States, 2020–21 Influenza Season. MMWR Recomm reports Morb Mortal Wkly report Recomm reports 2020;69(8):1–24. doi:10.15585/mmwr.rr6908a1.

45

Ortiz JR, Perut M, Dumolard L, et al. A global review of national influenza immunization policies: Analysis of the 2014 WHO/UNICEF Joint Reporting Form on immunization. Vaccine 2016;34(45):5400–5. doi:10.1016/j.vaccine.2016.07.045.

46

Regan AK, Munoz FM. Efficacy and safety of influenza vaccination during pregnancy: realizing the potential of maternal influenza immunization. Expert Rev Vaccines 2021;20(6):649–60. doi:10.1080/14760584.2021.1915138.

47

Principi N, Camilloni B, Esposito S, et al. Influenza immunization policies: Which could be the main reasons for differences among countries? Hum Vaccin Immunother 2018;14(3):684–92. doi:10.1080/21645515.2017.1405188.

48

European Centre for Disease Prevention and Control. Seasonal Influenza Vaccination and Antiviral Use in EU/EEA Member States – Overview of Vaccine Recommendations for 2017–2018 and Vaccination Coverage Rates for 2015–2016 and 2016–2017 Influenza Seasons 2018.

49

Omer SB. Maternal Immunization. N Engl J Med 2017;376(25):2497. doi:10.1056/NEJMc1705482.

50

Hulka JF. Effectiveness of Polyvalent Influenza Vaccine in Pregnancy. Report of a Controlled Study During an Outbreak of Asian Influenza. Obstet Gynecol 1964;23:830–7. http://www.ncbi.nlm.nih.gov/pubmed/14168242.

51

Sumaya C V, Gibbs RS. Immunization of pregnant women with influenza A/New Jersey/76 virus vaccine: reactogenicity and immunogenicity in mother and infant. J Infect Dis 1979;140(2):141–6. doi:10.1093/infdis/140.2.141.

52

Munoz FM, Greisinger AJ, Wehmanen OA, et al. Safety of influenza vaccination during pregnancy. Am J Obstet Gynecol 2005;192(4):1098–106. doi:10.1016/j.ajog.2004.12.019.

53

Pool V, Iskander J. Safety of influenza vaccination during pregnancy. Am J Obstet Gynecol 2006;194(4):1200; author reply 1201. doi:10.1016/j.ajog.2005.07.091.

54

Zaman K, Roy E, Arifeen SE, et al. Effectiveness of maternal influenza immunization in mothers and infants. N Engl J Med 2008;359(15):1555–64. doi:10.1056/NEJMoa0708630.

55

Steinhoff MC, Omer SB, Roy E, et al. Neonatal outcomes after influenza immunization during pregnancy: a randomized controlled trial. CMAJ 2012;184(6):645–53. doi:10.1503/cmaj.110754.

56

Juvet LK, Robertson AH, Laake I, et al. Safety of Influenza A H1N1pdm09 Vaccines: An Overview of Systematic Reviews. Front Immunol 2021;12:740048. doi:10.3389/fimmu.2021.740048.

57

Siston AM, Rasmussen SA, Honein MA, et al. Pandemic 2009 influenza A(H1N1) virus illness among pregnant women in the United States. JAMA 2010;303(15):1517–25. doi:10.1001/jama.2010.479.

58

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.e1–9. doi:10.1016/j.ajog.2011.06.047.

59

Mackenzie IS, MacDonald TM, Shakir S, et al. Influenza H1N1 (swine flu) vaccination: a safety surveillance feasibility study using self-reporting of serious adverse events and pregnancy outcomes. Br J Clin Pharmacol 2012;73(5):801–11. doi:10.1111/j.1365-2125.2011.04142.x.

60

Oppermann M, Fritzsche J, Weber-Schoendorfer C, et al. A(H1N1) v2009: a controlled observational prospective cohort study on vaccine safety in pregnancy. Vaccine 2012;30(30):4445–52. doi:10.1016/j.vaccine.2012.04.081.

61

Håberg SE, Trogstad L, Gunnes N, et al. Risk of fetal death after pandemic influenza virus infection or vaccination. N Engl J Med 2013;368(4):333–40. doi:10.1056/NEJMoa1207210.

62

Pasternak B, Svanström H, Mølgaard-Nielsen D, et al. Vaccination against pandemic A/H1N1 2009 influenza in pregnancy and risk of fetal death: cohort study in Denmark. BMJ 2012;344(may02 1):e2794. doi:10.1136/bmj.e2794.

63

Tavares F, Nazareth I, Monegal JS, et al. Pregnancy and safety outcomes in women vaccinated with an AS03-adjuvanted split virion H1N1 (2009) pandemic influenza vaccine during pregnancy: a prospective cohort study. Vaccine 2011;29(37):6358–65. doi:10.1016/j.vaccine.2011.04.114.

64

Heikkinen T, Young J, van Beek E, et al. Safety of MF59-adjuvanted A/H1N1 influenza vaccine in pregnancy: a comparative cohort study. Am J Obstet Gynecol 2012;207(3):177.e1–8. doi:10.1016/j.ajog.2012.07.007.

65

Abzug MJ, Nachman SA, Muresan P, et al. Safety and immunogenicity of 2009 pH1N1 vaccination in HIV-infected pregnant women. Clin Infect Dis 2013;56(10):1488–97. doi:10.1093/cid/cit057.

66

Tsatsaris V, Capitant C, Schmitz T, et al. Maternal immune response and neonatal seroprotection from a single dose of a monovalent nonadjuvanted 2009 influenza A(H1N1) vaccine: a single-group trial. Ann Intern Med 2011;155(11):733–41. doi:10.1059/0003-4819-155-11-201112060-00005.

67

Horiya M, Hisano M, Iwasaki Y, et al. Efficacy of double vaccination with the 2009 pandemic influenza A (H1N1) vaccine during pregnancy. Obstet Gynecol 2011;118(4):887–94. doi:10.1097/AOG.0b013e31822e5c02.

68

Regan AK, Mak DB, Hauck YL, et al. Trends in seasonal influenza vaccine uptake during pregnancy in Western Australia: Implications for midwives. Women Birth 2016;29(5):423–9. doi:10.1016/j.wombi.2016.01.009.

69

Seo J, Lim J. Trends in influenza vaccination coverage rates in South Korea from 2005 to 2014: Effect of public health policies on vaccination behavior. Vaccine 2018;36(25):3666–73. doi:10.1016/j.vaccine.2018.05.024.

70

Nunes MC, Cutland CL, Moultrie A, et al. Immunogenicity and safety of different dosing schedules of trivalent inactivated influenza vaccine in pregnant women with HIV: a randomised controlled trial. lancet HIV 2020;7(2):e91–103. doi:10.1016/S2352-3018(19)30322-4.

71

Simões EAF, Nunes MC, Carosone-Link P, et al. Trivalent influenza vaccination randomized control trial of pregnant women and adverse fetal outcomes. Vaccine 2019;37(36):5397–403. doi:10.1016/j.vaccine.2019.07.024.

72

Steinhoff MC, Katz J, Englund JA, et al. Year-round influenza immunisation during pregnancy in Nepal: a phase 4, randomised, placebo-controlled trial. Lancet Infect Dis 2017;17(9):981–9. doi:10.1016/S1473-3099(17)30252-9.

73

Regan AK, Tracey L, Blyth CC, et al. A prospective cohort study comparing the reactogenicity of trivalent influenza vaccine in pregnant and non-pregnant women. BMC Pregnancy Childbirth 2015;15:61. doi:10.1186/s12884-015-0495-2.

74

Regan AK, Tracey LE, Blyth CC, et al. A prospective cohort study assessing the reactogenicity of pertussis and influenza vaccines administered during pregnancy. Vaccine 2016;34(20):2299–304. doi:10.1016/j.vaccine.2016.03.084.

75

Glover C, Crawford N, Leeb A, et al. Active SMS-based surveillance of adverse events following immunisation with influenza and pertussis-containing vaccines in Australian pregnant women using AusVaxSafety. Vaccine 2020;38(31):4892–900. doi:10.1016/j.vaccine.2020.04.056.

76

Munoz FM, Jackson LA, Swamy GK, et al. Safety and immunogenicity of seasonal trivalent inactivated influenza vaccines in pregnant women. Vaccine 2018;36(52):8054–61. doi:10.1016/j.vaccine.2018.10.088.

77

Regan AK, Blyth CC, Effler P V. Using SMS technology to verify the safety of seasonal trivalent influenza vaccine for pregnant women in real time. Med J Aust 2013;199(11):744–6.

78

Leeb A, Regan AK, Peters IJ, et al. Using automated text messages to monitor adverse events following immunisation in general practice. Med J Aust 2014;200(7):416–8. doi:10.5694/mja13.11166.

79

Pillsbury A, Quinn H, Cashman P, et al. Active SMS-based influenza vaccine safety surveillance in Australian children. Vaccine 2017;35(51):7101–6. doi:10.1016/j.vaccine.2017.10.091.

80

Toback SL, Beigi R, Tennis P, et al. Maternal outcomes among pregnant women receiving live attenuated influenza vaccine. Influenza Other Respi Viruses 2012;6(1):44–51. doi:10.1111/j.1750-2659.2011.00266.x.

81

Moro PL, Broder K, Zheteyeva Y, et al. Adverse events in pregnant women following administration of trivalent inactivated influenza vaccine and live attenuated influenza vaccine in the Vaccine Adverse Event Reporting System, 1990–2009. Am J Obstet Gynecol 2011;204(2):146.e1–7. doi:10.1016/j.ajog.2010.08.050.

82

Bratton KN, Wardle MT, Orenstein WA, et al. Maternal influenza immunization and birth outcomes of stillbirth and spontaneous abortion: a systematic review and meta-analysis. Clin Infect Dis 2015;60(5):e11–9. doi:10.1093/cid/ciu915.

83

Fell DBB, Platt RWW, Lanes A, et al. Fetal death and preterm birth associated with maternal influenza vaccination: systematic review. BJOG 2015;122(1):17–26. doi:10.1111/1471-0528.12977.

84

Nunes MC, Aqil AR, Omer SB, et al. The Effects of Influenza Vaccination during Pregnancy on Birth Outcomes: A Systematic Review and Meta-Analysis. Am J Perinatol 2016;33(11):1104–14. doi:10.1055/s-0036-1586101.

85

Speake HA, Pereira G, Regan AK. Risk of adverse maternal and foetal outcomes associated with inactivated influenza vaccination in first trimester of pregnancy. Paediatr Perinat Epidemiol 2020;(June 2020):196–205. doi:10.1111/ppe.12715.

86

Polyzos KA, Konstantelias AA, Pitsa CE, et al. Maternal Influenza Vaccination and Risk for Congenital Malformations: A Systematic Review and Meta-analysis. Obstet Gynecol 2015;126(5):1075–84. doi:10.1097/AOG.0000000000001068.

87

Beau a B, Hurault-Delarue C, Vidal S, et al. Pandemic A/H1N1 influenza vaccination during pregnancy: a comparative study using the EFEMERIS database. Vaccine 2014;32(11):1254–8. doi:10.1016/j.vaccine.2014.01.021.

88

Chambers CD, Johnson D, Xu R, et al. Risks and safety of pandemic h1n1 influenza vaccine in pregnancy: Birth defects, spontaneous abortion, preterm delivery, and small for gestational age infants. Vaccine 2013;31(44):5026–32. doi:10.1016/j.vaccine.2013.08.097.

89

Chambers CD, Johnson DL, Xu R, et al. Safety of the 2010–11, 2011–12, 2012–13, and 2013–14 seasonal influenza vaccines in pregnancy: Birth defects, spontaneous abortion, preterm delivery, and small for gestational age infants, a study from the cohort arm of VAMPSS. Vaccine 2016;34(37):4443–9. doi:10.1016/j.vaccine.2016.06.054.

90

Sammon CJ, Snowball J, McGrogan A, et al. Evaluating the hazard of foetal death following H1N1 influenza vaccination; a population based cohort study in the UK GPRD. In: Baradaran HR. (ed.) PLoS One 2012;7(12):e51734. doi:10.1371/journal.pone.0051734.

91

Donahue JG, Kieke BA, King JP, et al. Association of spontaneous abortion with receipt of inactivated influenza vaccine containing H1N1pdm09 in 2010–11 and 2011–12. Vaccine 2017;35(40):5314–22. doi:10.1016/j.vaccine.2017.06.069.

92

Hansen KP, Benn CS, Aamand T, et al. Does Influenza Vaccination during Pregnancy Have Effects on Non-Influenza Infectious Morbidity? A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Vaccines 2021;9(12). doi:10.3390/vaccines9121452.

93

Clark DR, Omer SB, Tapia MD, et al. Influenza or Meningococcal Immunization During Pregnancy and Mortality in Women and Infants: A Pooled Analysis of Randomized Controlled Trials. Pediatr Infect Dis J 2020;39(7):641–4. doi:10.1097/INF.0000000000002629.

94

Sukumaran L, Mccarthy NL, Kharbanda EO, et al. Infant hospitalizations and mortality after maternal vaccination. Pediatrics 2018;141(3):e20173310. doi:10.1542/peds.2017-3310.

95

Mehrabadi A, Dodds L, MacDonald NE, et al. Association of Maternal Influenza Vaccination During Pregnancy With Early Childhood Health Outcomes. JAMA 2021;325(22):2285–93. doi:10.1001/jama.2021.6778.

96

Schofield FD, Tucker VM, Westbrook GR. Neonatal tetanus in New Guinea. Effect of active immunization in pregnancy. Br Med J 1961;2(5255):785–9. doi:10.1136/bmj.2.5255.785.

97

World Health Organizat. Tetanus vaccines: WHO position paper – February 2017. Wkly Epidemiol Rec 2017;6(92):53–76.

98

Yusuf N, Raza AA, Chang-Blanc D, et al. Progress and barriers towards maternal and neonatal tetanus elimination in the remaining 12 countries: a systematic review. Lancet Glob Heal 2021;9(11):e1610-e1617. doi:10.1016/S2214-109X(21)00338-7.

99

Yildirim I, Omer S. Tetanus. In: Leuridan E, Nunes MC, Jones CE. (eds.) Maternal Immunization, 1st edn. Elsevier, 2020:115–27.

100

Omer S, Jamieson DJ. Maternal Immunization. In: SA P, WA O, PA O, et al. (eds.) Vaccines, 7th edn. Elsevier, 2018:567–79.

101

Silveira CM, Cáceres VM, Dutra MG, et al. Safety of tetanus toxoid in pregnant women: a hospital-based case-control study of congenital anomalies. Bull World Health Organ 1995;73(5):605–8. http://www.ncbi.nlm.nih.gov/pubmed/8846486.

102

Czeizel AE, Rockenbauer M. Tetanus toxoid and congenital abnormalities. Int J Gynaecol Obstet 1999;64(3):253–8. doi:10.1016/s0020-7292(99)00008-9.

103

Catindig N, Abad-Viola G, Magboo F, et al. Tetanus toxoid and spontaneous abortions: is there epidemiological evidence of an association? Lancet (London, England) 1996;348(9034):1098–9. doi:10.1016/S0140-6736(05)64442-X.

104

Centers for Disease Control and Prevention (CDC). Updated recommendations for use of tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis vaccine (Tdap) in pregnant women–Advisory Committee on Immunization Practices (ACIP), 2012. MMWR Morb Mortal Wkly Rep 2013;62(7):131–5. Accessed March 15, 2017. http://www.ncbi.nlm.nih.gov/pubmed/23425962.

105

Amirthalingam G, Letley L, Campbell H, et al. Lessons learnt from the implementation of maternal immunization programs in England. Hum Vaccin Immunother 2016;12(11):2934–9. doi:10.1080/21645515.2016.1210730.

106

Maertens K, Edwards KM, Pertussis LE. In: Leuridan E, Nunes MC, Jones C. (eds.) Maternal Immunization, 1st edn. Elsevier, 2020:169–85.

107

Nasser R, Rakedzon S, Dickstein Y, et al. Are all vaccines safe for the pregnant traveller? A systematic review and meta-analysis. J Travel Med 2020;27(2). doi:10.1093/jtm/taz074.

108

Gkentzi D, Katsakiori P, Marangos M, et al. Maternal vaccination against pertussis: a systematic review of the recent literature. Arch Dis Child – Fetal Neonatal Ed 2017;102(5):F456-F463. doi:10.1136/archdischild-2016-312341.

109

Furuta M, Sin J, Ng ESW, et al. Efficacy and safety of pertussis vaccination for pregnant women – a systematic review of randomised controlled trials and observational studies. BMC Pregnancy Childbirth 2017;17(1):390. doi:10.1186/s12884-017-1559-2.

110

McMillan M, Clarke M, Parrella A, et al. Safety of tetanus, diphtheria, and pertussis vaccination during pregnancy: a systematic review. Obstet Gynecol 2017;129(3):560–73. doi:10.1097/AOG.0000000000001888.

111

Vygen-Bonnet S, Hellenbrand W, Garbe E, et al. Safety and effectiveness of acellular pertussis vaccination during pregnancy: a systematic review. BMC Infect Dis 2020;20(1):136. doi:10.1186/s12879-020-4824-3.

112

Petousis-Harris H, Walls T, Watson D, et al. Safety of Tdap vaccine in pregnant women: an observational study. BMJ Open 2016;6(4):e010911. doi:10.1136/bmjopen-2015-010911.

113

Fortner KB, Swamy GK, Broder KR, et al. Reactogenicity and immunogenicity of tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis vaccine (Tdap) in pregnant and nonpregnant women. Vaccine 2018;36(42):6354–60. doi:10.1016/j.vaccine.2018.07.012.

114

Kharbanda EO, Vazquez-Benitez G, Lipkind HS, et al. Maternal Tdap vaccination: Coverage and acute safety outcomes in the vaccine safety datalink, 2007–2013. Vaccine 2016;34(7):968–73. doi:10.1016/j.vaccine.2015.12.046.

115

Hoang HTT, Leuridan E, Maertens K, et al. Pertussis vaccination during pregnancy in Vietnam: Results of a randomized controlled trial Pertussis vaccination during pregnancy. Vaccine 2016;34(1). doi:10.1016/j.vaccine.2015.10.098.

116

Munoz FM, Bond NH, Maccato M, et al. Safety and immunogenicity of tetanus diphtheria and acellular pertussis (Tdap) immunization during pregnancy in mothers and infants. JAMA 2014;311(17):1760. doi:10.1001/jama.2014.3633.

117

Maertens K, Caboréb RN, Huygenb K, et al. Pertussis vaccination during pregnancy in Belgium: Results of a prospective controlled cohort study Kirsten. Vaccine 2016;34(1):151–9. doi:10.1016/j.vaccine.2015.10.098.

118

Layton JB, Butler AM, Li D, et al. Prenatal Tdap immunization and risk of maternal and newborn adverse events. Vaccine 2017;35(33):4072–8. doi:10.1016/j.vaccine.2017.06.071.

119

DeSilva M, Vazquez-Benitez G, Nordin JD, et al. Maternal Tdap vaccination and risk of infant morbidity. Vaccine 2017;35(29):3655–60. doi:10.1016/j.vaccine.2017.05.041.

120

Kharbanda EO, Vazquez-Benitez G, Lipkind HS, et al. Evaluation of the association of maternal pertussis vaccination with obstetric events and birth outcomes. JAMA 2014;312(18):1897–904. doi:10.1001/jama.2014.14825.

121

Guo Y, Murphy MSQ, Erwin E, et al. Birth outcomes following cesarean delivery on maternal request: a population-based cohort study. CMAJ 2021;193(18):E634-E644. doi:10.1503/cmaj.202262.

122

Griffin JB, Yu L, Watson D, et al. Pertussis Immunisation in Pregnancy Safety (PIPS) Study: A retrospective cohort study of safety outcomes in pregnant women vaccinated with Tdap vaccine. Vaccine 2018;36(34):5173–9. doi:10.1016/j.vaccine.2018.07.011.

123

Berenson AB, Hirth JM, Rahman M, et al. Maternal and infant outcomes among women vaccinated against pertussis during pregnancy. Hum Vaccin Immunother 2016;12(8):1965–71. doi:10.1080/21645515.2016.1157241.

124

Morgan JL, Baggari SR, McIntire DD, et al. Pregnancy outcomes after antepartum tetanus, diphtheria, and acellular pertussis vaccination. Obstet Gynecol 2015;125(6):1433–8. doi:10.1097/AOG.0000000000000862.

125

Donegan K, King B, Bryan P. Safety of pertussis vaccination in pregnant women in UK: observational study. BMJ 2014;349(9526):g4219. doi:10.1136/bmj.g4219.

126

Fakhraei R, Crowcroft N, Bolotin S, et al. Obstetric and perinatal health outcomes after pertussis vaccination during pregnancy in Ontario, Canada: a retrospective cohort study. C open 2021;9(2):E349. doi:10.9778/cmajo.20200239.

127

Petousis-Harris H, Jiang Y, Yu L, et al. A Retrospective Cohort Study of Safety Outcomes in New Zealand Infants Exposed to Tdap Vaccine in Utero. Vaccines 2019;7(4):147. doi:10.3390/vaccines7040147.

128

Shakib JH, Korgenski K, Sheng X, et al. Tetanus, diphtheria, acellular pertussis vaccine during pregnancy: pregnancy and infant health outcomes. J Pediatr 2013;163(5):1422–6.e1–4. doi:10.1016/j.jpeds.2013.06.021.

129

Halperin SASA, Langley JMJM, Ye L, et al. A randomized, controlled trial of the safety and immunogenicity of tetanus, diphtheria, and acellular pertussis vaccine (Tdap) immunization during pregnancy and Subsequent Infant Immune Response. Clin Infect Dis 2018;In press(7):1063–71. doi:10.1093/cid/ciy244.

130

Becerra-Culqui TA, Getahun D, Chiu V, et al. Prenatal Tetanus, Diphtheria, Acellular Pertussis Vaccination and Autism Spectrum Disorder. Pediatrics 2018;142(3). doi:10.1542/peds.2018-0120.

131

Becerra-Culqui TA, Getahun D, Chiu V, et al. The Association of Prenatal Tetanus, Diphtheria, and Acellular Pertussis (Tdap) Vaccination With Attention-Deficit/Hyperactivity Disorder. Am J Epidemiol 2020;189(10):1163–72. doi:10.1093/aje/kwaa074.

132

Laverty M, Crowcroft N, Bolotin S, et al. Health Outcomes in Young Children Following Pertussis Vaccination During Pregnancy. Pediatrics 2021;147(5). doi:10.1542/peds.2020-042507.

133

Allotey J, Stallings E, Bonet M, et al. Clinical manifestations, risk factors, and maternal and perinatal outcomes of coronavirus disease 2019 in pregnancy: living systematic review and meta-analysis. BMJ 2020;370:m3320. doi:10.1136/bmj.m3320.

134

Manca T, Baylis F, Munoz FM, et al. Prioritise research on vaccines for pregnant and breastfeeding women. Lancet (London, England) 2022;399(10328):890–3. doi:10.1016/S0140-6736(22)00379-8.

135

Prevention UC for DC and. COVID-19 Vaccines While Pregnant or Breastfeeding 2021. Accessed March 20, 2022. https://www.cdc.gov/coronavirus/2019-ncov/vaccines/recommendations/pregnancy.html.

136

American College of Obstetricians and Gynecologists. ACOG and SMFM Recommend COVID-19 Vaccination for Pregnant Individuals 2021. Accessed March 20, 2022. https://www.acog.org/news/news-releases/2021/07/acog-smfm-recommend-covid-19-vaccination-for-pregnant-individuals.

137

Israeli Ministry of Health. Vaccination recommendation for high-risk pregnant women (in Hebrew) 2021. Accessed March 20, 2022. https://www.gov.il/he/departments/news/19012021-05.

138

Berman Institute of Bioethics & Center for Immunization Research JHU. COVID-19 Maternal Immunization Tracker (COMIT): COVID-19 Vaccine policies for pregnant and lactating people worldwide 2021. Accessed January 14, 2022. www.comitglobal.org.

139

Pashaei Z, SeyedAlinaghi S, Qaderi K, et al. Prenatal and neonatal complications of COVID-19: A systematic review. Heal Sci reports 2022;5(2):e510. doi:10.1002/hsr2.510.

140

Nakahara A, Biggio JR, Elmayan A, et al. Safety-Related Outcomes of Novel mRNA COVID-19 Vaccines in Pregnancy. Am J Perinatol 2022. doi:10.1055/a-1745-1168.

141

Shimabukuro TT, Kim SY, Myers TR, et al. Preliminary findings of mRNA Covid-19 vaccine safety in pregnant persons. N Engl J Med 2021;384(24):2273–82. doi:10.1056/NEJMoa2104983.

142

Kachikis A, Englund JA, Singleton M, et al. Short-term Reactions Among Pregnant and Lactating Individuals in the First Wave of the COVID-19 Vaccine Rollout. JAMA Netw open 2021;4(8):e2121310. doi:10.1001/jamanetworkopen.2021.21310.

143

Blakeway H, Prasad S, Kalafat E, et al. COVID-19 vaccination during pregnancy: coverage and safety. Am J Obstet Gynecol 2022;226(2):236.e1–236.e14. doi:10.1016/j.ajog.2021.08.007.

144

Zauche LH, Wallace B, Smoots AN, et al. Receipt of mRNA Covid-19 Vaccines and Risk of Spontaneous Abortion. N Engl J Med 2021;385(16):1533–5. doi:10.1056/NEJMc2113891.

145

Kharbanda EO, Haapala J, DeSilva M, et al. Spontaneous abortion following COVID-19 vaccination during pregnancy. JAMA 2021;326(16):1629–31. doi:10.1001/jama.2021.15494.

146

Magnus MC, Gjessing HK, Eide HN, Wilcox AJ, Fell DB, Håberg SE. Covid-19 vaccination during pregnancy and first-trimester miscarriage. N Engl J Med 2021;385(21):2008–10. doi:10.1056/NEJMc2114466.

147

Magnus MC, Örtqvist AK, Dahlqwist E, et al. Association of SARS-CoV-2 Vaccination During Pregnancy With Pregnancy Outcomes. JAMA 2022;In press. doi:10.1001/jama.2022.3271.

148

Fell DB, Dhinsa T, Alton GD, et al. Association of COVID-19 Vaccination in Pregnancy With Adverse Peripartum Outcomes. JAMA 2022;In press. doi:10.1001/jama.2022.4255.

149

Fell DB, Dimanlig-Cruz S, Regan AK, et al. Risk of preterm birth, small for gestational age at birth, and stillbirth after covid-19 vaccination during pregnancy: population based retrospective cohort study. BMJ. 2022;378:e071416. doi:10.1136/bmj-2022-071416

150

Lipkind HS, Vazquez-Benitez G, DeSilva M, et al. Receipt of COVID-19 vaccine during pregnancy and preterm or small-for-gestational-age at birth – Eight integrated health care organizations, United States, December 15, 2020-July 22, 2021. MMWR Morb Mortal Wkly Rep 2022;71(1):26–30. doi:10.15585/mmwr.mm7101e1.

151

Goldshtein I, Steinberg DM, Kuint J, et al. Association of BNT162b2 COVID-19 vaccination during pregnancy with neonatal and early infant outcomes. JAMA Pediatr 2022. doi:10.1001/jamapediatrics.2022.0001.

152

Badell ML, Dude CM, Rasmussen SA, Jamieson DJ. Covid-19 vaccination in pregnancy. BMJ. 2022;378:e069741. doi:10.1136/bmj-2021-069741

153

Etti M, Calvert A, Galiza E, et al. Maternal vaccination: a review of current evidence and recommendations. Am J Obstet Gynecol 2021. doi:10.1016/j.ajog.2021.10.041.

154

Rawal S, Tackett RL, Stone RH, et al. COVID-19 Vaccination among Pregnant People in the U.S.: A Systematic Review. Am J Obstet Gynecol MFM 2022:100616. doi:10.1016/j.ajogmf.2022.100616.

155

Dagan N, Barda N, Biron-Shental T, et al. Effectiveness of the BNT162b2 mRNA COVID-19 vaccine in pregnancy. Nat Med 2021;27(10):1693–95. doi:10.1038/s41591-021-01490-8.

156

Goldshtein I, Nevo D, Steinberg DM, et al. Association Between BNT162b2 Vaccination and Incidence of SARS-CoV-2 Infection in Pregnant Women. JAMA 2021;326(8):728–35. doi:10.1001/jama.2021.11035.

157

Halasa NB, Olson SM, Staat MA, et al. Effectiveness of Maternal Vaccination with mRNA COVID-19 Vaccine During Pregnancy Against COVID-19-Associated Hospitalization in Infants Aged <6 Months – 17 States, July 2021-January 2022. MMWR Morb Mortal Wkly Rep 2022;71(7):264–70. doi:10.15585/mmwr.mm7107e3.

158

Carlsen EØ, Magnus MC, Oakley L, et al. Association of COVID-19 Vaccination During Pregnancy With Incidence of SARS-CoV-2 Infection in Infants. JAMA Intern Med. Published online June 1, 2022. doi:10.1001/jamainternmed.2022.2442

159

Kharbanda EO, Vazquez-Benitez G. COVID-19 mRNA Vaccines During Pregnancy: New Evidence to Help Address Vaccine Hesitancy. JAMA. 2022;327(15):1451-1453. doi:10.1001/jama.2022.2459

160

Perrett KP, Nolan TM. Immunization During Pregnancy: Impact on the Infant. Paediatr Drugs 2017;19(4):313–24. doi:10.1007/s40272-017-0231-7.

161

Engmann C, Fleming JA, Khan S, et al. Closer and closer? Maternal immunization: current promise, future horizons. J Perinatol 2020;40(6):844–57. doi:10.1038/s41372-020-0668-3.

162

Groom HC, Irving SA, Koppolu P, et al. Uptake and safety of Hepatitis B vaccination during pregnancy: A Vaccine Safety Datalink study. Vaccine. 2018;36(41):6111-6116. doi:10.1016/j.vaccine.2018.08.074

163

Groom HC, Smith N, Irving SA, et al. Uptake and safety of hepatitis A vaccination during pregnancy: A Vaccine Safety Datalink study. Vaccine 2019;37(44):6648–55. doi:10.1016/j.vaccine.2019.09.043.

164

Moro PL, Museru OI, Niu M, et al. Reports to the Vaccine Adverse Event Reporting System after hepatitis A and hepatitis AB vaccines in pregnant women. Am J Obstet Gynecol 2014;210(6):561.e1–6. doi:10.1016/j.ajog.2013.12.036.

165

Moro PL, Zheteyeva Y, Barash F, et al. Assessing the safety of hepatitis B vaccination during pregnancy in the Vaccine Adverse Event Reporting System (VAERS), 1990–2016. Vaccine 2018;36(1):50–4. doi:10.1016/j.vaccine.2017.11.039.

166

Celzo F, Buyse H, Welby S, et al. Safety evaluation of adverse events following vaccination with Havrix, Engerix-B or Twinrix during pregnancy. Vaccine 2020;38(40):6215–23. doi:10.1016/j.vaccine.2020.07.041.

167

Bi D, Apter D, Eriksson T, et al. Safety of the AS04-adjuvanted human papillomavirus (HPV)-16/18 vaccine in adolescents aged 12–15 years: end-of-study results from a community-randomized study up to 6.5 years. Hum Vaccin Immunother 2020;16(6):1392–403. doi:10.1080/21645515.2019.1692557.

168

Tapia MD, Sow SO, Tamboura B, et al. Maternal immunisation with trivalent inactivated influenza vaccine for prevention of influenza in infants in Mali: a prospective, active-controlled, observer-blind, randomised phase 4 trial. Lancet Infect Dis 2016;16(9):1026–35. doi:10.1016/S1473-3099(16)30054-8.

169

Becerra-Culqui TA, Sy LS, Ackerson BK, et al. Safety of MenACWY-CRM vaccine exposure during pregnancy. Vaccine 2020;38(12):2683–90. doi:10.1016/j.vaccine.2020.02.008.

170

Myers TR, McNeil MM, Ng CS, et al. Adverse events following quadrivalent meningococcal diphtheria toxoid conjugate vaccine (Menactra®) reported to the Vaccine Adverse Event Reporting System (VAERS), 2005–2016. Vaccine 2020;38(40):6291–8. doi:10.1016/j.vaccine.2020.07.039.

171

Myers TR, McNeil MM, Ng CS, et al. Adverse events following quadrivalent meningococcal CRM-conjugate vaccine (Menveo®) reported to the Vaccine Adverse Event Reporting system (VAERS), 2010–2015. Vaccine 2017;35(14):1758–63. doi:10.1016/j.vaccine.2017.02.030.

172

Chaithongwongwatthana S, Yamasmit W, Limpongsanurak S, et al. Pneumococcal vaccination during pregnancy for preventing infant infection. Cochrane database Syst Rev 2015;1:CD004903. doi:10.1002/14651858.CD004903.pub4.

173

Lopes CRC, Berezin EN, Ching TH, et al. Ineffectiveness for infants of immunization of mothers with pneumococcal capsular polysaccharide vaccine during pregnancy. Braz J Infect Dis 2009;13(2):104–6.

174

O’Dempsey TJ, McArdle T, Ceesay SJ, et al. Immunization with a pneumococcal capsular polysaccharide vaccine during pregnancy. Vaccine 1996;14(10):963–70. doi:10.1016/0264-410x(96)00009-6.

175

Munoz FM, Englund JA, Cheesman CC, et al. Maternal immunization with pneumococcal polysaccharide vaccine in the third trimester of gestation. Vaccine 2001;20(5–6):826–37. doi:10.1016/s0264-410x(01)00397-8.

176

Quiambao BP, Nohynek H, Käyhty H, et al. Maternal immunization with pneumococcal polysaccharide vaccine in the Philippines. Vaccine 2003;21(24):3451–4. doi:10.1016/s0264-410x(03)00349-9.

177

Shahid NS, Steinhoff MC, Hoque SS, et al. Serum, breast milk, and infant antibody after maternal immunisation with pneumococcal vaccine. Lancet (London, England) 1995;346(8985):1252–7. doi:10.1016/s0140-6736(95)91861-2.

178

Obaro SK, Deubzer HE, Newman VO, et al. Serotype-specific pneumococcal antibodies in breast milk of Gambian women immunized with a pneumococcal polysaccharide vaccine during pregnancy. Pediatr Infect Dis J 2004;23(11):1023–9. doi:10.1097/01.inf.0000143651.54880.09.

179

McHugh L, Binks M, Ware RS, et al. Birth outcomes in Aboriginal mother-infant pairs from the Northern Territory, Australia, who received 23-valent polysaccharide pneumococcal vaccination during pregnancy, 2006–2011: The PneuMum randomised controlled trial. Aust N Z J Obstet Gynaecol 2020;60(1):82–7. doi:10.1111/ajo.13002.

180

Weinberg A, Muresan P, Laimon L, et al. Safety, immunogenicity, and transplacental antibody transport of conjugated and polysaccharide pneumococcal vaccines administered to pregnant women with HIV: a multicentre randomised controlled trial. lancet HIV 2021;8(7):e408–19. doi:10.1016/S2352-3018(20)30339-8.

181

Miller ER, Moro PL, Cano M, et al. Post-licensure safety surveillance of 23-valent pneumococcal polysaccharide vaccine in the Vaccine Adverse Event Reporting System (VAERS), 1990–2013. Vaccine 2016;34(25):2841–6. doi:10.1016/j.vaccine.2016.04.021.

182

Abazeed ME, Cinti S. Rabies prophylaxis for pregnant women. Emerg Infect Dis 2007;13(12):1966–7. doi:10.3201/eid1312.070157.

183

Chutivongse S, Wilde H, Benjavongkulchai M, et al. Postexposure rabies vaccination during pregnancy: effect on 202 women and their infants. Clin Infect Dis 1995;20(4):818–20. doi:10.1093/clinids/20.4.818.

184

Huang G, Liu H, Cao Q, et al. Safety of post-exposure rabies prophylaxis during pregnancy: a follow-up study from Guangzhou, China. Hum Vaccin Immunother 2013;9(1):177–83. doi:10.4161/hv.22377.

185

Chutivongse S, Wilde H. Postexposure rabies vaccination during pregnancy: experience with 21 patients. Vaccine 1989;7(6):546–8. doi:10.1016/0264-410x(89)90280-6.

186

Sudarshan MK, Madhusudana SN, Mahendra BJ. Post-exposure prophylaxis with purified vero cell rabies vaccine during pregnancy–safety and immunogenicity. J Commun Dis 1999;31(4):229–36. http://www.ncbi.nlm.nih.gov/pubmed/10937299.

187

Global Task Force on Cholera Control (GTFCC) WHO. Evidence of the Risks and Benefits of Vaccinating Pregnant Women with WHO Pre-Qualified Cholera Vaccines during Mass Campaigns, 2016. https://www.who.int/cholera/vaccines/Risk_Benefits_vaccinating_pregnant_women_Technical_Note.pdf?ua=1.

188

Grout L, Martinez-Pino I, Ciglenecki I, et al. Pregnancy Outcomes after a Mass Vaccination Campaign with an Oral Cholera Vaccine in Guinea: A Retrospective Cohort Study. PLoS Negl Trop Dis 2015;9(12):e0004274. doi:10.1371/journal.pntd.0004274.

189

Ciglenecki I, Bichet M, Tena J, et al. Cholera in pregnancy: outcomes from a specialized cholera treatment unit for pregnant women in Léogâne, Haiti. PLoS Negl Trop Dis 2013;7(8):e2368. doi:10.1371/journal.pntd.0002368.

190

Ali M, Nelson A, Luquero FJ, et al. Safety of a killed oral cholera vaccine (Shanchol) in pregnant women in Malawi: an observational cohort study. Lancet Infect Dis 2017;17(5):538–44. doi:10.1016/S1473-3099(16)30523-0.

191

Hashim R, Khatib AM, Enwere G, et al. Safety of the recombinant cholera toxin B subunit, killed whole-cell (rBS-WC) oral cholera vaccine in pregnancy. PLoS Negl Trop Dis 2012;6(7):e1743. doi:10.1371/journal.pntd.0001743.

192

Khan AI, Ali M, Chowdhury F, et al. Safety of the oral cholera vaccine in pregnancy: Retrospective findings from a subgroup following mass vaccination campaign in Dhaka, Bangladesh. Vaccine 2017;35(11):1538–43. doi:10.1016/j.vaccine.2017.01.080.

193

Khan AI, Ali M, Lynch J, et al. Safety of a bivalent, killed, whole-cell oral cholera vaccine in pregnant women in Bangladesh: evidence from a randomized placebo-controlled trial. BMC Infect Dis 2019;19(1):422. doi:10.1186/s12879-019-4006-3.

194

Zhang Y, Zhang H, Wang B, et al. Pregnancy outcomes after a mass vaccination campaign with an oral cholera vaccine: a systematic review and meta-analysis. BJOG 2020;127(9):1066–73. doi:10.1111/1471-0528.16260.

195

Keller-Stanislawski B, Englund JA, Kang G, et al. Safety of immunization during pregnancy: a review of the evidence of selected inactivated and live attenuated vaccines. Vaccine 2014;32(52):7057–64. doi:10.1016/j.vaccine.2014.09.052.

196

Psarris A, Sindos M, Daskalakis G, et al. Immunizations during pregnancy: How, when and why. Eur J Obstet Gynecol Reprod Biol 2019;240:29–35. doi:10.1016/j.ejogrb.2019.06.019.

197

Jaffe E, Goldfarb IT, Lyerly AD. The Costs of Contradictory Messages About Live Vaccines in Pregnancy. Am J Public Health 2021;111(3):498–503. doi:10.2105/AJPH.2020.306045.

198

World Health Organization. WHO Director-General declares the ongoing monkeypox outbreak a Public Health Emergency of International Concern. Published 2022. Accessed August 19, 2022. https://www.who.int/europe/news/item/23-07-2022-who-director-general-declares-the-ongoing-monkeypox-outbreak-a-public-health-event-of-international-concern

199

World Health Organizat. Vaccines and immunization for monkeypox. Published 2022. Accessed August 19, 2022. https://apps.who.int/iris/bitstream/handle/10665/356120/WHO-MPX-Immunization-2022.1-eng.pdf

200

Khalil A, Samara A, O’Brien P, et al. Monkeypox and pregnancy: what do obstetricians need to know? Ultrasound Obstet Gynecol. 2022;60(1):22-27. doi:10.1002/uog.24968

201

Badell ML, Meaney-Delman D, Tuuli MG, et al. Risks Associated With Smallpox Vaccination in Pregnancy: A Systematic Review and Meta-analysis. Obstet Gynecol. 2015;125(6):1439-1451. doi:10.1097/AOG.0000000000000857

202

Ryan MAK, Gumbs GR, Conlin AMS, et al. Evaluation of preterm births and birth defects in liveborn infants of US military women who received smallpox vaccine. Birth Defects Res A Clin Mol Teratol. 2008;82(7):533-539. doi:10.1002/bdra.20470

203

Laris-González A, Bernal-Serrano D, Jarde A, Kampmann B. Safety of Administering Live Vaccines During Pregnancy: A Systematic Review and Meta-Analysis of Pregnancy Outcomes. Vaccines. 2020;8(1). doi:10.3390/vaccines8010124

204

Marin M, Willis ED, Marko A, et al. Closure of varicella-zoster virus-containing vaccines pregnancy registry &ndash, United States, 2013. MMWR Morb Mortal Wkly Rep 2014;63(33):732–3. http://www.ncbi.nlm.nih.gov/pubmed/25144545.

205

Khodr ZG, Hall C, Chang RN, et al. Japanese encephalitis vaccination in pregnancy among U.S. active duty military women. Vaccine 2020;38(29):4529–35. doi:10.1016/j.vaccine.2020.05.023.

206

Prevention UC for DC and. HPV Vaccination Recommendations. Vaccines and Preventable Diseases 2021. Accessed April 1, 2022. https://www.cdc.gov/vaccines/vpd/hpv/hcp/recommendations.html#:~:text=HPV vaccine is not recommended,before vaccination is not needed.

207

Panagiotou OA, Befano BL, Gonzalez P, et al. Effect of bivalent human papillomavirus vaccination on pregnancy outcomes: long term observational follow-up in the Costa Rica HPV Vaccine Trial. BMJ 2015;351:h4358. doi:10.1136/bmj.h4358.

208

Faber MT, Duun-Henriksen AK, Dehlendorff C, et al. Adverse pregnancy outcomes and infant mortality after quadrivalent HPV vaccination during pregnancy. Vaccine 2019;37(2):265–71. doi:10.1016/j.vaccine.2018.11.030.

209

Scheller NM, Pasternak B, Mølgaard-Nielsen D, Svanström H, Hviid A. Quadrivalent HPV Vaccination and the Risk of Adverse Pregnancy Outcomes. N Engl J Med 2017;376(13):1223–33. doi:10.1056/nejmoa1612296.

210

Bukowinski AT, Hall C, Chang RN, et al. Maternal and infant outcomes following exposure to quadrivalent human papillomavirus vaccine during pregnancy. Vaccine 2020;38(37):5933–9. doi:10.1016/j.vaccine.2020.06.073.

211

Baril L, Rosillon D, Willame C, et al. Risk of spontaneous abortion and other pregnancy outcomes in 15–25 year old women exposed to human papillomavirus-16/18 AS04-adjuvanted vaccine in the United Kingdom. Vaccine 2015;33(48):6884–91. doi:10.1016/j.vaccine.2015.07.024.

212

Moro PL, Zheteyeva Y, Lewis P, et al. Safety of quadrivalent human papillomavirus vaccine (Gardasil) in pregnancy: adverse events among non-manufacturer reports in the Vaccine Adverse Event Reporting System, 2006–2013. Vaccine 2015;33(4):519–22. doi:10.1016/j.vaccine.2014.11.047.

213

Goss MA, Lievano F, Buchanan KM, et al. Final report on exposure during pregnancy from a pregnancy registry for quadrivalent human papillomavirus vaccine. Vaccine 2015;33(29):3422–8. doi:10.1016/j.vaccine.2015.04.014.

214

Dana A, Buchanan KM, Goss MA, et al. Pregnancy outcomes from the pregnancy registry of a human papillomavirus type 6/11/16/18 vaccine. Obstet Gynecol 2009;114(6):1170–8. doi:10.1097/AOG.0b013e3181c2a122.

215

López-Fauqued M, Zima J, Angelo M-G, et al. Results on exposure during pregnancy from a pregnancy registry for AS04-HPV-16/18 vaccine. Vaccine 2017;35(40):5325–30. doi:10.1016/j.vaccine.2017.08.042.

216

Madhi SA, Polack FP, Piedra PA, et al. Respiratory Syncytial Virus Vaccination during Pregnancy and Effects in Infants. N Engl J Med 2020;383(5):426–39. doi:10.1056/NEJMoa1908380.

217

Muňoz FM, Swamy GK, Hickman SP, et al. Safety and Immunogenicity of a Respiratory Syncytial Virus Fusion (F) Protein Nanoparticle Vaccine in Healthy Third-Trimester Pregnant Women and Their Infants. J Infect Dis 2019;220(11):1802–15. doi:10.1093/infdis/jiz390.

218

Donders GGG, Halperin SA, Devlieger R, et al. Maternal immunization with an investigational trivalent Group B Streptococcal vaccine: a randomized controlled trial. Obstet Gynecol 2016;127(2):213–21. doi:10.1097/AOG.0000000000001190.

219

Heyderman RS, Madhi SA, French N, et al. Group B streptococcus vaccination in pregnant women with or without HIV in Africa: a non-randomised phase 2, open-label, multicentre trial. Lancet Infect Dis 2016;16(5):546–55. doi:10.1016/S1473-3099(15)00484-3.

220

Swamy GK, Metz TD, Edwards KM, et al. Safety and immunogenicity of an investigational maternal trivalent group B streptococcus vaccine in pregnant women and their infants: Results from a randomized placebo-controlled phase II trial. Vaccine 2020;38(44):6930–40. doi:10.1016/j.vaccine.2020.08.056.

221

Madhi SA, Cutland CL, Jose L, et al. Safety and immunogenicity of an investigational maternal trivalent group B streptococcus vaccine in healthy women and their infants: a randomised phase 1b/2 trial. Lancet Infect Dis 2016;16(8):923–34. doi:10.1016/S1473-3099(16)00152-3.

222

Vress D. Future vaccines in pregnancy. Best Pract Res Clin Obstet Gynaecol 2021;76:96–106. doi:10.1016/j.bpobgyn.2021.03.009.

223

Legardy-Williams JK, Carter RJ, Goldstein ST, et al. Pregnancy Outcomes among Women Receiving rVSVΔ-ZEBOV-GP Ebola Vaccine during the Sierra Leone Trial to Introduce a Vaccine against Ebola. Emerg Infect Dis 2020;26(3):541–8. doi:10.3201/eid2603.191018.

224

Samai M, Seward JF, Goldstein ST, et al. The Sierra Leone Trial to Introduce a Vaccine Against Ebola: An Evaluation of rVSV∆G-ZEBOV-GP Vaccine Tolerability and Safety During the West Africa Ebola Outbreak. J Infect Dis 2018;217(suppl_1):S6-S15. doi:10.1093/infdis/jiy020.

225

Juan-Giner A, Tchaton M, Jemmy J-P, et al. Safety of the rVSV ZEBOV vaccine against Ebola Zaire among frontline workers in Guinea. Vaccine 2019;37(48):7171–7. doi:10.1016/j.vaccine.2018.09.009.

226

Halperin SA, Arribas JR, Rupp R, et al. Six-Month Safety Data of Recombinant Vesicular Stomatitis Virus-Zaire Ebola Virus Envelope Glycoprotein Vaccine in a Phase 3 Double-Blind, Placebo-Controlled Randomized Study in Healthy Adults. J Infect Dis 2017;215(12):1789–98. doi:10.1093/infdis/jix189.

227

Agnandji ST, Fernandes JF, Bache EB, et al. Safety and immunogenicity of rVSVΔG-ZEBOV-GP Ebola vaccine in adults and children in Lambaréné, Gabon: A phase I randomised trial. PLoS Med 2017;14(10):e1002402. doi:10.1371/journal.pmed.1002402.

228

Krubiner CB, Faden RR, Karron RA, et al. Pregnant women & vaccines against emerging epidemic threats: Ethics guidance for preparedness, research, and response. Vaccine 2019. doi:10.1016/j.vaccine.2019.01.011.

229

Kochhar S, Edwards KM, Ropero Alvarez AM, et al. Introduction of new vaccines for immunization in pregnancy – Programmatic, regulatory, safety and ethical considerations. Vaccine 2019;37(25):3267–77. doi:10.1016/j.vaccine.2019.04.075.

230

Sobanjo-Ter Meulen A, Munoz FM, Kaslow DC, et al. Maternal interventions vigilance harmonization in low- and middle-income countries: Stakeholder meeting report; Amsterdam, May 1–2, 2018. Vaccine 2019;37(20):2643–50. doi:10.1016/j.vaccine.2019.03.060.

231

Munoz FM, Van Damme P, Dinleyici E, et al. The Fourth International Neonatal and Maternal Immunization Symposium (INMIS 2017): Toward Integrating Maternal and Infant Immunization Programs. mSphere 2018;3(6). doi:10.1128/mSphere.00221-18.

232

Krishnaswamy S, Lambach P, Giles ML. Key considerations for successful implementation of maternal immunization programs in low and middle income countries. Hum Vaccin Immunother 2019;15(4):942–50. doi:10.1080/21645515.2018.1564433.

233

Berrueta M, Ciapponi A, Bardach A, et al. Maternal and neonatal data collection systems in low- and middle-income countries for maternal vaccines active safety surveillance systems: A scoping review. BMC Pregnancy Childbirth 2021;21(1):217. doi:10.1186/s12884-021-03686-9.

234

Sobanjo-ter Meulen A, Abramson J, Mason E, et al. Path to impact: A report from the Bill and Melinda Gates Foundation convening on maternal immunization in resource-limited settings; Berlin – January 29–30, 2015. Vaccine 2015;33(47):6388–95. doi:10.1016/j.vaccine.2015.08.047.

235

McNeil MM, Gee J, Weintraub ES, et al. The Vaccine Safety Datalink: successes and challenges monitoring vaccine safety. Vaccine 2014;32(42):5390–8. doi:10.1016/j.vaccine.2014.07.073.

236

Neophytou AM, Kioumourtzoglou M-A, Goin DE, et al. Addressing special cases of bias that frequently occur in perinatal epidemiology. Int J Epidemiol 2021;50(1):337–45. doi:10.1093/ije/dyaa252.

237

Fell DB, Dimitris MC, Hutcheon JA, et al. Guidance for design and analysis of epidemiological studies of fetal and newborn outcomes following COVID-19 vaccination during pregnancy. Vaccine 2021;https://www.doi:https://doi.org/10.1016/j.vaccine.2021.02.070.

238

Savitz DA, Fell DB, Ortiz JR, et al. Does influenza vaccination improve pregnancy outcome? Methodological issues and research needs. Vaccine 2015;33(47):6430–5. doi:10.1016/j.vaccine.2015.08.041

Online Study Assessment Option
All readers who are qualified doctors or allied medical professionals can now automatically receive 2 Continuing Professional Development credits from FIGO plus a Study Completion Certificate from GLOWM for successfully answering 4 multiple choice questions (randomly selected) based on the study of this chapter.
Medical students can receive the Study Completion Certificate only.

 

(To find out more about FIGO’s Continuing Professional Development awards programme CLICK HERE)