Next Article in Journal
SIV Infection Regulates Compartmentalization of Circulating Blood Plasma miRNAs within Extracellular Vesicles (EVs) and Extracellular Condensates (ECs) and Decreases EV-Associated miRNA-128
Next Article in Special Issue
Congenital SARS-CoV-2 Infection in Two Neonates with Confirmation by Viral Culture of the Placenta in One Case
Previous Article in Journal
The Kinetics of Humoral and Cellular Responses after the Booster Dose of COVID-19 Vaccine in Inflammatory Arthritis Patients
Previous Article in Special Issue
Four Waves of the COVID-19 Pandemic: Comparison of Clinical and Pregnancy Outcomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunisation against COVID-19 in Pregnancy and of Women Planning Pregnancy

by
Justin C. Konje
1,2,3,*,
Mariam Al Beloushi
4,5 and
Badreldeen Ahmed
1,2,5
1
Feto-Maternal Centre Al Markhiya, Doha P.O. Box 34181, Qatar
2
Obstetrics and Gynecology Department, Weill Cornell Medicine Qatar, Doha P.O. Box 24144, Qatar
3
Obstetrics and Gynaecology, Department of Health Sciences, University of Leicester, Leicester LE2 7LX, UK
4
Women’s Wellness and Research Centre, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
5
Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(3), 621; https://doi.org/10.3390/v15030621
Submission received: 23 October 2022 / Revised: 15 February 2023 / Accepted: 16 February 2023 / Published: 24 February 2023
(This article belongs to the Special Issue SARS-CoV-2 in Pregnancy and Reproduction)

Abstract

:
Following reports of the first human SARS-CoV2 infection in December 2019 from Wuhan Province, China, there was such rapid spread that by March 2021, the World Health Organization (WHO) had declared a pandemic. Over 6.5 million people have died from this infection worldwide, although this is most likely an underestimate. Until vaccines became available, mortality and severe morbidity were costly in terms of life lost as well as the cost of supporting the severely and acutely ill. Vaccination changed the landscape, and following worldwide adoption, life has gradually been returning to normal. The speed of production of the vaccines was unprecedented and undoubtedly ushered in a new era in the science of fighting infections. The developed vaccines were on the already known platforms for vaccine delivery: inactivated virus, virus vector, virus-like particles (VLP) subunit, DNA and mRNA. The mRNA platform was used for the first time to deliver vaccines to humans. An understanding of these platforms and the pros and cons of each are important for clinicians who are often challenged by the recipients on the advantages and risks of these vaccines. These vaccines have so far and reassuringly been shown to be safe in reproduction (with no effect on gametes) and pregnancy (not associated with congenital malformations). However, safety remains paramount and continuing vigilance is critical, especially against rare fatal complications such as vaccine-induced thrombocytopenia and myocarditis. Finally, the waning immunity months after vaccination means repeated immunisation is likely to be ongoing, but just how often and how many such revaccinations should be recommended remains uncertain. Research into other vaccines and alternate delivery methods should continue as this infection is likely to be around for a long time.

1. Introduction

Coronaviruses cause various infections in avians and mammals. They specifically attack the respiratory system causing damage, which in some cases can cause death [1]. Three new coronaviruses related severe outbreaks of zoonotic origin have occurred in the last two decades—the Severe Acute Respiratory Syndrome Coronavirus-1 (SARS-CoV-1) in 2002–2003, the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) in 2012 and recently the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) in 2019, which was declared a pandemic by the WHO in March 2020 [2] and has been the most devastating. As of September 2022, more than 600 million cases have been confirmed, with over 6.5 million deaths, giving a mortality rate of 1.18% [3], but this is thought to be an underestimation as testing and reporting of deaths have not been universal, and furthermore, data on asymptomatic cases are often not captured.
The Severe Acute Respiratory Syndrome Coronoavirus-2 (SARS-CoV-2) is a novel enveloped ribonucleic acid (RNA) betacoronavirus belonging to the subfamily Coronavirinae in the Coronoviridae family [4,5,6]. Within this family of viruses are four genera—Alphacoronoavirus, Betacoronavirus, Gammacoronavirus and Deltacoronavirus [7]. Other members of the family that have caused infections in humans with varying severity include SARS-CoV-1 and MERS-CoV.
SARS-CoV-2 is spherical in shape with a core shell and surface with proteins, some of which project like the spikes of a crown, hence named corona (crown in Latin). The proteins are the spike (S) which is responsible for attachment to the host cell membrane receptor, followed by fusion and finally entry into the host cell, the membrane (M), which is the most abundant, the envelope (E) is characterised by a comparatively higher basic reproductive number (Ro) [Ro is referred to as the reproductive number. It is used to gauge how infectious a contagious infection is. The basic reproductive number (Ro) of COVID-19 has been initially estimated by the WHO to range between 1.4 and 2.5] which explains its rapid spread worldwide, and the N protein, which makes part of the helical nucleocapsid that includes the genome RNA [8]. The virus has a single-stranded positive-sense RNA of around 30 kb that is non-segmented. Phylogenetic analysis has shown that SARS-CoV-2 has an 80% genetic sequence similarity to that of SARS-CoV-1 and a 96.2% similarity to that of the bat coronavirus RaTG13 [9]. SARS-CoV-2 has a higher reproductive number (Ro) than SARS-CoV-1, implying that it spreads more efficiently [10].
Although the total number of people infected with SARS-CoV-2 worldwide continues to increase, the rate of increase has probably peaked and may indeed be falling. Pregnant women, by virtue of their relatively immunosuppressed status, tend to develop severe consequences from infections that ordinarily will not pose problems in the non-pregnant, and indeed have been shown to have a tendency to develop severe complications with SARS-CoV-2 [11]. Like most viral infections, effective treatment has been elusive, and the only approach that has been adopted globally to reduce the spread and burden of SARS-CoV-2 infection is immunisation. Concordant with the wide adoption of immunisation against SARS-CoV-2 are concerns about safety in pregnancy and around reproduction.

2. COVID-19 Vaccination

The severe health (morbidity and mortality), social and economic consequences of the Coronaviruses Infectious Diseases-2019 (COVID-19) pandemic led to the development of vaccines with unparalleled speed. These developments were across all six platforms for vaccine delivery (i.e., inactivated virus vaccines, live attenuated virus vaccines, recombinant viral vector vaccines, recombinant subunit vaccines, DNA vaccines and RNA vaccines) [12]. As of early 2022, at least seven different vaccines across three of these platforms had been approved in several countries. The most commonly offered ones are the Pfizer-BioNTech vaccine (mRNA), Moderna vaccine (mRNA), the Johnson & Johnson’s Janssen Ad26.COV2.S vaccine (adenoviral vector), the AstraZeneca, University of Oxford and Serum Institute of India ChAsOx1 CoV-19 vaccine (adenoviral vector-based), the Chinese Sinovac Research and Development CoronaVac and the Russian Sputnik V (Gam-COVID-Vac), an adenovirus vector vaccine. For most of these vaccines, phase II and III clinical trials did not include pregnant women, and vaccine phobia plus negative social media reports significantly affected uptake in general and more so in vulnerable groups, which include pregnant women.
The first of the approved vaccines is a messenger RNA (mRNA) vaccine made by in-vitro transcription of a target gene from a linearised DNA template [12]. Vaccines of this type, mRNA, are non-infectious and non-integrating. Hence, they are unable to cause infectious or insertional mutagenesis. They are also easily degraded, with a half-life that can be regulated through modification to the template or delivery methods [13]. Antigen-specific cellular and humoral immune responses are enhanced by the strong immune-stimulatory effect and intrinsic adjuvant activity of the in-vitro transcribed mRNA [14,15,16,17]. Other advantages of this platform include no anti-vector response (it is the minimal genetic vector), large amounts of antigen can be produced from a minimal vaccine dose since mRNA can be replicated intracellularly, and mRNA is relatively easy to produce compared to recombinant protein and live attenuated virus vaccines. Furthermore, the manufacturing process for these types of vaccines can be standardised to produce many encoded protein immunogens, making mRNA the preferred platform for rapid response during a pandemic [12]. However, some mRNA vaccines may induce potent type I interferon reactions, especially in those at an increased risk of autoimmune response. Furthermore, the lipid nano peptide (NLP) used for packaging the mRNA means the extracellular RNA they deliver can induce thrombosis [12]. Indeed, vaccine-induced thrombosis (VIT) has been reported as a complication in a small number of cases [18]. In pregnancy, this risk may be increased because of the thrombotic state induced by the physiological changes of pregnancy [1]. The Pfizer-BioNtech and Moderna COVID-19 vaccines are lipid nanoparticle-formulated mRNA vaccines which do not contain ingredients that are known to be harmful to pregnant women or to the fetus. These vaccines do not enter the nucleus and do not alter human DNA in recipients. As a result, the mRNA in the vaccine is translated and transcribed by the body to produce the spike protein, which then acts as an intracellular antigen to stimulate the immune response. The mRNA in the vaccine is normally degraded within a few days and cannot be incorporated into the host genome. Thus, it cannot cause any genetic changes [12].
The second platform for COVID-19 vaccines that received approval for use is the viral vector platform. The vaccines of this platform are the Johnson and Jonson-Janssen, Astra-Zeneca and Russian Sputnik vaccines [19]. When attenuated viruses are used as platforms for delivering vaccines for highly infectious pathogens such as SARS-CoV-2, Ebola virus and Zika virus, there is the danger that these viruses may revert to their pathogenic forms and cause disease (i.e., recover their virulence) [20]. Therefore, recombinant viral vectors are developed that mimic the natural pathogen but are not virulent. This is achieved by cloning the target pathogen into an avirulent viral host—in this case, an adenovirus. The adenovirus (host-virus genome) carries out all the viral transmission and amplification functions, including antigen production, to present the chosen antigen to the patient’s immune system [12]. The vaccines in this category introduce DNA for the SARS-CoV-2 spike protein into the cells. The DNA is first copied into mRNA, which is then used to produce copies of the spike protein that stimulates the body’s immune system to generate antibodies. The main advantage of this platform is the fact that since it presents protective antigen epitopes in the context of a live, replicating virus without concern about reversion to a pathogenic state, it induces a more robust immune response and allows for antigen sparing as the viral vectors have limited nucleotide activity [12]. A disadvantage of this platform is that the immunogenicity induced resulting from both the encoded antigen and the viral vector antigen may lower or modify the vaccine’s efficacy by diverting immune responses away from the target antigens [12]. In the unlikely event that an individual has pre-existing immunity to the viral vector, it can lead to premature clearance of the vaccine before an immune response can be mounted to the antigen of interest carried by the vector making it ineffective [21,22].
The third platform that COVID-19 vaccines belong to is the antigenic sub-unit, with examples in this group including Ahnui Zhifei Longcom Biopharmaceutical Novovax, GSK Sanofi Pasteur and United Biomedical COVAXX vaccines. These do not contain any live component of the pathogen (SARs-CoV-2) but contain an antigenic sub-unit of the pathogen that induces the protective immune response [23]. Vaccines on this platform thus include only parts of the virus or bacteria necessary to cause a protective immune response; hence side effects are less common (local ones are negligible) since the sub-units do not contain other molecules of the pathogen [23]. Another advantage of these groups of vaccines is that they are less likely to induce an eosinophilic immunopathology or antibody-mediated disease enhancement, hence are safe for use in those who are immunosuppressed [24].
A summary of the information on the four vaccines with published outcomes following use in pregnancy is shown in Table 1. The mRNA vaccines have been shown to have a higher efficacy in the prevention of symptomatic illness with efficacies of about 95% compared to that of about 70% with adenoviruses vaccines. With regard to severe COVID-19, there are no obvious differences in efficacies.

3. Immunological Response to Vaccination in Pregnant Woman

Of the four vaccines that have been used extensively, three are administered as two or more vaccine doses three to twelve weeks apart, while the 4th, the Johnson and Johnson-Janssen vaccine, is administered once. Consideration should, however, also be given for a booster dose about six months later for all of them. While most studies on the immune response have been on non-pregnant women, Prabhu et al. [36], Collier and McMahan [37], and Gray et al. [38] have all shown that the immunological response in pregnant women from measured IgG increase is similar to that in non-pregnant women. Interestingly IgG has been measured in the cord blood and breastmilk of mothers who were vaccinated during pregnancy in New York [36]. In the New York study, it was shown that antibody production (IgG) started five days after immunization. By day 16, the antibody production was detectable in cord blood. The implication of these findings is that if mothers are vaccinated at least 16 days prior to delivery, the baby should be born with passive immunity, which would then be supplemented with the antibodies secreted in breast milk if they breastfeed. Children of mothers vaccinated after 20 weeks of gestation have been shown to have a lower risk of hospitalisation compared to those of mothers vaccinated before 20 weeks (85% vs. 32%). These results suggest a declining passive immunity as the antibody levels in the mother wane [39]. Several studies have shown a decline in antibodies by 4–6 months after the booster dose, and following these studies, 3rd and 4th booster doses have been recommended, especially for vulnerable groups. It would therefore seem logical to consider a 3rd dose in women who are vaccinated prior to pregnancy and have the second booster dose early in pregnancy or, indeed, prior to pregnancy. Table 2 is a summary of the data from some of the studies on the immunogenicity of COVID-19 vaccination in pregnancy. The data from these studies suggest that the early third trimester may be the optimal time for a booster vaccine dose (supporting our recommendation above) to allow for optimal maternal-to-fetal antibody transfer for appropriate neonatal immunity. While the transplacental transfer of antibodies starts in the second trimester, it is most efficient in the third trimester [40].

4. COVID-19 Vaccination and Pregnancy

The effectiveness of vaccines in reducing risk of moderate to severe illness, hospitalisation, and deaths from COVID-19 has been shown to be extremely high (69–95%) [1,59,60,61,62], although immunity has been demonstrated to wane 4–6 months after booster doses [63]. Furthermore, the regular emergence of new/mutated variants has been regarded as a potential factor affecting vaccine efficacy [64,65].
Concerns about safety with the rapidity with which these vaccines were developed have, in some cases, resulted in vaccine hesitance, especially in vulnerable groups such as pregnant women and others at risk. Several studies, most of which are cohort studies, have investigated adverse pregnancy outcomes in women following COVID-19 vaccination (Table 3) [42,44,45,50,53,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82]. The best evidence for the safety of COVID-19 vaccination in pregnancy comes from the USA Centers for Disease Control and Prevention (CDC) tracking of mRNA vaccine recipients who were either pregnant at the time of vaccination (V-safe) or shortly after. In the latest report published on 35,691 women v-safe participants aged 16 to 54 years identified as pregnant who received the Pfizer-Biotech Vaccine, injection-site pain was reported more frequently among pregnant persons than among non-pregnant women, whereas headache, myalgia, chills, and fever were reported less frequently. Of those who completed their pregnancies, 115 (13.9%) were pregnancy losses, and 712 (86.1%) were live births. Adverse neonatal outcomes included preterm birth (9.4%) and small size for gestational age (3.2%); no neonatal deaths were reported. Although not directly comparable, calculated proportions of adverse pregnancy and neonatal outcomes in persons vaccinated against COVID-19 who had a completed pregnancy were similar to incidences reported in studies involving pregnant women that were delivered before the COVID-19 pandemic [70]. From all these studies, we are able to conclude that there have so far been no safety concerns in pregnant women who received especially mRNA and, to a lesser extent, the adenovirus COVID-19 vaccines. However, more longitudinal follow-up, including follow-up of large numbers of women vaccinated early in pregnancy, is necessary to inform maternal, pregnancy, and infant outcomes [1,70]. These follow-up studies are continuing, and hopefully, some will be on the adenovirus platform vaccines. Overall, the evidence from these supports the conclusion that the risks of COVID-19 in the un-vaccinated outweigh those of the vaccines.

5. COVID-19 Vaccination and Transplacental Transfer of Antibodies

Maternal antibodies (IgG) produced in response to infections cross the placenta and provide passive immunity to the baby when exposed to the infective pathogen after birth. Some vaccines, when administered to pregnant women, offer protection to both the mother and infant as the induced antibodies also cross the placenta. Examples include the TDap vaccine (against acellular pertussis, diphtheria and tetanus toxoids) [83,84]. With regards to COVID-19 vaccines, there is evidence that generated antibodies from vaccinated mothers reach their fetuses transplacentally. In a study evaluating SARS-COV-2 antibody titre in cord blood from 16 neonates whose mothers received the SARS-COV-2 mRNA vaccine (BNT162b2), all maternal and cord blood samples were positive for SARS-CoV-2 spike (S) protein antibody. The anti-S antibody titre in the cord blood increased the longer the interval (weeks) between the first dose of the vaccine and delivery [47]. In another study, there was a positive correlation between the number of weeks from the first or second vaccine and the ratio of the umbilical cord to the maternal SARS-CoV-2 anti-S antibody [85]. Mithal et al. studied 27 vaccinated pregnant women (23 had received the BNT162b2/Pfizer or Spikevax (Moderna) vaccine, four had received an unknown SARS-CoV-2 vaccine) and their infants (28 including a pair of twins). Of these women, 74% received both doses before delivery. IgG against SARS-CoV-2 was present in all but three of the infants whose mothers had received their first dose of vaccine less than three weeks before delivery. They concluded that “receiving both vaccine doses before delivery and longer latency from vaccination to delivery was associated with a higher IgG concentration in infants and stronger immunity” [52]. This result was also confirmed in other studies [86,87]. Prabhu et al. studied 122 pregnant women, of whom 55 and 67 had received their first and second mRNA vaccine doses, respectively, before delivery. They showed that maternal antibody production started five days after the first vaccine dose, and transplacental transfer to the fetus began after the 16th day post-vaccination. These findings emphasise the importance of a second vaccination dose, with the data showing that 99% of women receiving both doses had IgG antibodies in their cord blood samples; however, only 44% of women who had received one dose of the vaccine had cord blood IgG antibodies against SARS-CoV-2. They concluded from this that transplacental antibody transfer rises with the increase in weeks between the second vaccine and birth [36]. It would seem from these data that an interval of four weeks between maternal vaccination and delivery is necessary to enable adequate antibodies to be generated and transferred to the fetus for the protection of the infant against SARS-CoV-2. Since adequate antibody production is achieved two weeks after the second dose of the vaccine (bearing in mind that there usually are at least 21 or 28 days between first and second doses with different vaccines) and that transplacental antibody transfer begins approximately at 16 weeks of gestation, vaccinating pregnant women at the beginning of the second trimester might lead to the highest levels of antibodies being transferred to the newborn [88]. Comparisons of vaccination in early and late third trimesters showed that early vaccination increases antibody transfer through the placenta and increases neonatal neutralizing antibody levels [89]. With regards to the transplacental antibody transfer, the highest transfer occurred in women vaccinated in the first followed by the second trimester; antibody titres declined more when the vaccination was in the first trimester. This would imply that a booster vaccine in the third trimester would be of maximum benefit to the fetus [90]. Data on the protective effect of vaccine-induced antibodies against SARS-COV-2 are limited. In their study, Atyeo et al. [38] did show that the IgG antibody levels against the S protein in maternal and cord samples were not statistically different although cord levels were lower.

6. COVID-19 Vaccination and Breastfeeding

Antibodies against SARS-CoV-2 are present in breast milk following maternal infection and offer passive immunity to the newborn. The same will apply if vaccine-induced antibodies are also present in breastmilk. In a study of fourteen lactating women who received two doses of the BNT162b2 (Pfizer) vaccine, it was found that 3–7 days after the second dose of the vaccine, there was a peak in the amount of SARS-CoV-2 IgA and IgG antibodies in breastmilk. IgG remained stable until 4–6 weeks, but IgA levels fell significantly. There was also a minimal transfer of vaccine mRNA into breastmilk. Breastfed infants showed no adverse effects during the first 28 days of follow-up [91]. These findings were further confirmed by another study which found IgA and IgG antibodies in breastmilk as early as two and four weeks after the first dose of the vaccine, respectively [92]. Studies of post-vaccination lactating women who received the mRNA vaccines similarly found SARS-CoV-2 IgA and IgG antibodies in breastmilk samples that were absent before vaccination [93]. A recent systematic review concluded that after the first vaccine, 64% and 30% of breastmilk samples were positive for IgA and IgG, respectively, and these rose to 70% and 91% after the second dose [94]. Evidence on the ability of these antibodies to neutralise antigens is limited. In an in-vitro study of 34 cases, the neutralizing capacity of antibodies in milk samples obtained pre- and post-SARS-COV-2 infection (with IgA and IgG antibodies) were compared; 62% of the post-infection samples compared to none of the pre-infection samples were able to neutralise antigen suggesting that these antibodies are capable of providing passive immunity [95]. With regards to vaccine-generated antibodies, a study by Young et al. showed that 60% and 85% of human milk samples had neutralizing capacity against the wild-type SARS-CoV-2 virus after the first and second vaccine doses, respectively; of the post-infection samples, 80% and 100% showed neutralizing capacity after 28 and 90 days, respectively. Perez et al. [96] showed that the neutralizing capacity of breastmilk three and six months post-vaccination was 83.3%, 70.4% and 25%, respectively and correlated strongly with IgG levels in the milk [97]. Taken together, we can conclude that vaccinated-generated antibodies cross to the baby through breastmilk and that these antibodies are able to offer passive immunity to the baby. These additional benefits of vaccination should form part of the counselling of pregnant women.

7. Vaccination and Reproduction

SARS-CoV-2, through its spike (S) protein, binds to the angiotensin-converting enzyme 2 (ACE-2) receptor [98] after it enters the body. The interaction between the virus and the receptor is mediated by the transmembrane protease serine 2 (TMPRSS2) or cathepsins B and L (encoded by the genes CTSB and CTSL, respectively) when TMPRSS2 is absent [99]. TMPRSS2 and ACE-2 receptors [100] have been localised in parts of the female genital tract, such as the ovaries, where ACE-2 receptor activity is expressed in relation to folliculogenesis and maturation, steroid synthesis and ovulation. The distribution of these receptors is, at best, described as sparse. Because of their presence, therefore these parts of the female reproductive tract are a potential target for SARS-CoV-2. There is the potential for the SARs virus to cause damage via the ACE2 /TMPRSS2 pathway [101,102]. In males, ACE-2 receptors and TMPRSS2 (targets of the virus) are expressed on testicular tissue, again implying a potential of damage by SARS-CoV-2 to the testis and possibly having an adverse effect on its function [103,104]. Furthermore, testosterone has been shown to facilitate SARS-CoV-2 spread via activation of TMPRSS2 [105], thus increasing the attractiveness of the virus onto the testicular tissue. SARS-CoV-2, through binding to the ACE2 receptor, may potentially cause orchitis and possible testicular atrophy and sub-fertility [106]. Furthermore, there have been reports of isolation of SARS-CoV-2 in the semen of infected males [92], raising the possibility of sexual transmission. Recent studies have shown that COVID-19 (even moderate disease) causes adverse changes in semen parameters, but these were found to recover quickly after the infection [107,108].
Clinical trials with COVID-19 vaccines neither included pregnant women nor those undergoing fertility treatment or, indeed, those in early pregnancy. Hence the impact/effect of these vaccines on gametes and early embryos is unknown. In a joint statement, however, the International Federation of Fertility Societies (IFFS) and the European Society of Human Reproduction and Embryology (ESHRE) advised that women planning to conceive in an environment with ineffective control of COVID-19 and/or with limited resources for vaccination should adopt measures to mitigate the risk of exposure and defer pregnancy pending improvement with regards to SARS-CoV-2 infection or until vaccination is available, thus indicating that vaccination is not contra-indicated in these women [109,110,111,112,113,114,115]. In a recent prospective study of the effect of the COVID-19 vaccine on sperm parameters, Gonzalez et al. [116] not only show that there were no adverse effects with the mRNA vaccine but that there seems to be an improvement in parameters in oligozoospermic men. Furthermore, the vaccine has been shown to have no effect on response to ovulation stimulation as well as parameters of successful stimulation [117,118,119], and in animals, no adverse effects on reproduction have been reported [27,120].

8. Complications of COVID-19 Vaccines

Side effects of COVID-19 vaccines can be localised or systemic. The most commonly-reported ones are headache, fatigue, muscle and joint pain, fever and chills and pain at the site of injection [121]. From the CDC’s Vaccine Adverse Events Reporting System (VAERS) uncommon side effects include anaphylaxis reported in approximately five cases per one million vaccine doses administered, thrombocytopenia syndrome (TTS) a rare but serious adverse event after the Johnson and Johnson-Janssen (J & J/Janssen) COVID-19 vaccination, reported in approximately four cases per one million doses administered, Guillain-Barre Syndrome (GBS) (a rare disorder where the body’s immune system damages nerve cells, causing muscle weakness and sometimes paralysis) reported mainly in men aged 50 years and older who received the J & J/Janssen COVID-19 vaccine and myocarditis and pericarditis mostly reported after Pfizer-BioNTech or Moderna (mRNA COVID-19 vaccines), particularly in male adolescents and young adults, the rates being highest after the second dose: 70.7 cases per one million doses of Pfizer-BioNTech in those age 12–15 years, 105.9 cases per one million doses of Pfizer-BioNTech in those aged 16–17 years and 52.4 cases and 56.3 cases per million doses of Pfizer-BioNTech and Moderna in those age 18–24 years, respectively [122,123].
Of these rare complications, the prothrombotic syndrome (vaccine-induced thrombotic thrombocytopenia (VITT) or vaccine-induced prothrombotic immune thrombocytopenia (VIPIT) has the potential to be serious in pregnancy (Johnson & Johnson-Janssen and AstraZeneca, University of Oxford) [124]. This is thought to be caused by immunoglobin (Ig) antibodies that bind to platelet factor 4 (PF4), also known as CXCL4. Platelets are activated following this binding to PF4 with the resultant stimulation of the coagulation system and the ensuing clinically significant thromboembolic complications [125]. The thrombosis caused by these vaccines includes cerebral venous thrombosis, splanchnic vein thrombosis (e.g., mesenteric vein, portal vein, splenic and hepatic vein), adrenal vein thrombosis (which may present as adrenal failure), pulmonary embolism and arterial thrombosis (including ischaemic stroke and acute limb ischaemia) [124,125]. Although a very rare complication, it is associated with mortality. Since pregnancy per se more than doubles the risk of VTE (because of the hypercoagulability state secondary to physiological changes in pregnancy) and severe SARS-CoV-2 doubles the risk of VTE in pregnant women with severe illness, it would be prudent to avoid the adenovirus vaccines in those who are planning a pregnancy or are pregnant and have risk factors for venous thromboembolism (VTE).

9. Recommendations

From these data and the risk-benefit analysis, various bodies and authorities, including the Centers for Disease Control and Prevention (CDC) [126], the Society for Maternal-Fetal Medicine (SMFM) [127,128], the American College of Obstetricians and Gynecologists (ACOG) [29,129], the International Federation of Gynaecology and Obstetrics (FIGO) and the Royal College of Obstetricians and Gynaecologists, on balance all recommend that COVID-19 vaccines should be offered to both pregnant and lactating mothers [130]. Several countries have also given regulatory approval for their use in pregnancy and in women planning a pregnancy or undergoing treatment for infertility. The recommendations and positions of the various societies on COVID-19 vaccination in pregnancy and reproduction are shown in Table 4. In counselling women for vaccination, particular attention should be given to those who are at high-risk. These include women from ethnic minorities, overweight/obese and/or with co-morbidities [131].

10. Conclusions

The COVID-19 pandemic undoubtedly changed all aspects of clinical practice and, indeed, life. In the absence of effective treatment, vaccination remains the only option for controlling the infection. The rapidity with which these vaccines were developed left several questions in the vaccine doubters and, indeed, was partly responsible for the reluctance of some to receive them. The benefits of vaccination greatly outweigh the side-effects. The available accumulated evidence since the vaccines were introduced overwhelmingly confirms their safety in all stages of pregnancy and in those planning pregnancies. It is the recommendation of most international and national societies that women planning a pregnancy or are pregnant or lactating should be offered vaccination against COVID-19. Where vaccination was received before pregnancy or in early pregnancy, consideration should be given to a booster dose in the third trimester to allow for the maximum transplacental transfer of antibodies to the baby for passive immunity afterbirth. While the mRNA vaccines are preferred by many, the J & J Janssen vaccine as well as the protein platform vaccine, are also acceptable options. We believe that while this is indeed a good practice to offer these vaccines, caution must continue to be exercised going forward, and more data must be collected and regularly reviewed, especially that which includes vulnerable groups like pregnant women, those who are immunocompromised, children and the elderly. With the levels of antibodies waning after vaccination, questions remain on how often this should be administered and whether repeated vaccinations are associated with more complications.

Author Contributions

Conceptualization, J.C.K. and B.A.; methodology, J.C.K. and M.A.B.; writing—original draft preparation, M.A.B., J.C.K. and B.A.; writing—review and editing, J.C.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hamad, W.A.B.; Al Beloushi, M.; Ahmed, B.; Konje, J.C. Severe acute respiratory syndrome (SARS) coronavirus-2 infection (COVID-19) in pregnancy—An overview. Eur. J. Obstet. Gynecol. Reprod. Bio. 2021, 263, 106–116. [Google Scholar] [CrossRef]
  2. WHO. WHO Director-General’s Opening Remarks at the Media Briefing on COVID-19—11 March 2020; World Health Organization (WHO): Geneva, Switzerland, 2020.
  3. Available online: https://coronavirus.jhu.edu/map.html (accessed on 24 September 2022).
  4. Cui, J.; Li, F.; Shi, Z.-L. Origin and evolution of pathogenic coronaviruses. Nat. Rev. 2019, 17, 181–192. [Google Scholar] [CrossRef] [Green Version]
  5. Weiss, S.R.; Navas-Martin, S. Coronavirus pathogenesis and the emerging pathogen severe acute respiratory syndrome coronavirus. Microbiol. Mol. Rev. 2005, 69, 635–664. [Google Scholar] [CrossRef] [Green Version]
  6. Hasoksuz, M.; Alekseev, K.; Vlasova, A.; Zhang, X.; Spiro, D.; Halpin, R.; Wang, S.; Ghedin, E.; Sail, L.J. Biologic, antigenic, and full-length genomic characterization of a bovine-like coronavirus isolated from a giraffe. J. Virol. 2007, 81, 4981–4990. [Google Scholar] [CrossRef] [Green Version]
  7. Christian, M.D.; Poutanen, S.M.; Loutfy, M.R.; Muller, M.P.; Low, D.E. Severe acute respiratory syndrome. Clin. Infect. Dis. 2004, 38, 1420–1427. [Google Scholar] [CrossRef] [Green Version]
  8. Yan, R.; Zhang, Y.; Li, Y.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef] [Green Version]
  9. Cevik, M.; Bamford, C.G.G.; Ho, A. COVID-19 pandemic—A focused review for clinicians. Clin. Microbiol. Infect. 2020, 26, 842–847. [Google Scholar] [CrossRef]
  10. Liu, J.; Xie, W.; Wang, Y.; Xiong, Y.; Chen, S.; Han, J.; Wu, Q. A comparative overview of COVID-19, MERS and SARS: Review article. Int. J. Surg. 2020, 81, 1–8. [Google Scholar] [CrossRef]
  11. Allotey, J.; Fernandez, S.; Bonet, M.; Stallings, E.; Yap, M.; Kew, T.; Zhou, D.; Coomar, D.; Sheikh, J.; Lawson, H.; 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. [Google Scholar] [CrossRef]
  12. Verdecia, M.; Kokai0Kun, J.F.; Kibbey, M.; Acharya, A.; Venema, J.; Atouf, F. COVID-19 vaccine platforms: Delivering on promise? Hum. Vaccin. Immunother. 2021, 17, 2873–2893. [Google Scholar] [CrossRef]
  13. Jackoson, N.A.C.; Kester, K.E.; Casimito, D.; Gurunanthan, S.; DeRosa, F. The promise of mRNA vaccines: A biotech and industrial perspective. NPJ Vaccines 2020, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  14. Hoerr, I.; Obst, R.; Rammensee, H.G.; Jung, G. In vivo application of RNA leads to induction of specific cytotoxic T lymphocytes and antibodies. Eur. J. Immunol. 2000, 30, 1–7. [Google Scholar] [CrossRef]
  15. Bourquin, C.; Schmidt, L.; Hornung, V.; Wurzenberger, C.; Anz, D.; Sandholzer, N.; Schreiber, S.; Voelkl, A.; Hartmann, G.; Endres, S.; et al. Immunostimulatory RNA oligonucleotides trigger an antigen-specific cytotoxic T-cell and IgG2a response. Blood 2007, 109, 2953–2960. [Google Scholar] [CrossRef]
  16. Sander, L.E.; Davis, M.J.; Boekschoten, M.V.; Amsen, D.; Dascher, C.C.; Ryffel, B.; Swanson, J.A.; Müller, M.; Blander, J.M. Detection of prokar- yotic mRNA signifies microbial viability and promotes immunity. Nature 2011, 22, 385–389. [Google Scholar] [CrossRef] [Green Version]
  17. Weissman, D.; Ni, H.; Scales, D.; Dude, A.; Capodici, J.; McGibney, K.; Abdool, A.; Isaacs, S.N.; Cannon, G.; Karikó, K.; et al. HIV gag mRNA transfection of dendritic cells (DC) delivers encoded antigen to MHC class I and II molecules, causes DC maturation, and induces a potent human in vitro primary immune response. J. Immunol. 2000, 15, 4710–4717. [Google Scholar] [CrossRef] [Green Version]
  18. Warkentin, T.E.; Cuker, A. COVID-19, Vaccine-Induced Immune Thrombotic Thrombocytopenia (VITT). Available online: https://www.ncbi.nlm.nih.gov/books/NBK570605/ (accessed on 7 May 2021).
  19. Kallel, H.; Kamen, A.A. Large-scale adenovirus and poxvirus-vectored vaccine manufacturing to enable clinical trials. Biotechnol. J. 2015, 10, 741–747. [Google Scholar] [CrossRef]
  20. Stowe, J.; Andrews, N.; Taylor, B.; Miller, E. No evidence of an increase of bacterial and viral infections following measles, mumps and rubella vaccine. Vaccine 2009, 25, 1422–1425. [Google Scholar] [CrossRef]
  21. Rollier, C.S.; Reyes-Sandoval, A.; Cottingham, M.G.; Ewer, K.; Hill, A.V. Viral vectors as vaccine platforms: Deployment in sight. Curr. Opin. Immunol. 2011, 23, 377–382. [Google Scholar] [CrossRef]
  22. Fausther-Bovendo, H.; Kobinger, G.P. Pre-existing immunity against Ad vectors: Humoral, cellular, and innate response, what’s important? Hum. Vaccin. Immunother. 2014, 10, 2875–2884. [Google Scholar] [CrossRef] [Green Version]
  23. Wang, M.; Jiang, S.; Wang, Y. Recent advances in the production of recombinant subunit vaccines in Pichia pastoris. Bioengineered 2016, 7, 155–165. [Google Scholar] [CrossRef] [Green Version]
  24. Oyston, P.; Robinson, K. The current challenges for vaccine development. J. Med. Microbiol. 2012, 61, 889–894. [Google Scholar] [CrossRef]
  25. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gudman, A.; Lockhaart, S.; Perez, J.L.; Marc, G.P.; Moreira, E.; Zerbini, C.; et al. C4591001 Clinical Trial Group. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
  26. Rosenblum, H.G.; Hadler, S.C.; Moulia, D.; Shimabukuro, T.T.; Su, J.R.; Tepper, N.K.; Ess, K.C.; Woo, E.J.; Mba-Jonas, A.; Alimchandani, M.; et al. Use of COVID-19 vaccines after reports of adverse events among adult recipients of Janssen (Johnson & Johnson) and mRNA COVID-19 vaccines (Pfizer-BioNTech and moderna): Update from the Advisory Committee on Immunization Practices—United States, July 2021. MMWR Morb. Mortal. Wkly Rep. 2021, 70, 1094–1099. [Google Scholar]
  27. Bowman, C.J.; Bouressam, M.; Campion, S.N.; Cappon, G.D.; Catlin, N.R.; Cutler, M.W.; Diekmann, J.; Rhode, C.M.; Sellers, R.S.; Lindermann, C.L. Lack of effects on female fertility and prenatal and postnatal offspring development in rats with BNT162b2, a mRNA-based COVID-19 vaccine. Reprod. Toxicol. 2021, 103, 28–35. [Google Scholar] [CrossRef]
  28. American College of Obstetricians and Gy-Necologists. COVID-19 FAQs for Obstetrician-Gynecologists, Obstetrics. 2020. Available online: https://www.acog.org/clinical-information/physician-faqs/covid-19-faqs-for-ob-gyns-ob-stetrics (accessed on 7 February 2022).
  29. American College of Obstetricians and Gynecologists. COVID-19 Vaccination Considerations for Obstetrice-Gynecologic Care. Practice Advisory. 2020. Available online: https://www.acog.org/clinical/clinical-guidance/practice-advisory/articles/2020/12/covid-19-vaccination-considerations-for-obstetricgynecologic-care (accessed on 7 February 2022).
  30. Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416. [Google Scholar] [CrossRef]
  31. Southwestern Vaccine Science UT, Review Committee. Scientific Review of Moderna COVID-19 Vaccine (mRNA-1273). 2020. Available online: https://AOG.0000000000004438www.utsouthwestern.edu/covid-19/assets/moderna-review.pdf (accessed on 23 October 2022).
  32. Sadoff, J.; Gray, G.; Vandebosch, A.; Cardenas, V.; Shukarev, G.; Grinsztejn, B.; Goepfert, P.A.; Truyers, C.; Fennema, H.; Speissens, B.; et al. Safety and Efficacy of Single-Dose Ad26.COV2.S Vaccine against Covid-19. N. Engl. J. Med. 2021, 384, 2187–2201. [Google Scholar] [CrossRef]
  33. U.S. Food and Drug Administration. Emergency Use Authorization (EUA) of the Janssen COVID-19 Vaccine to Prevent Coronavirus Disease 2019 (COVID-19). 2022. Available online: https://www.fda.gov/media/146304/download (accessed on 23 October 2022).
  34. Voysey, M.; Clemens, S.A.C.; Madhi, S.A.; Weckx, L.Y.; Folegatti, P.M.; Aley, P.K.; Angus, B.; Baillie, V.L.; Barnabas, S.L.; Oxford COVID Vaccine Trial Group; et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2, an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021, 397, 99–111. [Google Scholar] [CrossRef]
  35. Stebbings, R.; Maguire, S.; Armour, G.; Jone, C.; Goodman, J.; Maguire, A.K.; Tang, C.M.; Skellett, V.; Harris, J. Developmental and reproductive safety of AZD1222 (ChAdOx1 nCoV-19) in mice. Reprod. Toxicol. 2021, 104, 134–142. [Google Scholar] [CrossRef]
  36. Prabhu, M.; Murphy, E.A.; Sukhu, A.C. Antibody response to coronavirus disease 2019 (COVID-19) messenger RNA vaccination in pregnant women and transplacental passage into cord blood. Obstet. Gynecol. 2021, 138, 278–280. [Google Scholar] [CrossRef]
  37. Collier, A.Y.; McMahan, K.; Yu, J. Immunogenicity of COVID-19 mRNA vaccines in pregnant and lactating women. JAMA 2021, 325, 2370–2380. [Google Scholar] [CrossRef]
  38. Atyeo, C.; Shook, L.L.; Nziza, N.; Deriso, E.A.; Muir, C.; Baez, A.M.; Lima, R.S.; Demidkin, S.; Brigida, S.; De Guzman, R.M.; et al. COVID-19 booster dose induces robust antibody response in pregnant, lactating, and nonpregnant women. Am. J. Obstet. Gynecol. 2023, 228, 68.e1–68.e12. [Google Scholar] [CrossRef]
  39. Halasa, N.B.; Olson, S.M.; Staat Margaret, M.A.; Newhams, M.M.; Price, A.M.; Boom, J.A.; Sahni, L.C.; Cameron, M.H.; Pannaraj, P.S.; Bline, K.E.; 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, 18, 264–270. [Google Scholar]
  40. Badell, M.L.; Dude, C.M.; Rasmussen, S.; Jamieson, D.J. COVID-19 vaccination in pregnancy. BMJ 2022, 378, e069741. [Google Scholar] [CrossRef]
  41. Shen, C.J.; Fu, Y.C.; Lin, Y.P.; Shen, C.F.; Sun, D.J.; Chen, H.Y.; Cheng, C.M. Evaluation of Transplacental Antibody Transfer in SARS-CoV-2-Immunized Pregnant Women. Vaccines 2022, 10, 101. [Google Scholar] [CrossRef]
  42. Beharier, O.; Mayo, R.P.; Raz, T.; Sacks, K.N.; Schreiber, L.; Suisse-Cohen, Y.; Chen, R.; Gomez-Tolub, R.; Hadar, E.; Gabby-Benziv, R.; et al. Efficient maternal to neonatal transfer of antibodies against SARS-CoV-2 and BNT162b2 mRNA COVID-19 vaccine. J. Clin. Invest. 2021, 131, e154834. [Google Scholar] [CrossRef]
  43. Ben-Mayor Bashi, T.; Amikam, U.; Ashwal, E.; Hershkovitz, G.; Attali, E.; Berkovitz-Shperling, R.; Domibsky, O.; Halperin, T.; Goldshmidt, H.; Gamzu, R.; et al. The association of maternal SARS-CoV-2 vaccination-to-delivery interval and the levels of maternal and cord blood antibodies. Int. J. Gynaecol. Obstet. 2022, 156, 436–443. [Google Scholar] [CrossRef]
  44. Bookstein Peretz, S.; Regev, N.; Novick, L.; Nachshol, M.; Goffer, E.; Ben-David, a.; Asraf, K.; Doolman, R.; Levin, E.G.; Yochay, G.R.; et al. Short-term outcome of pregnant women vaccinated with BNT162b2 mRNA COVID-19 vaccine. Ultrasound Obstet. Gynecol. 2021, 58, 450–456. [Google Scholar] [CrossRef]
  45. Kashani-Ligumsky, L.; Lopian, M.; Cohen, R.; Senderovich, H.; Czeiger, S.; Halpin, A.; Chaim, A.B.; Kremer, I.; Lessing, J.B.; Somekh, E.; et al. Titers of SARS-CoV-2 antibodies in cord blood of neonates whose mothers contracted SARS-CoV-2 (COVID-19) during pregnancy and in those whose mothers were vaccinated with mRNA to SARS-CoV-2 during pregnancy. J. Perinatol. 2021, 41, 2621–2624. [Google Scholar] [CrossRef]
  46. Kugelman, N.; Nahshon, C.; Shaked-Mishan, P.; Cohen, N.; Sher, M.L.; Gruber, M.; Marom, I.; Zolotarevsky, A.; Lavie, O.; Damti, A.; et al. Maternal and Neonatal SARS-CoV-2 Immunoglobulin G Antibody Levels at Delivery After Receipt of the BNT162b2 Messenger RNA COVID-19 Vaccine During the Second Trimester of Pregnancy. JAMA Pediatr. 2022, 176, 290–295. [Google Scholar] [CrossRef]
  47. Nir, O.; Schwartz, A.; Toussia-Cohen, S.; Leibovitch, L.; Strauss, T.; Asraf, K.; Doolam, R.; Sgarabi, S.; Cohen, C.; Lustig, Y.; et al. Maternal-neonatal transfer of SARS-CoV-2 immunoglobulin G antibodies among parturient women treated with BNT162b2 messenger RNA vaccine during pregnancy. Am. J. Obstet Gynecol. MFM 2022, 4, 100492. [Google Scholar] [CrossRef]
  48. Rottenstreich, A.; Zarbiv, G.; Oiknine-Djian, E.; Zigron, R.; Wolf, D.G.; Porat, S. 27 Efficient Maternofetal Transplacental Transfer of Anti- Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Antibodies After Antenatal SARS-CoV-2 BNT162b2 Messenger RNA Vaccination. Clin. Infect. Dis. 2021, 73, 1909–1912. [Google Scholar] [CrossRef]
  49. Rottenstreich, A.; Zarbiv, G.; Oiknine-Djian, E.; Vorontsom, O.; Zigron, R.; Kleinstern, G.; Wolf, D.G.; Porat, S. The effect of gestational age at BNT162b2 mRNA vaccination on maternal and neonatal SARS-CoV-2 antibody levels. Clin. Infect. Dis. 2022, 75, e603–e610. [Google Scholar] [CrossRef]
  50. Rottenstreich, M.; Sela, H.Y.; Rotem, R.; Kadish, E.; Wiener-Well, Y.; Grisaru-Granovsky, S. COVID-19 vaccination during the third trimester of pregnancy: Rate of vaccination and maternal and neonatal outcomes, a multicentre retrospective cohort study. BJOG 2022, 129, 248–255. [Google Scholar] [CrossRef]
  51. Atyeo, C.; DeRiso, E.A.; Davis, C.; Bordt, E.A.; DeGuzman, R.M.; Shook, L.L.; Yonker, L.M.; Fasaro, A.; Akinwunmi, B.; Lauffenburger, D.A.; et al. COVID-19 mRNA vaccines drive differential antibody Fc-functional profiles in pregnant, lactating, and nonpregnant women. Sci. Transl. Med. 2021, 13, eabi8631. [Google Scholar] [CrossRef]
  52. Mithal, L.B.; Otero, S.; Shanes, E.D.; Goldstein, J.A.; Miller, E.S. Cord blood antibodies following maternal coronavirus disease 2019 vaccination during pregnancy. Am. J. Obstet. Gynecol. 2021, 225, 192–194. [Google Scholar] [CrossRef]
  53. Shanes, E.D.; Otero, S.; Mithal, L.B.; Mupanomunda, C.A.; Miller, E.S.; Goldstein, J.A. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccination in Pregnancy: Measures of Immunity and Placental Histopathology. Obstet. Gynecol. 2021, 138, 281–283. [Google Scholar] [CrossRef]
  54. Gray, K.J.; Bordt, E.A.; Atyeo, C.; Deriso, E.; Akinwunmi, B.; Young, N.; Baez, A.M.; Shook, L.L.; Cvrk, D.; James., K.; et al. Coronavirus disease 2019 vaccine response in pregnant and lactating women: A cohort study. Am. J. Obstet. Gynecol. 2021, 225, 303.e1–303.e17. [Google Scholar] [CrossRef]
  55. Trostle, M.E.; Aguero-Rosenfeld, M.E.; Roman, A.S.; Lighter, J.L. High antibody levels in cord blood from pregnant women vaccinated against COVID-19. Am. J. Obstet. Gynecol. MFM 2021, 3, 100481. [Google Scholar] [CrossRef]
  56. Citu, I.M.; Citu, C.; Gorun, F.; Sas, I.; Tomescu, L.; Neamtu, R.; Motoc, A.; Gorum, A.M.; Burlea, B.; Brastosin, F.; et al. Immunogenicity Following Administration of BNT162b2 and Ad26.COV2.S COVID-19 Vaccines in the Pregnant Population during the Third Trimester. Viruses 2022, 14, 307. [Google Scholar] [CrossRef]
  57. Gloeckner, S.; Hornung, F.; Heimann, Y.; Schleussner, E.; Deinhardt-Emmer, S.; Loeffler, B.; Zoellkan, J. Newborns’ passive humoral SARS-CoV-2 immunity following heterologous vaccination of the mother during pregnancy. Am. J. Obstet. Gynecol. 2022, 226, 261–262. [Google Scholar] [CrossRef]
  58. Yang, Y.J.; Murphy, E.A.; Singh, S.; Sukhu, A.C.; Wolfe, I.; Adurty, S.; Eng, D.; Yee, J.; Mohammed, I.; Zhao, Z.; et al. Association of Gestational Age at Coronavirus Disease 2019 (COVID-19) Vaccination, History of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection, and a Vaccine Booster Dose with Maternal and Umbilical Cord Antibody Levels at Delivery. Obstet. Gynecol. 2022, 139, 373–380. [Google Scholar] [CrossRef]
  59. Maertens, K.; Orije, M.R.P.; Van Damme, P.; Leuridan, E. Vaccination during pregnancy: Current and possible future recommendations. Eur. J. Pediatr. 2020, 179, 235–242. [Google Scholar] [CrossRef]
  60. Andrews, N.; Stowe, J.; Kirsebom, F.; Toffa, S.; Rickeard, T.; Gallagher, E.; Gower, C.; Kall, M.; Groves, N.; O’Connell, A.M.; et al. COVID-19 Vaccine Effectiveness against the Omicron (B.1.1.529) Variant. N. Engl. J. Med. 2022, 21, 1532–1546. [Google Scholar] [CrossRef]
  61. The Institute for Health Metrics and Evaluation (IHME). COVID-19 Vaccine Efficacy Summary. Available online: https://www.healthdata.org/covid/covid-19-vaccine-efficacy-summary (accessed on 17 May 2022).
  62. Dagan, N.; Barda, N.; Biron-Shental, T. Effectiveness of the BNT162b2 mRNA COVID-19 vaccine in pregnancy. Nat. Med. 2021, 27, 1693–1695. [Google Scholar] [CrossRef]
  63. Abu-Raddad, L.; Chemaitelly, H.; Ayoub, H.; AlMukdad, S.; Yassine, H.; Al-Khatib, H.; Smatti, M.; Tang, P.; Hasan, M.; Coyle, P.; et al. Effect of mRNA Vaccine Boosters against SARS-CoV-2 Omicron Infection in Qatar. N. Engl. J. Med. 2022, 386, 1804–1816. [Google Scholar] [CrossRef]
  64. Noureddine, F.Y.; Chakkour, M.; El Roz, A.; Reda, J.; Al Sahily, R.; Assi, A.; Joma, M.; Salami, H.; Hashem, S.J.; Harb, B.; et al. The Emergence of SARS-CoV-2 Variant(s) and Its Impact on the Prevalence of COVID-19 Cases in the Nabatieh Region, Lebanon. Med. Sci. 2021, 9, 40. [Google Scholar] [CrossRef]
  65. Vasireddy, D.; Vanaparthy, R.; Mohan, G.; Malayala, S.V.; Atluri, P. Review of COVID-19 Variants and COVID-19 Vaccine Efficacy: What the Clinician Should Know? J. Clin. Med. Res. 2021, 13, 317–325. [Google Scholar] [CrossRef]
  66. Kharbanda, E.O.; Haapala, J.; DeSilva, M.; Vazquez-Benitez, G.; Vesco, K.K.; Naleway, A.L.; Lipkind, H.S. Spontaneous Abortion Following COVID-19 Vaccination During Pregnancy. JAMA 2021, 326, 1629–1631. [Google Scholar] [CrossRef]
  67. Lipkind, H.S.; Vazquez-Benitez, G.; DeSilva, M.; Vesco, K.K.; Ackerman-Banks, C.; Zhu, J.; Boyce, T.G.; Daley, M.F.; Fuller., C.C.; Getahun, D.; et al. Receipt of COVID-19 Vaccine During Pregnancy and Preterm or Small-for-Gestational-Age at Birth; Eight Integrated Health Care Organizations: United. R Morb Mortal Wkly Rep. 2022, 7, 26–30. [Google Scholar] [CrossRef]
  68. Morgan, J.A.; Biggio, J.R., Jr.; Mussarat, N.; Chawla, H.K.; Puri, P.; Williams, F.B. Maternal Outcomes After Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection in Vaccinated Compared with Unvaccinated Pregnant Patients. Obstet. Gynecol. 2022, 139, 107–109. [Google Scholar] [CrossRef]
  69. Ruderman, R.S.; Mormol, J.; Trawick, E.; Perry, M.F.; Allen, C.C.; Millan, D.; Miller, E.S. Association of COVID-19 Vaccination During Early Pregnancy with Risk of Congenital Fetal Anomalies. JAMA Pediatr. 2022, 176, 717–719. [Google Scholar] [CrossRef]
  70. Shimabukuro, T.T.; Kim, S.Y.; Myers, T.R.; Moro, P.L.; Oduyebo, T.; Panagiotakopoulos, L.; Marquez, P.L.; Olson, C.; Liu, R.; Chang, K.T.; et al. Preliminary Findings of mRNA COVID-19 Vaccine Safety in Pregnant Persons. N. Engl. J. Med. 2021, 384, 2273–2282. [Google Scholar] [CrossRef] [PubMed]
  71. Theiler, R.N.; Wick, M.; Mehta, R.; Weaver, A.L.; Virk, A.; Swift, M. Pregnancy and birth outcomes after SARS-CoV-2 vaccination in pregnancy. Am. J. Obstet. Gynecol. MFM 2021, 3, 100467. [Google Scholar] [CrossRef]
  72. Trostle, M.E.; Limaye, M.A.; Avtushka, V.; Lighter, J.L.; Penfield, C.A.; Roman, A.S. COVID-19 vaccination in pregnancy: Early experience from a single institution. Am. J. Obstet. Gynecol. MFM 2021, 3, 100464. [Google Scholar] [CrossRef]
  73. Zauche, L.H.; Wallace, B.; Smoots, A.N.; Olson, C.K.; Oduyebo, T.; Kim, S.Y.; Petersen, E.E.; Ju, J.; Beauregard, J.; Wilcox, A.J.; et al. Receipt of mRNA COVID-19 Vaccines and Risk of Spontaneous Abortion. N. Engl. J. Med. 2021, 385, 1533–1535. [Google Scholar] [CrossRef]
  74. Bleicher, I.; Kadour-Peero, E.; Sagi-Dain, L.; Sagi, S. Early exploration of COVID-19 vaccination safety and effectiveness during pregnancy: Interim descriptive data from a prospective observational study. Vaccine 2021, 39, 6535–6538. [Google Scholar] [CrossRef]
  75. Dick, A.; Rosenbloom, J.I.; Gutman-Ido, E.; Lessans, N.; Cahen-Peretz, A.; Chill, H.H. Safety of SARS-CoV-2 vaccination during pregnancy-obstetric outcomes from a large cohort study. BMC Pregnancy Childbirth 2022, 22, 166. [Google Scholar] [CrossRef]
  76. Goldshtein, I.; Steinberg, D.M.; Kuint, J.; Chopik, G.; Segal, Y.; David, S.S.B.; Ben-Tov, A. Association of BNT162b2 COVID-19 Vaccination During Pregnancy with Neonatal and Early Infant Outcomes. JAMA Pediatr. 2022, 176, 470–477. [Google Scholar] [CrossRef] [PubMed]
  77. Wainstock, T.; Yoles, I.; Sergienko, R.; Sheiner, E. Prenatal maternal COVID-19 vaccination and pregnancy outcomes. Vaccine 2021, 39, 6037–6040. [Google Scholar] [CrossRef]
  78. Magnus, M.C.; Örtqvist, A.K.; Dahlqwist, E.; Ljung, R.; Skar, F.; Oakley, L.; Macsali, F.; Pasternak, B.; Gjessing, H.K.; Haberg, S.E.; et al. Association of SARS-CoV-2 Vaccination During Pregnancy with Pregnancy Outcomes. JAMA 2022, 327, 1469–1477. [Google Scholar] [CrossRef]
  79. Magnus, M.C.; Gjessing, H.K.; Eide, H.N.; Wilcox, A.J.; Fell, D.B.; Håberg, S.E. COVID-19 Vaccination during Pregnancy and First-Trimester Miscarriage. N. Engl. J. Med. 2021, 385, 2008–2010. [Google Scholar] [CrossRef]
  80. Fell, D.B.; Dhinsa, T.; Alton, G.D.; Torok, E.; Dimanlig-Cruz, S.; Regan, A.K.; Sprague, A.e.; Buchan, S.A.; Kwong, J.C.; Wilson, S.E.; et al. Association of COVID-19 Vaccination in Pregnancy with Adverse Peripartum Outcomes. JAMA 2022, 327, 1478–1487. [Google Scholar] [CrossRef]
  81. Sadarangani, M.; Soe, P.; Shulha, H.P.; Valiquette, L.; Vanderkooi, O.G.; Kellner, J.D.; Muller, M.P.; Top, K.A.; Isenor, J.E.; McGeer, A.; et al. For the Canadian Immunization Research Network. Safety of COVID-19 vaccines in pregnancy: A Canadian National Vaccine Safety (CANVAS) network cohort study. Lancet Infec. Dis. 2022, 22, 1553–15564. [Google Scholar] [CrossRef]
  82. Blakeway, H.; Prasad, S.; Kalafat, E.; Heath, P.; Ladhani, S.N.; Le Doare, K.; Magee, L.A.; O’Brien, P.; Rezvani, A.; von Dadelszen, P.; et al. COVID-19 vaccination during pregnancy: Coverage and safety. Am. J. Obstet. Gynecol. 2022, 226, 236.e1–236.e14. [Google Scholar] [CrossRef]
  83. Vojtek, I.; Dieussaert, I.; Doherty, T.M.; Franck, V.; Hanssens, L.; Miller, J.; Bekkat-Berkani, R.; Kandell, W.; Prado-Cohrs, D.; Vyse, A. Maternal immunization: Where are we now and how to move forward? Ann. Med. 2018, 50, 193–208. [Google Scholar] [CrossRef] [Green Version]
  84. Adhikari, E.H.; Spong, C.Y. COVID-19 Vaccination in Pregnant and Lactating Women. JAMA 2021, 325, 1039–1040. [Google Scholar] [CrossRef]
  85. Zdanowski, W.; Wasniewski, T. Evaluation of SARS-CoV-2 Spike Protein Antibody Titers in Cord Blood after COVID-19 Vaccination during Pregnancy in Polish Healthcare Workers: Preliminary Results. Vaccines 2021, 9, 675. [Google Scholar] [CrossRef]
  86. Sourouni, M.; Braun, J.; Oelmeier, K.; Möllers, M.; Willy, D.; Hennies, M.T.; Köster, H.A.; Pecks, U.; Klockenbusch, W.; Schmitz, R. Assessment of Neonatal Cord Blood SARS-CoV-2 Antibodies after COVID-19 Vaccination in Pregnancy: A Prospective Cohort Study. Geburtshilfe Frauenheilkd. 2022, 82, 510–516. [Google Scholar] [CrossRef]
  87. Popescu, D.-E.; Cîtu, C.; Jura, A.M.C.; Lungu, N.; Navolan, D.; Craina, M.; Semenescu, A.M.; Gorum, F.; Jura, M.-A.; Belengeanu, V.; et al. The Benefits of Vaccination against SARS-CoV-2 during Pregnancy in Favor of the Mother/Newborn Dyad. Vaccines 2022, 10, 848. [Google Scholar] [CrossRef]
  88. Munoz, F.M. Can we protect pregnant women and young infants from COVID-19 through maternal immunization. JAMA Pediatr. 2021, 175, 561–562. [Google Scholar] [CrossRef]
  89. Rottenstreich, A.; Zarbiv, G.; Oiknine-Djian, E.; Vorontsov, O.; Zigron, R.; Kleinstern, G.; Wolf, D.G.; Porat, S. Timing of SARS-CoV-2 vaccination during the third trimester of pregnancy and transplacental antibody transfer: A prospective cohort study. Clinic. Microbiol. Infect. 2022, 28, 419–425. [Google Scholar] [CrossRef]
  90. Atyeo, C.G.; Shook, L.L.; Brigida, S.; De Guzman, R.M.; Demidkin, S.; Muir, C.; Akinwunmi, B.; Baez, A.M.; Sheehan, M.L.; McSweeney, E.; et al. Maternal immune response and placental antibody transfer after COVID-19 vaccination across trimester and platforms. Nat. Comm. 2022, 13, 3571. [Google Scholar] [CrossRef]
  91. Low, J.M.; Gu, Y.; Ng, M.S.F.; Amin, Z.; Lee, L.Y.; Ng, Y.P.M.; Shunmuganathan, B.D.O.; Niu, Y.; Gupta, R.; Tambyah, P.A.; et al. Codominant IgG and IgA expression with minimal vaccine mRNA in milk of BNT162b2 vaccinees. NPJ Vaccines. 2021, 6, 105. [Google Scholar] [CrossRef]
  92. Perl, S.H.; Uzan-Yulzari, A.; Klainer, H.; Asiskovich, L.; Youngster, M.; Rinott, E.; Youngster, I. SARS-CoV-2–Specific Antibodies in Breast Milk After COVID-19 Vaccination of Breastfeeding Women. JAMA 2021, 325, 2013–2014. [Google Scholar] [CrossRef]
  93. Baird, J.K.; Jensen, S.M.; Urba, W.J.; Fox, B.A.; Baird, J.R. SARS-CoV-2 antibodies detected in mother’s milk post-vaccination. J. Hum. Lact. 2021, 37, 492–498. [Google Scholar] [CrossRef]
  94. Whited, N.; Cervantes, J. Antibodies Against SARS-CoV-2 in Human Breast Milk After Vaccination: A Systematic Review and Meta-Analysis. Breastfeed. Med. 2022, 17, 475–483. [Google Scholar] [CrossRef]
  95. Pace, R.M.; Williams, J.E.; Jarvinen, K.M.; Belfort, M.B.; Pace, C.D.W.; Lackey, K.A.; Gogel, A.C.; Nguyen-Contant, P.; Karagaiah, P.; Fitzgerald, T.; et al. Characterization of SARS-CoV-2 RNA, Antibodies, and Neutralizing Capacity in Milk Produced by Women with COVID-19. Epidemiology 2021, 12, e03192-20. [Google Scholar] [CrossRef]
  96. Young, B.E.; Seppo, A.E.; Diaz, N.; Rosen-Carole, C.; Nowak-Wegrzyn, A.; Cruz Vasquez, J.M.; Ferri-Huerta, R.; Nguyen-Constant, P.; Fitzgerald, t.; Sangster, M.Y.; et al. Association of Human Milk Antibody Induction, Persistence, and Neutralizing Capacity with SARS-CoV-2 Infection vs mRNA Vaccination. JAMA Pediatrics. 2022, 176, 159–168. [Google Scholar] [CrossRef]
  97. Perez, S.E.; Luna Centeno, L.D.; Cheng, W.A.; Marentes Ruiz, C.J.; Lee, Y.; Congrave-Wilson, Z.; Powell, R.L.; Stellwagen, L.; Pannaraj, P.S. Human milk SARS-CoV-2 antibodies up to 6 months after vaccination. Pediatrics 2022, 149, e2021054260. [Google Scholar] [CrossRef]
  98. Rodriguez-Garcia, M.; Biswas, N.; Patel, M.V.; Barr, F.D.; Crist, S.G.; Ochsenbauer, C.; Fahey, J.V.; Wira, C.R. Estradiol reduces susceptibility of CD4+ T cells and macrophages to HIV-infection. PLoS ONE 2013, 8, e62069. [Google Scholar] [CrossRef] [Green Version]
  99. Goad, J.; Rudolph, J.; Rajkovic, A. Female reproductive tract has low concentration of SARS-CoV2 receptors. PLoS ONE 2020, 15, e0243959. [Google Scholar] [CrossRef]
  100. Singh, B.; Gornet, M.; Sims, H.; Kisanga, E.; Knight, Z.; Segars, J. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and its effect on gametogenesis and early pregnancy. Am. J. Reprod. Immunol. 2020, 84, 1–9. [Google Scholar] [CrossRef] [PubMed]
  101. Dashraath, P.; Wong, J.L.J.; Lim, M.X.K.; Kim, L.M.; Li, S.; Biswas, A.; CHoolani, M.; Matter, C.; Su, L.L. Coronavirus disease 2019 (COVID-19) pandemic and pregnancy. Am. J. Obstet. Gynecol. 2020, 222, 521–531. [Google Scholar] [CrossRef]
  102. Wang, Z.; Xu, X. scRNA-seq profiling of human testes reveals the presence of the ACE2 receptor, a target for SARS-CoV-2 infection in spermatogonia, Leydig and Sertoli cells. Cells 2020, 9, 920. [Google Scholar] [CrossRef] [Green Version]
  103. Paoli, D.; Pallotti, F.; Turriziani, O.; Mazzuti, L.; Antonelli, G.; Lenzi, A.; Lombardo, F. SARS-CoV-2 presence in seminal fluid: Myth or reality. Andrology 2021, 9, 23–26. [Google Scholar] [CrossRef]
  104. Pozzilli, P.; Lenzi, A. Testosterone, a key hormone in the context of COVID-19 pandemic [Commentary]. Metabolism 2020, 108, 154252. [Google Scholar] [CrossRef]
  105. Xu, J.; Qi, L.; Chi, X.; Yang, J.; Wei, X.; Gong, E.; Peh, S.; Gu, J. Orchitis: A complication of severe acute respiratory syndrome (SARS). Biol. Reprod. 2006, 74, 410–416. [Google Scholar] [CrossRef] [Green Version]
  106. Batiha, O.; Al-Deeb, T.; Al-zoubi, E.A.; Alsharu, E. Impact of COVID-19 and other viruses on reproductive health. Andrologia 2020, 52, e13791. [Google Scholar] [CrossRef]
  107. Li, D.; Jin, M.; Bao, P.; Zhao, W.; Zhang, S. Clinical characteristics and results of semen tests among men with coronavirus disease 2019. JAMA Network Open 2020, 3, e208292. [Google Scholar] [CrossRef] [PubMed]
  108. A Guide to WHO’s Guidance on COVID-19 [Internet]. Available online: https://www.who.int/news-room/feature-stories/detail/a-guide-to-who-s-guidance (accessed on 27 April 2021).
  109. Aksak, T.; Satar, D.A.; Bagci, R.; Gultekin, E.O.; Coskun, A.; Demirdelen, U. Investigation of the effect of COVID-19 on sperm count, motility and morphology. J. Med. Virol. 2022, 94, 5201–5205. [Google Scholar] [CrossRef]
  110. Gharagozloo, P.; Cartagena, S.; Moazamian, A.; Drevet, J.R.; Somkuti, S.; Aitken, R.J. Rapid impact of COVID-19 infection on semen quality: A case report. Transl. Androl. Urol. 2022, 11, 110–115, PMCID:8824823. [Google Scholar] [CrossRef] [PubMed]
  111. American College of Obstetricians and Gynecologists. COVID-19 FAQs for Obstetrician–Gynecologists, Gynecology. COVID-19 FAQs. Washington, DC: ACOG. 30 May 2020. Available online: https://www.acog.org/clinical-information/physician-faqs/covid19-faqs-for-ob-gyns-gynecology (accessed on 27 April 2021).
  112. Royal College of Obstetricians and Gynecologists. Corona Virus (COVID-19) and gynaecological services. RCOG. 2020. Available online: https://www.rcog.org.uk/en/guidelines-research-services/coronavirus-covid-19-pregnancy-and-womens-health/coronavirus-covid-19-and-gynaecological-services/ (accessed on 27 April 2021).
  113. International Federation of Gynecology and Obstetrics. COVID-19 (Corona Virus) Statement. FIGO.2020. Available online: https://www.figo.org/covid-19-coronavirus-statement (accessed on 27 April 2021).
  114. Veiga, A.; Gianaroli, L.; Ory, S.; Horton, M.; Feinberg, E.; Penzias, A. Assisted reproduction and COVID-19, a joint statement of ASRM, ESHRE and IFFS. Hum. Reprod. Open 2020, 1, 3. [Google Scholar]
  115. Vaiarelli, A.; Bulletti, C.; Cimadomo, D.; Borini, A.; Alviggi, C.; Ajossa, S.; Anserini, P.; Gennarelli, G.; Guido, M.; Levi-Setti, P.; et al. COVID-19 and ART: The view of the Italian Society of Fertility and Sterility and Reproductive Medicine. Reprod. Biomed. Online. 2020, 40, 755–759. [Google Scholar] [CrossRef]
  116. Gonzalez, D.C.; Nassau, D.E.; Khodamoradi, K.; Ibrahim, E.; Blachman-Braun, R.; Ory, J.; Ramasamy, R. Sperm parameters before and after COVID-19 mRNA vaccination. JAMA 2021, 326, 273–274. [Google Scholar] [CrossRef] [PubMed]
  117. Ory, S.; Veiga, A.; Horton, M.; Gianaroli, L. Joint IFFS/ESHRE statement on COVID-19 vaccination for pregnant women and those considering pregnancy. Hum. Reprod. Open. 2021, 2, hoab016, PMCID:8050407. [Google Scholar] [CrossRef] [PubMed]
  118. Rao Orvieto, R.; Noach-Hirsh, M.; Segev-Zahav, A.; Haas, J.; Nahum, R.; Aizer, A. Does mRNA SARS-CoV-2 vaccine influence patients’ performance during IVF-ET cycle? Reprod. Biol. Endocrinol. 2021, 19, 69. [Google Scholar] [CrossRef]
  119. Bentov, Y.; Beharier, O.; Moav-Zafrir, A.; Kabessa, M.; Godin, M.; Greenfield, C.; Broder, E.A.; Holzer, H.; Wolf, D.; Oiknine-Djian, E.; et al. Klement Ovarian follicular function is not altered by SARS–CoV-2 infection or BNT162b2 mRNA COVID-19 vaccination. Hum. Reprod. 2021, 36, 2506–2513. [Google Scholar] [CrossRef]
  120. Zaçe, D.; La Gatta, E.; Petrella, L.; Di Pietro, M.L. The impact of COVID-19 vaccines on fertility—A systematic review and meta-analysis . Vaccine 2022, 40, 6023–6034, Epub 2022 Sep 12. [Google Scholar] [CrossRef]
  121. Kachikis, A.; Englund, J.A.; Singleton, M.; Covelli, I.; Drake, A.L.; Eckert, L.O. Short-term Reactions among Pregnant and Lactating Individuals in the First Wave of the COVID-19 Vaccine Rollout. JAMA Netw. Open. 2021, 4, e2121310. [Google Scholar] [CrossRef]
  122. Available online: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/expect/after.html (accessed on 17 October 2022).
  123. Logunov, D.Y.; Dolzhikova, I.V.; Tukhvatullin, A.I.; Shcheblyakov, D.V. Safety and efficacy of the Russian COVID-19 vaccine: More information needed—Authors’ reply. Lancet 2020, 396, 54–55. [Google Scholar] [CrossRef]
  124. Kloff, F.A.; Pai, M.; Huisman, M.V.; Makris, M. Vaccine-induced immune thrombotic thrombocytopenia. Lancet Haematol. 2021, 9, 73–80. [Google Scholar] [CrossRef]
  125. Briller, J.; Aggarwal, N.; Davis, M.; Hamed, A.B.; Malhame, I.; Mahmoud, Z.; McDonald, E.G.; de Oliveira, G.; Quesada, O.; Scott, N.S.; et al. Cardiovascular Disease in Women Committee Cardiovascular Complications of Pregnancy—Associated COVID-19 Infections. JACC Adv. 2022, 1, 100057. [Google Scholar] [CrossRef]
  126. Centers for Disease Control and Prevention. COVID-19 Vaccines While Pregnant or Breastfeeding. Available online: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/recommendations/pregnancy.html (accessed on 18 May 2022).
  127. Society for Maternal-Fetal Medicine SMFM: Provider Considerations for Engaging in COVID-19 Vaccine Counseling with Pregnant and Lactating Patients. 1.11.2022 (Last Published 12.20.21). Available online: https://s3.amazonaws.com/cdn.smfm.org/media/3290/Provider_Considerations_for_Engaging_in_COVID_Vaccination_Considerations_1-11-22_%28final%29_KS.pdf (accessed on 17 October 2022).
  128. Society for Maternal-Fetal Medicine SMFM: COVID-19 and Pregnancy: What Maternal-Fetal Medicine Subspecialists Need to Know. 3.2.22 (update of draft originally posted on 1.3.2022). Available online: https://s3.amazonaws.com/cdn.smfm.org/media/3402/COVID19-What_MFMs_need_to_know_revision_3-1-22_%28final%29.pdf (accessed on 17 October 2022).
  129. Available online: https://www.acog.org/clinical/clinical-guidance/practice-advisory/articles/2020/12/vaccinating-pregnant-and-lactating-patients-against-covid-19 (accessed on 22 March 2021).
  130. Royal College of Obstetricians and Gynaecologists. Coronavirus (COVID-19) Infection in Pregnancy. Information for Healthcare Professionals. Version 15, Published Monday 7 March 2022. Available online: https://www.rcog.org.uk/media/xsubnsma/2022-03-07-coronavirus (accessed on 17 October 2022).
  131. Chakkour, M.; Salami, A.; Olleik, D.; Kamal, I.; Noureddine, F.Y.; Roz, A.E.; Ghssein, G. Risk Markers of COVID-19, a Study from South-Lebanon. COVID 2022, 2, 867–876. [Google Scholar] [CrossRef]
Table 1. Authorised vaccines for use in pregnancy, including their efficacy and safety.
Table 1. Authorised vaccines for use in pregnancy, including their efficacy and safety.
VaccineManufacturer/CountryPlatform/
Technology
No of DosesEfficacy from Randomised TrialsReported Adverse EffectsReproductive Toxicology Studies
Pfizer-BioNTech mRNA (BNT16b2) [25]USA—PfizermRNA—encodes stabilised spike, lipid nanoparticle2 doses at least 3 weeks apart95% against symptomatic COVID-19Myocarditis—more after 2nd dose. Reported rate is 3.5/1,000,000 after 2nd dose. Highest in age 18–29 years [26]No reported safety concerns in rats given vaccine before and during pregnancy—investigated—effect on gonads/gametes fertility, embryo-fetal development, growth and neurofunction [27,28,29]
Moderna mRNA 1273 [30]Moderna USAmRNA—encodes stabilised spike, lipid nanoparticle2 doses 4 weeks apart94.1% against symptomatic COVID-19Myocarditis– especially after 2nd dose—rate 3.5/1,000,000; highest in 18–29 age group [26]No reported safety concerns in rats given vaccine before and during pregnancy—investigated—effect on gonads/gametes fertility, embryo-fetal development, growth and neurofunction [28,29,31]
Johnson and Johnson-Janssen Ad26 COV2.S [32] Netherlands, Belgium and USAReplication incompetent human adenovirus type 26 vector-stabilised spikeOne dose66.1% against moderate to severe-critical COVID-19; 85.4% against severe-critical COVID-9Thrombosis with thrombocytopenia syndrome—rate 3/1,000,000 cases. Highest in females aged 30–49 years. Guillain-Barre syndrome—7.8/1,000,000 cases; highest in 50–64 age group [26]No reported safety concerns in rats given vaccine before and during pregnancy—investigated—effect on gonads/gametes fertility, embryo-fetal development, growth and neurofunction [28,29,33]
Oxford-AstraZenecaChAdOx1 (AZS1222) [34]AstraZeneca
UK
Recombinant replication of deficient chimpanzee adenoviral vector encoding SARS-CoV-2 spike proteinTwo doses 4–12 weeks apart70.4% against asymptomatic COVID-19; 100% against severe-critical COVID-19 Development and reproductive toxicology studies have not shown harmful effects in pregnant animals and their offspring [35]
Table 2. Evidence of immunoreactivity from published studies in pregnancy (adapted from Badell et al. [40]).
Table 2. Evidence of immunoreactivity from published studies in pregnancy (adapted from Badell et al. [40]).
Author, Year and Study TypeVaccine TypeGestational Age (Weeks) at VaccinationSample SizeSeropositive Maternal BloodSeropositive Cord Blood
Shen et al., 2022
Prospective cohort [41]
Moderna28–33 Total—29 (1st dose only 4, 1st & 2nd dose 25)Antibody against
  • wild type SARS-CoV-2—neutralizing IgG 40% > one dose and 97.5% > 2 doses
  • Delta variant—neutralizing IgG 4% > 1 dose and 80.5% > 2 doses
Antibody against
  • Wild type—Neutralizing IgG 43.3% > 1 dose and 97.4% > 2 doses
  • Delta variant—Neutralizing IgG 1.4% >1 dose and 66.3% > 2 doses
Beharier et al., 2021 Multicentre cohort [42]Pfizer34 Vaccinated—86; infected—65 and control—62Antibody against SARS-CoV-2
  • Rise in anti-S and anti-RBD titres > 15 days of 1st dose
  • Additional rise after 2nd dose
  • Vaccinated higher anti-S1 and anti-RBD but natural infection—higher anti-S2 and N antibodies
Antibody against SARS-CoV-2
  • Levels of anti-IgG and anti-RBD were the same after vaccination and natural infection, but transfer ratios for anti-S1, anti-S2 and anti-RBD than with natural infection
Ben-Mayor et al., 2021 Prospective multicentre [43]Pfizer34–37 weeks58 (1st does 19, 2nd dose 29)Antibody against SARS-CoV-2
  • Anti-IgG anti-S > 50 AU/mL detected in 53 samples
Antibody against SARS-CoV-2
  • Detected in 51 cord samples and negative in 7, all with a 1st dose to delivery interval of <27 days
  • Maternal antibodies tires positively correlated with cord titres
Bookstein et al., 2021 [44]PfizerUnavailable—not provided650 (390—pregnant and 260—non-pregnant controls)Antibody against SARS-CoV-2
Pregnant women had significantly lower serum IgG titres than non-pregnant women
No measured
Kashani-Ligumsky et al., 2021, Cohort [45]PfizerNo reported29—vaccinated, 29—infected, 21—un-vaccinated controlsAntibody against SARS-CoV-2
Mean titre of 224.7 U/mL in vaccinated v 83.U/mL in infected
Antibody against SARS-CoV-2
  • Anti-S IgG positive in 100% of cord samples in vaccinated
  • Mean neonatal titres significantly higher in vaccinated vs. unvaccinated (225 U/mL vs. 83.7 U/M, p ≤ 0.05)
Kulgelman et al., 2021, Prospective cohort [46]Pfizer25130Antibody against SARS-CoV-2
  • IgG antibody positive in 100% of cases
  • Change in maternal antibody level per week increase from 2nd vaccine dose to birth—10.9%
Antibody against SARS-CoV-2
  • 100% for IgG titre 2.6 times higher than maternal titres
  • Change in antibody titre per week increase from 2nd vaccine doe to birth (−11.7%)
Nir et al., 2021; Prospective cohort [47]Pfizer33 Total—64 fully vaccinated (1st & 2nd dose); unvaccinated and recovered from COVID-19 = 11Antibody against
  • 100% positive for SARS-CoV-2 IgG
  • Recovered from COVID-19
  • IgG-positive 26.1% in vaccinated
  • IgG positive in 2.6% in unvaccinated
Antibody against SARS-CoV-2
  • 98.3% in fully vaccinated
  • 20.2% in recovered vaccinated vs. 3.3% in recovered unvaccinated
Rottenstreich A et al., 2021; Prospective cohort [48]Pfizer3rd trimester20 fully vaccinated (1st & 2nd dose)Antibody against
SARS-CoV-2 anti-S and anti-RBD in 100% of cases
Antibody against SARS-CoV-2 anti-S and anti-RBD
100%
  • Cord levels positively correlated with maternal levels
  • Cord levels increased with increasing time from 1st vaccine to delivery
Rottenstreich A et al., 2021; Cohort [49]Pfizer3rd trimester (27–36)171 Antibody against
SARS-CoV-2 anti-S and anti-RBD—100% in all cases at time of delivery
Titres are lower in those vaccinated in early vs. late third trimester
Antibody against SARS-CoV-2 anti-S and anti-RBD—100% in neonatal serum; higher in those whose mothers were vaccinated earlier
Transfer higher after early vaccination
Positive correlation of antibody titre with duration from vaccination
Rottenstreich M et al., 2022; Prospective Cohort [50]Pfizer1st, 2nd and 3rd trimesters1st trimester = 90
2nd trimester = 124
3rd trimester = 188
Quantified IgG antibodies in maternal serum
  • 1st trimester—anti-S 76 AU/mL; anti-RBD 478 AU/mL
  • 2nd trimester—anti-S 126 AU/mL, anti-RBD-1263 AU/mL
  • 3rd trimester—anti-S-240 AU/mL, anti-RBD-5855 Au/mL
  • Booster in 3rd trimester of vaccine in 1st trimester—anti-S—1665 AU/mL, ani-RBD-20,956 AU/mL
Antibodies against SARS-CoV-2
  • positive for anti-S and anti-RBD in all neonates
  • 1st trimester—anti-S 126 AU/mL, anti-RBD-1140 AU/mL
  • 2nd trimester—anti-S-204 AU/mL; anti-RBD-8038 AU/mL
  • 3rd trimester—anti-S255 AU/mL, anti-RBD-8038 AU/mL
  • Booster in 3rd trimester after 1st trimester vaccine—anti-S-528 AU/mL, anti-RBD-4225 AU/mL
Atyeo et al., 2021 [51]Pfizer and Moderna16–3264
Moderna—1st dose—32, 2nd dose—19
Pfizer—1st dose—32, 2nd dose—17
Non-pregnant controls—14
Antibody against SARS-CoV-2
  • Fc receptor antibody titres are significantly lower in pregnancy compared to non-pregnant > 1st dose
  • Fc receptor antibody increase significantly > 2nd dose but is still lower than in non-pregnant women
Antibody against SARS-CoV-2
  • Vaccinated maternal/cord blood pairs—8, maternal titres of all antibodies higher than cord titres
  • Enriched RBD-specific FcyR3a binding in cord
Collier et al., 2021 [36]Pfizer and ModernaAll three trimestersVaccinated—103 (pregnant—30; non-pregnant—57; infected—28 of which 22—pregnant and 6 nn-pregnant0
Pfizer—11
Antibody against SARS-CoV-2
  • Pregnant vaccinated mean RDB IgG—27,601 vs. 37,839 for non-pregnant
  • Mean RBD IgG of 1321in pregnant infected vs. 910 in non-pregnant infected
  • Mean neutralizing antibody titre of 148 in pregnant vs. 901 in non-pregnant
Antibody against SARS-CoV-2
  • Vaccinated maternal and cord-paired samples—9;
Median cord RDB IgG tires higher than maternal blood titres 919,873 vs. 14,953)
Median cord neutralizing IgG lower h = than maternal blood (324 vs. 1016)
Mithal et al., 2021, Prospective cohort [52]Pfizer, Moderna and unknown3328 total (Pfizer—18, Moderna—6, unknown—4)Antibody against SARS-CoV-2
97% IgG positive
Antibody against SARS-CoV-2
  • IgG antibodies were positive in 25/28 cases
  • 3 negative—had 1st dose < 3 weeks before delivery
  • Increased latency from vaccination to delivery is associated with an increased transfer ratio
  • 2nd dose before delivery is associated with increased infant IgG levels
  • Latency from vaccination to delivery assocaity4ed with increased infant IgG levels
Shanes et al., 2021 [53]Pfizer, Moderna and unknownAll 3rd trimester84 vaccinated (Pfizer—49; Moderna—25, mRNA unknown—9) and 116 unvaccinated Antibody against SARS-CoV-2
  • Anti-IgG level 22.8 in vaccinated and 0.04 in unvaccinated
Antibody against SARS-CoV-2
Not measured
Gray et al., Prospective cohort [54]Pfizer and ModernaAll trimesters (1st—11, 2nd—39 and 3rd—34)Total—100
Pregnant (Pfizer—41, Moderna—43), non-pregnant (Pfizer—8, Moderna—8)
Antibody against SARS-CoV-2
  • Comparable IgG levels between pregnant and non-pregnant-(i.e., no difference)
  • All IgG titres higher in non-pregnant than pregnant with previous SARS-CoV-2 infection
Antibody against SARS-CoV-2
  • 10 cord samples studies
All positive for anti-S and anti-RBD IgG
Neutralizing antibodies lower in cord than maternal serum—but not statistically significant
Trostle et al., 2021 [55]Pfizer and ModernaAll trimesters (1st—2, 2nd—30, 3rd—4)Total—36 (Pfizer—26, Moderna—10)No measuredAntibody against SARS-CoV-2
  • IgG anti-S in 100% of cases
  • in 34 cases, titre > 250 IU/mL, 2 < 250 U/mL (both vaccinated > 20 weeks before delivery)
Citu et al., 2022 [56]Pfizer and Janssen Third trimester227 vaccinated
Unvaccinated 608
Vaccinated without COVID-19 history—173
Unvaccinated without COVID-19 history—529
Non-pregnant—227
Antibody against SARS-CoV-2
Prior to vaccination—N = 173 seronegative (A) vs. 54 seropositive (B) (in the pregnant cohort)
  • Before vaccination—0.41 vs. 145 U/mL
  • 2 months > vaccination IgG increases to 1697 U/mL in A vs. 14,571 in B
  • 4 months > vaccination 1083 U/mL vs. 12,571 U/mL (A vs. B), respectively)
Seronegative pregnant (N = 173) vs. non-pregnant without a history of COVID-19 (N = 173)—IgG levels
  • Before vaccination—0.41 vs. 0.40 U/mL
  • 2 months > vaccination 1697 vs. 1705 U/mL
  • 4 months > vaccination 1083 vs. 1114 U/mL
Not studied
Gloeckner et al., 2021 [57] Observational cohortPfizer, Moderna or AstraZeneca21–28Primary vaccine AstraZeneca, booster 12 weeks later with Pfizer or ModernaAntibody against SARS-CoV-2
IgG-S antibodies detected in all samples
Antibody against SARS-CoV-2
IgG-S antibodies detected in all samples
Yang et al., 2021, Retrospective cohort [58]Pfizer, Moderna and JanssenAll trimesters (pre-pregnancy—38, 1st trimester—193, 2nd trimester—699, 3rd trimester—429)1359 (Pfizer—1025, Moderna—301, Janssen—33)Antibody against SARS-CoV-2
Anti-S IgG measured in all women
  • Pre-pregnancy—(Pfizer group—3.7; Moderna group—4.8 & Janssen group—NA)
  • 1st trimester (Pfizer—3.9; Modernal—4.8; Janssen—3.6)
  • 2nd trimester (Pfizer—4.8; Moderna—5.7; Janssen—3.0)
  • 3rd trimester (Pfizer—6.2; Moderna—6.6; Janssen—2.5)
Anti-IgG in fully vaccinated women ≥ 14 days after 2nd dose
  • Pre-pregnancy (Pfizer—3.7; Moderna—4.8; Janssen—NA)
  • 1st trimester (Pfizer—3.9; Moderna—4.8; Janssen—3.6)
  • 2nd trimester (Pfizer—4.8; Moderna—5.7; Janssen—3.0)
  • 3rd trimester (Pfizer—6.4; Moderna 7.1; Janssen—2.5)
Antibody against SARS-CoV-2
Table 3. Summary from studies that report on pregnancy outcomes after COVID-19 vaccination.
Table 3. Summary from studies that report on pregnancy outcomes after COVID-19 vaccination.
Author and YearType of Study and CountryVaccine Type and Number VaccinatedPregnancy Outcome
Kharbanda et al., 2021 [66]Case-controlled—USAVaccinated = 23,375 [Pfizer (n = 8267) Moderna (n = 15,108), Janssen (n = 528)] versus unvaccinated—90,338No increase in odds of spontaneous miscarriage
Spontaneous miscarriage within 28 days of vaccination—1128/13,160) 8%)
Lipkind et al., 2022 [67]Observational retrospective—USAVaccinated 10,064 (Pfizer—5478; Moderna—4162; Janssen—424)
Unvaccinated—36,015
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • FGR/SGA—8.2% vs. 8.2% (NS)
  • Preterm birth—4.9% vs. 7.0% (NS; p = 0.06)
Morgan et al., 2022 [68]Retrospective cohort
USA
Vaccinated 1332
incompletely vaccinated or unvaccinated—8760
Pfizer (883), Moderna (382), Janssen (67)
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • Stillbirth—0% vs. 0.07% (NS)
  • Maternal death—0% vs. 0.01% (NS)
Ruderman et al., 2022 [69]Cohort USAVaccine type not defined
Vaccinated in the organogenesis window—1149
Vaccinated outside the window of organogenesis—1473
Unvaccinated—534
Congenital malformations—main outcome—no difference
  • Vaccinated 30 days before conception and up to 14 weeks—4.2%
  • Vaccinated from 10 weeks—4.0%
  • vaccinated outside organogenesis period—4.2%
  • Unvaccinated—4.4%
Shanes et al., 2021 [53]Cohort USAPfizer
Vaccinated—84
Unvaccinated—116
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • Vaginal delivery—79% vs. 65% (p = 0.04)
  • Decidual arteriopathy (NS)
  • Fetal vascular malperfusion (NS)
  • Low or high-grade villitis (NS)
Shimabukuro et al., 2021 [70]V-safe Surveillance System and VAERS Registry—USAVaccinated (Pfizer 1st dose—9052; 2nd dose—6638; Moderna—1st dose—7930, 2nd dose—5635)Outcomes not different from unvaccinated uninfected population
  • Spontaneous miscarriage—12.6%
  • Major congenital malformations—2.2%
  • Preterm birth—9.6%
  • SGA/FGR—3.2%
Theiler et al., 2021 [71]Cohort—USAPfizer—127; Moderna—12, Janssen—1Outcomes presented in vaccinated vs. unvaccinated, respectively
  • FGR/SGA—7.9% vs. 6.5% (NS)
  • Any antenatal complication—5% vs. 4.9% (NS)
  • Stillbirth—0.0% vs. 0.3% (NS)
  • Cesarean section—31.4% vs. 29.8% (NS)
  • NICU admission—0.07% vs. 0.6% (NS)
Trostle et al., 2021 [72]Case series—USAVaccinated (n = 424) [Pfizer = 332; Moderna = 92) delivered liveborn n = 85)SM rate of 6.5%, preterm birth rate of 5.9%, FGR/SGA rate of 12.2%, cesarean delivery rate of 35.5%, congenital abnormality rate of 1.2%, stillbirth rate of 0%; NICU admission rate of 15.3%, any antenatal complication risk—23.5%
All risks comparable to those of unvaccinated pregnancies
Zauche et al., 2021 [73]Cohort study—USAVaccinated (n = 2456) [Pfizer =1294; Moderna 1162)]Cumulative spontaneous miscarriage (SM) rate < 20 weeks (14.1 95 CI 12.1–16.1%)
Risk of SM in standardised against maternal age for this population was 12.8%; 95 CI 10.8–14.8%), hence no difference in miscarriage rate/risk
Beharier et al., 2021 [42]Multicentre cohort
Israel
Pfizer
Vaccinated—92; infected—74; control—66; reported
delivery outcomes—92
Vaccinated versus infected versus controls, respectively
  • Preterm birth—7.6% vs. 10.5% vs. 4.3%
  • NICU admission—4.3% vs. 2.7% vs. 1.6%
Bleicher et al., 2021 [74]Prospective cohort
Israel
Pfizer
Vaccinated—202
Unvaccinated—124
Outcomes presented in vaccinated vs. unvaccinated, respectively
Bookstein et al., 2021 [44]Observational case-control IsraelPfizer
Vaccinated—57
Preterm birth—0%
FGA/SGA—5.3%
Hypertensive disorder of pregnancy—1.8%
Caesarean section rate—17.6%
Stillbirth/Neonatal death—0%
NICU admission—3.5%
Dick et al., 2022 [75]Retrospective—IsraelVaccinated (Pfizer or Moderna)—2305 and unvaccinated—3313Outcomes presented in vaccinated vs. unvaccinated, respectively
  • SGA/FGR—6.2% vs. 7.050
  • Preterm birth—5.5% vs. 6.2% (NS)—vaccinated in 1st trimester
  • Vaccinated in 2nd trimester - Preterm birth 8.1% vs. 6.2%, p < 0.001)
  • Caesarean section—15.5% vs. 16.0% (NS)
  • Stillbirth rate 0.87% vs. 1.0% (NS)
Goldshtein et al., 2022 [76]Cohort—IsraelVaccinated—7591, unvaccinated—16,697
Pfizer
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • The relative risk of fetal malformations—0.69 (CI 44–1.04)
  • The relative risk of cardiac malformations—0.46 (CI 0.24—0.82)
  • Preterm birth rate—4.4% vs. 4.1% (NS)
  • FGA/SGA—6.6% vs. 6.7% (NS)
Kashani-Ligumsky et al., 2021 [45]Cohort
Israel
Pfizer
Vaccinated—29
Infected—29
Control—21
Vaginal delivery—89.7% in vaccinated vs. 82.8% in infected vs. 85.7% in controls (p = 0.86)
Rottensteich M et al., 2022 [50]Multicentre retrospective—IsraelVaccinated with Pfizer vaccine—712; unvaccinated—1063Outcomes presented in vaccinated vs. unvaccinated, respectively
  • Preterm birth rate—1% vs. 0.9% (NS)
  • FGR/SGA—11.4% vs. 9.2% (NS)
  • Stillbirth rate—0.7% vs. 0.5% (NS)
  • Caesarean section rate—15.%5 vs. 10.8% (p < 0.05)
  • NICU admission—4.1% vs. 4.5% (NS)
  • Composite adverse neonatal outcome—7.9% vs. 11.4% (p = 0.02)
  • Composite adverse maternal outcome—24.2% vs. 23.6% (NS)
Wainstock et al., 2021 [77]Retrospective cohort
Israel
Pfizer
Vaccinated—913
Unvaccinated—3438
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • FGA/SGA—2.8% vs. 3.8% (NS)
  • Caesarean section—19.9% vs. 17.2% (NS)
Magnus et al., 2022 [78]Case-control—NorwayVaccinated (n = 1003) [Pfizer =790; Moderna = 137; AstraZeneca = 76]
Unvaccinated—13,613
No difference in SM risk with an adjusted odd ratio of 0.81 for vaccinated versus unvaccinated for the previous 5 weeks
Magnus et al., 2022 [79]Registry-based retrospective
Norway and Sweden
Vaccinated—28,506; unvaccinated—129,015
Pfizer—20,424
Moderna—7607
AstraZeneca—475
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • FGA/SGA—7.8% vs. 8.5% (NS)
  • Preterm birth—6.2/10,000 vs. 4.9/10,000 (NS)
  • Stillbirth rate—2.1/100,000 vs. 2.4/100,000 (NS)
  • NICU admission—8.5% vs. 8.5% (NS)
Fell et al., 2022 [80]Retrospective
Canada
Vaccinated—22,660 (Pfizer—18,101, Moderna—4507, Other—52)
Vaccinated after pregnancy—44,815; Unvaccinated—30,115
Outcomes presented in vaccinated vs. unvaccinated vs. vaccinated after pregnancy, respectively
Caesarean section—30.8% vs. 28.5 % vs. 32.2% (NS)
NICU admission—11.0% vs. 12.8% vs. 13.3% (NS)
Sadarangani et al., 2022 [81]Observational cohort CanadaPregnant mRNA (Moderna and Pfizer)
  • 5597—received 1st dose
  • 3108—received 2nd dose
Non-pregnant
  • 174,765—received 1st dose
  • 91,131—received 2nd dose
Unvaccinated controls
  • Pregnant—339
  • Not pregnant—5840
Outcomes presented in vaccinated vs. unvaccinated vs. vaccinated
Miscarriage and stillbirth rate—1.5% vs. 2.1% (NS)
Any adverse pregnancy outcome—0.9% vs. 0.6% (NS)
Blakeway et al., 2022 [82]Retrospective cohort
UK
Pfizer
Vaccinated—133
Unvaccinated—399
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • Fetal abnormality—2.2% vs. 2.5% (NS)
  • FGR/SGA—12% vs. 12% (NS)
  • Caesarean section—30.8% vs. 34.1% (NS)
  • NICU admission—5.3% vs. 5.0% (NS)
Citu et al., 2022 [56]Prospective—RomaniaPfizer or Moderna
Vaccinated (n = 173), unvaccinated (n = 529)
Outcomes presented in vaccinated vs. unvaccinated, respectively
  • Preterm birth—8.1% vs. 6.9% (NS)
  • Caesarean section—11.5% vs. 13.0% (NS)
Table 4. Recommendations and positions of the various societies on COVID-19 vaccination in pregnancy and reproduction.
Table 4. Recommendations and positions of the various societies on COVID-19 vaccination in pregnancy and reproduction.
SocietyRecommendRecommended VaccineLast Updated
American College of Obstetricians and Gynaecologists (ACOG)Administration to all pre-pregnancy and throughout pregnancy and lactationModerna mRNA Vaccine
Pfizer-BioNTech mRNA (preferred over J & J Janssen vaccine)
Dated April 2022
Society of Gynecology and Obstetrics of Canada (SGOC)Administration to women during pregnancy in any trimester and while breastfeeding All available COVID-19 vaccines approved in Canada can be used during pregnancy and breastfeeding. Presently, preference is given for the use of mRNA vaccinations during pregnancy as more data on safety and efficacy during pregnancy is available for these vaccines14 March 2022
Royal Australian and New Zealand College of Obstetricians and Gynaecologists (RANZCOG)Pregnant women in Australia and Aotearoa, New Zealand priority group for COVID-19 vaccination and should be routinely offered
Women who are trying to become pregnant do not need to delay vaccination or avoid becoming pregnant after vaccination
Up to 3 doses of the Pfizer vaccine (Comirnaty) or Moderna (Spikevax) at any stage of pregnancy.
Pfizer (Comirnaty) and Moderna (Spikevax) are mRNA vaccines. Global evidence has shown that the Pfizer and Moderna vaccines are safe for pregnant women.
Novavax COVID-19 vaccine, a protein-based vaccine, can also be used in pregnancy (but is not currently approved for use in third or fourth doses). While there are no immunogenicity or safety data, there are no theoretical safety concerns relating to its use in pregnancy, since the Novavax COVID-19 vaccine, like other COVID-19 vaccines, is not a live vaccine
3 July 2022
International Federation of Gynecology and Obstetrics (FIGO)Supports offering COVID-19 vaccination to pregnant and breastfeeding womenNo specified vaccine2 March 2021
National Health Service (NHS)Administration to all pregnant and lactating women (considered high-risk group) and to those planning a pregnancyModerna mRNA Vaccine
Pfizer-BioNTech mRNA preferred over J & J Janssen vaccine
March 2022
Royal College of Obstetricians and Gynaecologists (RCOG)Strongly recommend administration to all pregnant and lactating women as well as those considering pregnancyComirnaty/Pfizer BioNTech or Moderna Spikevax mRNA vaccines, where available
Women who had already had one dose of Oxford-AstraZeneca (before they became pregnant or earlier on in pregnancy) are advised to complete vaccination with a second dose of Oxford-AstraZeneca
15 December 2022
Society of Maternal Fetal Medicine (SMFM)Administration to all unvaccinated individuals who are considering pregnancy, pregnant, recently pregnant, or lactating receive vaccination against COVID-19Pfizer or Moderna bivalent mRNA COVID-19 vaccine22 September 2022
Latin American Federation of Obstetrics and Gynecology Societies (FLASOG)Vaccination against COVID-19 should be offered to pregnant or breastfeeding women Not specified18 February 2022
European Society of Human Reproduction and embryology (ESHRE)Administration, if available to men and women attempting to conceive through assisted reproduction. Offer the COVID-19 vaccine before starting treatment or at any time during the fertility treatment or pregnancyNo particular vaccine preferred25 February 2022
American Society of Reproductive Medicine (ASRM)Administration to couples attempting pregnancy and during pregnancy
ASRM Task Force recommends that physicians should follow CDC guidance
mRNA-preferred, but people aged 18–49 who received J & J Janssen COVID-19 vaccine and booster who are not moderately or severely immunocompromised should receive a second booster dose using an mRNA COVID-19 vaccine at least 4 months after the first Janssen booster doseNovember 2021
European Board and College of Obstetrics and Gynaecology (EBCOG)The possibility of vaccination should be offered to all pregnant women after being adequately informed of the benefits and risks. EBCOG urges all Health Authorities and Governments to make vaccination available to all pregnant women wishing to take them.
EBCOG supports that COVID-19 vaccination be recommended to all breastfeeding women in the absence of a specific contraindication
Vaccine type not specified14 May 2021
Center for Disease Prevention and Control (CDC-USA)Administration to pregnant, lactating and those trying to conceivemRNA and J & J Janssen vaccinesApril 2022
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Konje, J.C.; Al Beloushi, M.; Ahmed, B. Immunisation against COVID-19 in Pregnancy and of Women Planning Pregnancy. Viruses 2023, 15, 621. https://doi.org/10.3390/v15030621

AMA Style

Konje JC, Al Beloushi M, Ahmed B. Immunisation against COVID-19 in Pregnancy and of Women Planning Pregnancy. Viruses. 2023; 15(3):621. https://doi.org/10.3390/v15030621

Chicago/Turabian Style

Konje, Justin C., Mariam Al Beloushi, and Badreldeen Ahmed. 2023. "Immunisation against COVID-19 in Pregnancy and of Women Planning Pregnancy" Viruses 15, no. 3: 621. https://doi.org/10.3390/v15030621

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop