You are on page 1of 17

Original Research ajog.

org

OBSTETRICS
Coronavirus disease 2019 vaccine response in pregnant
and lactating women: a cohort study
Kathryn J. Gray, MD, PhD; Evan A. Bordt, PhD; Caroline Atyeo, BS; Elizabeth Deriso, PhD;
Babatunde Akinwunmi, MD, MPH, MMSc; Nicola Young, BA; Aranxta Medina Baez, BS; Lydia L. Shook, MD; Dana Cvrk, CNM;
Kaitlyn James, PhD, MPH; Rose De Guzman, PhD; Sara Brigida, BA; Khady Diouf, MD; Ilona Goldfarb, MD, MPH;
Lisa M. Bebell, MD; Lael M. Yonker, MD; Alessio Fasano, MD; S. Alireza Rabi, MD; Michal A. Elovitz, MD; Galit Alter, PhD;
Andrea G. Edlow, MD, MSc

BACKGROUND: Pregnant and lactating women were excluded from RESULTS: Vaccine-induced antibody titers were equivalent in pregnant
initial coronavirus disease 2019 vaccine trials; thus, data to guide vaccine and lactating compared with nonpregnant women (pregnant, median,
decision making are lacking. 5.59; interquartile range, 4.68e5.89; lactating, median, 5.74; inter-
OBJECTIVE: This study aimed to evaluate the immunogenicity and quartile range, 5.06e6.22; nonpregnant, median, 5.62; interquartile
reactogenicity of coronavirus disease 2019 messenger RNA vaccination range, 4.77e5.98, P¼.24). All titers were significantly higher than those
in pregnant and lactating women compared with: (1) nonpregnant induced by severe acute respiratory syndrome coronavirus 2 infection
controls and (2) natural coronavirus disease 2019 infection in during pregnancy (P<.0001). Vaccine-generated antibodies were present
pregnancy. in all umbilical cord blood and breastmilk samples. Neutralizing antibody
STUDY DESIGN: A total of 131 reproductive-age vaccine recipients titers were lower in umbilical cord than maternal sera, although this finding
(84 pregnant, 31 lactating, and 16 nonpregnant women) were enrolled in a did not achieve statistical significance (maternal sera, median, 104.7;
prospective cohort study at 2 academic medical centers. Titers of severe interquartile range, 61.2e188.2; cord sera, median, 52.3; interquartile
acute respiratory syndrome coronavirus 2 spike and receptor-binding range, 11.7e69.6; P¼.05). The second vaccine dose (boost dose)
domain immunoglobulin G, immunoglobulin A, and immunoglobulin M increased severe acute respiratory syndrome coronavirus 2especific
were quantified in participant sera (n¼131) and breastmilk (n¼31) at immunoglobulin G, but not immunoglobulin A, in maternal blood and
baseline, at the second vaccine dose, at 2 to 6 weeks after the second breastmilk. No differences were noted in reactogenicity across the groups.
vaccine, and at delivery by Luminex. Umbilical cord sera (n¼10) titers CONCLUSION: Coronavirus disease 2019 messenger RNA vaccines
were assessed at delivery. Titers were compared with those of pregnant generated robust humoral immunity in pregnant and lactating women,
women 4 to 12 weeks from the natural infection (n¼37) by enzyme-linked with immunogenicity and reactogenicity similar to that observed in
immunosorbent assay. A pseudovirus neutralization assay was used to nonpregnant women. Vaccine-induced immune responses were statisti-
quantify neutralizing antibody titers for the subset of women who delivered cally significantly greater than the response to natural infection. Immune
during the study period. Postvaccination symptoms were assessed via transfer to neonates occurred via placenta and breastmilk.
questionnaire. Kruskal-Wallis tests and a mixed-effects model, with
correction for multiple comparisons, were used to assess differences Key words: antibodies, breastfeeding, breastmilk, cord blood, COVID-
among groups. 19 vaccine, maternal immunity, mRNA, neonatal immunity, pregnancy

Introduction pregnant women compared with their Pregnant women have long been left out
More than 73,600 infections and 80 nonpregnant counterparts, with an of therapeutic and vaccine research,
maternal deaths have occurred in preg- increased risk of hospital admission, reportedly owing to heightened safety
nant women in the United States alone as intensive care unit stay, and death.2 concerns in this population.5e8
of March 1, 2021.1 Severe acute respira- Despite their higher risk, pregnant and Although the American College of Ob-
tory syndrome coronavirus 2 (SARS- lactating women were not included in stetricians and Gynecologists and the
CoV-2) infection is more severe in any initial coronavirus disease 2019 Society for Maternal-Fetal Medicine
(COVID-19) vaccine trials, although the encouraged the Food and Drug Admin-
first vaccine trial began in pregnant istration to include pregnant women in
Cite this article as: Gray KJ, Bordt EA, Atyeo C, et al.
Coronavirus disease 2019 vaccine response in pregnant
women in February 2021 (Pfizer/Bio- the COVID-19 vaccine emergency use
and lactating women: a cohort study. Am J Obstet NTech, ClinicalTrials.gov identifier: authorization (EUA) owing to the risk of
Gynecol 2021;225:303.e1-17. NCT04754594). increased disease severity in this popu-
The COVID-19 pandemic has given lation, evidence about vaccine immu-
0002-9378
ª 2021 The Author(s). Published by Elsevier Inc. This is an rise to hundreds of vaccine platforms in nogenicity to guide patient decision
open access article under the CC BY license (http:// development to fight SARS-CoV-2.3,4 making and provider counseling is
creativecommons.org/licenses/by/4.0/). However, few of these platforms have lacking.9e11 In particular, given the
https://doi.org/10.1016/j.ajog.2021.03.023
been tested or are specifically designed to novelty of the first emergency approved
elicit immunity in vulnerable pop- COVID-19 vaccines, both of which use
ulations, including pregnant women. messenger RNA (mRNA) to deliver

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e1


Original Research OBSTETRICS ajog.org

at 80 C. Breastmilk was collected by


AJOG at a Glance the lactating participant into study-
Why was this study conducted? provided breastmilk bottles or breast-
Because pregnant and lactating women were excluded from initial coronavirus milk bags depending on volume.
disease 2019 (COVID-19) vaccine trials, data are lacking regarding vaccine effi- Breastmilk was centrifuged at 2000 rpm
cacy and infant humoral protection in this population. at 4 C for 25 minutes, supernatant was
aliquoted into cryogenic vials and stored
Key findings at 80 C.
Pregnant and lactating women elicited comparable vaccine-induced humoral
immune responses with nonpregnant controls and generated higher antibody Antibody quantification
titers than those observed after severe acute respiratory syndrome coronavirus 2 Antibody quantification was performed
infection in pregnancy. Vaccine-generated antibodies were present in umbilical as described previously.14 Briefly, a
cord blood and breastmilk after maternal vaccination. multiplexed Luminex assay was used to
determine relative titer of antigen-
What does this add to what is known? specific isotypes and subclasses using
This study provides data from a large cohort on maternal antibody generation in the following antigens: SARS-CoV-2 re-
response to COVID-19 vaccination, compares vaccine-generated immunity with ceptor-binding domain (RBD), S1, and
that from natural infection in pregnancy, and suggests that vaccination of S2 (all Sino Biologic, Beijing, China),
pregnant and lactating women can confer robust maternal and neonatal and SARS-CoV-2 spike (LakePharma,
immunity. Inc, San Carlos, CA). Antigen-specific
antibody titers were log10 transformed
for time course analyses. Phosphate-
SARS-CoV-2 spike to educate the im- COVID-19 vaccine doses, type of buffer saline (PBS) background in-
mune system,12,13 it remains unclear COVID-19 vaccine received (BNT162b2 tensity was reported for each antigen as a
whether this novel vaccine approach will Pfizer/BioNTech or mRNA-1273 Mod- threshold for positivity. Titers resulting
drive immunity in the context of preg- erna/National Institutes of Health from natural infection and vaccination-
nancy and whether antibodies will be [NIH]), and side effects after each vac- induced antibodies against SARS-CoV-
transferred efficiently to neonates via the cine dose (injection site soreness, injec- 2 RBD and spike were quantified from
cord and breastmilk. Here, vaccine- tion site skin reaction or rash, headache, the same plate using enzyme-linked
induced immunity was profiled in myalgias, fatigue, fever or chills, immunosorbent assay as previously
vaccinated pregnant, lactating, and allergic reaction, or others [reaction described.15 Additional detail regarding
nonpregnant controls compared with detailed]). A cumulative symptom and antibody quantification may be found in
women infected with SARS-CoV-2 dur- reactogenicity score was generated by Supplemental Methods.
ing pregnancy. assigning 1 point to each side effect.
Antibody neutralization assay
Materials and Methods Sample collection and processing On the morning of the experiment,
Study design Blood and breastmilk from lactating 17,000 angiotensin-converting enzyme 2
Women at 2 tertiary care centers were women were collected at V0 (at the time (ACE2) cells were plated in each well of a
approached for enrollment in an insti- of the first vaccine dose/baseline), at V1 flat-bottom 96-well plate in 100 mL of
tutional review boardeapproved (at the time of the second vaccine dose/ D10 (Dulbecco’s modified Eagle medi-
COVID-19 pregnancy and lactation “prime” profile), at V2 (2e6 weeks after umþ10% fetal bovine serum). Notably, 6
biorepository study between December the second vaccine dose/“boost” profile), hours later, the serum samples were heat

17, 2020, and February 23, 2021. Eligible and at delivery (for pregnant partici- inactivated by incubation at 56 C for 1
women were: (1) pregnant, (2) lactating, pants who delivered during the study hour. A solution containing virus at 1.9 ng
or (3) nonpregnant and of reproductive time frame). Umbilical cord blood was equivalent of p24 per mL was prepared in
age (18e45 years), 18 years old, able to also collected at delivery for pregnant D10. The heat-inactivated serum was
provide an informed consent, and participants. The V2 time point reflects diluted in this virus-containing media
receiving the COVID-19 vaccine. full antibody complement, achieved 1 1:5-fold, and then 3-fold serial dilutions
week after Pfizer/BioNTech and 2 weeks were done in the same virus-containing
Participants and procedures after Moderna/NIH.12,13 Blood was media. The virus and serum samples
Eligible study participants were identi- collected by venipuncture (or from the were incubated at 37 C for 2 hours; 50 mL
fied by practitioners at the participating umbilical vein following delivery for of the virus-serum mix was then added to
hospitals or were self-referred. A study cord blood) into serum separator tubes. the ACE2 cells. Therefore, the lowest final
questionnaire was administered to assess Blood was centrifuged at 1000 g for 10 dilution of each serum sample is 15-fold.
pregnancy and lactation status, previous min at room temperature. Sera were The cells were incubated at 37 C for 48
SARS-CoV-2 infection, timing of aliquoted into cryogenic vials and stored hours, and the red fluorescent protein was

303.e2 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

quantified using the flow cytometer (BD with natural SARS-CoV-2 infection in received both vaccine doses (median,
Accuri C6, BD Biosciences, San Jose, CA). pregnancy are detailed in Supplemental 36.5 days (IQR, 30e42) from the first
Additional details about this assay may be Table 1. These participants all had vaccine and 14 days (IQR, 11e16) from
found in the Supplemental Methods. symptomatic SARS-CoV-2 with known the second vaccine). One participant
timing of infection. delivered 17 days after vaccine 1, with
Statistical analyses spontaneous preterm labor at 35 weeks’
Participant characteristics were summa- Vaccination characteristics gestation. Lactating participant charac-
rized with frequency statistics. Contin- At the time of the study, 2 COVID-19 teristics are detailed in Table 2.
uous outcome measures were reported as vaccines had received EUA: Pfizer/Bio-
either mean (standard deviation) or me- NTech (Mainz, Germany) and Moderna The maternal vaccine response
dian (interquartile range [IQR]). Corre- (Cambridge, MA). Both vaccines use IgM, IgG, and IgA responses to the spike
lation analyses were performed using mRNA to deliver the SARS-CoV-2 spike (S), RBD, S1 segment of S, and S2
Spearman coefficients. Within- and antigen to the immune system,12,13 rep- segment of S were measured. A signifi-
between-group analyses of log10 trans- resenting a novel vaccine platform never cant rise in all isotypes across all antigens
formed antibody levels in serum or before tested in pregnancy. Although was observed from V0 to V1, with a
breastmilk across multiple time points mRNA vaccines have shown highly further rise in IgG levels from V1 to V2
were evaluated by a repeated measures effective immune induction in nonpreg- in both the pregnant and lactating
mixed-effects model, followed by post nant adults, the immunogenicity and groups (Figure 1, AeD; Supplemental
hoc Tukey’s multiple comparisons test. reactogenicity of this platform in preg- Figure 1). Spike titers rose more rapidly
Differences between paired maternal and nancy remain unclear. An equivalent than RBD titers after the first (V1/prime
cord sera immunoglobulin (Ig) G and number of pregnant women receiving the time point) and second (V2/boost time
neutralization titers were evaluated by Pfizer/BioNTech and Moderna vaccines point) vaccine dose, but the magnitude
Wilcoxon matched-pairs signed rank test. were included in our study. Of pregnant of the response did not differ across
Statistical significance was defined as participants, the mean gestational age at pregnant or lactating women. In contrast
P<.05. Statistical analyses were per- the first vaccine dose was 23.2 weeks, to IgG responses, IgM and IgA responses
formed using GraphPad Prism 9 (San with 11 women (13%) receiving their were induced robustly after the prime
Diego, CA) and Stata/IC version 16.1 first vaccine dose in the first trimester, 39 and were poorly induced after boosting,
(College Station, TX). (46%) in the second trimester, and 34 across all groups (Figure 1, C and D).
(40%) in the third trimester. Side effect Higher S- and RBD-specific IgA re-
Results profiles between participant groups sponses were noted in Moderna vacci-
From December 17, 2020, to March 2, following vaccination were similar and nees than Pfizer/BioNTech vaccinees
2021, samples were obtained from 131 are detailed in Table 1. The cumulative (Supplemental Figure 2, AeC), poten-
enrolled participants: 84 pregnant, 31 symptom score after the first dose in all 3 tially related to the extended boosting
lactating, and 16 nonpregnant groups was low. After the second dose, window used for the Moderna vaccine.
reproductive-age women. Of the preg- there was no significant difference be- By 2 weeks after the second vaccine, the
nant vaccine recipients, 13 delivered tween groups with respect to cumulative dominant serum antibody response was
during the study time frame, and cord symptom score (median, 2 (IQR, 1e3); 3 IgG for pregnant, lactating, and
blood was collected at delivery from 10. (IQR, 2e4); and 2.5 (IQR, 1e4.5) in nonpregnant women (Figure 1, E;
Banked sera from 37 pregnant women pregnant, lactating, and nonpregnant Supplemental Figure 1, C). Vaccine-
infected with SARS-CoV-2 in pregnancy groups respectively; P¼.40). Vaccine- induced maternal antibody titers in
and enrolled between March 24, 2020, related fevers or chills were reported by sera did not differ by trimester of vacci-
and December 11, 2020, were included 32% of pregnant women (25 of 77) after nation (Supplemental Figure 3). Strik-
as a second comparison group. the boost dose and 50% of nonpregnant ingly higher levels of SARS-CoV-2
women (8 of 16) (P¼.25). antibodies were observed in all vacci-
Participant characteristics nated women compared with pregnant
Participant demographic and clinical Delivery outcomes and women with natural infection 4 to 12
characteristics, sampling time points, characteristics of lactating women weeks before (Figure 1, F) (Kruskal-
and side effect profiles are presented in Delivery information for the 13 pregnant Wallis P<.001), highlighting the robust
Table 1. The study population consisted participants who delivered during the humoral immune responses induced by
primarily of white, non-Hispanic study period is detailed in Table 2. All 13 mRNA vaccination.
women, reflecting the healthcare were vaccinated in the third trimester.
worker population at the 2 hospitals. A Notably, 3 women delivered at hospitals Impact of maternal vaccination on
total of 5 participants reported previous other than the study sites, and cord breastmilk antibody transfer
SARS-CoV-2 infection: 2 pregnant, 2 blood samples were not available. Of the mRNA vaccination resulted in the in-
lactating, and 1 nonpregnant. The 10 umbilical cord blood samples avail- duction of antibodies in the circulation
characteristics of the comparison group able for analysis, 9 of 10 mothers had of vaccinated women (Figure 1).

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e3


Original Research OBSTETRICS ajog.org

TABLE 1
Cohort demographic characteristics
Characteristic Nonpregnant (n¼16), n (%) Pregnant (n¼84), n (%) Lactating (n¼31), n (%)
Participant age, mean (SD), y 38.4 (8.3) 34.1 (3.3) 34.6 (2.6)
Race
White 12 (75) 75 (89) 27 (87)
Black 2 (12) 2 (2) 0 (0)
Asian 0 (0) 6 (7) 2 (6)
Multiracial 0 (0) 1 (1) 1 (3)
Other 1 (6) 0 (0) 1 (3)
Unknown 1 (6) 0 (0) 0 (0)
Ethnicity
Hispanic or Latino 0 (0) 5 (6) 2 (6)
Not Hispanic or Latino 14 (88) 79 (94) 28 (90)
Unknown or not reported 2 (12) 0 (0) 1 (3)
Maternal comorbidities
Chronic hypertension 1 (6) 3 (4) 3 (10)
Diabetes mellitus or gestational diabetes 0 (0) 3 (4) 3 (10)
BMI of >30 kg/m 2
2 (12) 10 (12) 3 (10)
Asthma 2 (12) 16 (19) 7 (23)
Immunosuppression/cancer 0 (0) 3 (4) 0 (0)
Previous SARS-CoV-2 infection 1 (6) 2 (2) 2 (6)
Vaccine type
Pfizer-BioNTech 8 (50) 41 (49) 16 (52)
Moderna 8 (50) 43 (51) 15 (48)
Gestational age at first vaccine dose n/a 23.2 (16.3e32.1) n/a
Trimester of first vaccine dose n/a n/a
- First 11 (13)
- Second 39 (46)
- Third 34 (40)
Time points for blood collection
- Baseline/at first dose (V0) 1 (6) 31 (37) 14 (45)
- At second dose (V1) 15 (94) 78 (93) 26 (84)
- 2e5.5 wk after second dose (V2) 16 (100) 17 (20) 13 (42)
Time points for milk collection
- Baseline or at first dose (V0) — 3 (4) 16 (52)
- At second dose (V1) — 26 (31) 28 (90)
- 2e5.5 wk after second dose (V2) — 0 (0) 13 (42)
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021. (continued)

However, whether these antibodies were breastmilk of lactating mothers IgM levels did not increase with boost-
transferred efficiently to infants (Figure 2, AeC). Robust induction of ing, in synchrony with a minimal boost
remained unclear. Thus, we next exam- IgG, IgA, and IgM was observed after the in these isotypes in serum (Figure 1, C
ined the levels of antibodies in prime and boost. Interestingly, IgA and and D; Supplemental Figure 1, AeE).

303.e4 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

TABLE 1
Cohort demographic characteristics (continued)
Characteristic Nonpregnant (n¼16), n (%) Pregnant (n¼84), n (%) Lactating (n¼31), n (%)
Side effects at first vaccine dosea
- Injection site soreness 12 (75) 73 (88) 20 (67)
- Injection site reaction or rash 0 (0) 1 (1) 0 (0)
- Headache 5 (31) 7 (8) 9 (30)
- Muscle aches 2 (12) 2 (2) 4 (13)
- Fatigue 6 (38) 12 (14) 4 (13)
- Fever or chills 1 (6) 1 (1) 1 (3)
- Allergic reaction 0 (0) 0 (0) 0 (0)
- Otherb 2 (6) 3 (4) 0 (0)
c
Side effects at second vaccine dose
- Injection site soreness 12 (75) 44 (57) 17 (61)
- Injection site reaction or rash 0 (0) 1 (1) 0 (0)
- Headache 6 (38) 25 (32) 11 (39)
- Muscle aches 7 (44) 37 (48) 16 (57)
- Fatigue 9 (56) 41 (53) 14 (50)
- Fever or chills 8 (50) 25 (32) 12 (43)
- Allergic reaction 0 (0) 1 (1) 0 (0)
d
- Other 2 (12) 7 (9) 7 (25)
BMI, body mass index; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; SD, standard deviation.
a
Not all participants provided side effect data after the first dose: 2 patients (1 pregnant, 1 lactating) did not provide information. Thus, percentages are based off of 16 nonpregnant, 79 pregnant, and
30 lactating participants; b “Other” side effects reported after vaccine dose 1: elevated heart rate, joint pain, nausea, swollen lymph node, or sore throat; c Not all participants received the second
dose at the time of analysis: 16 nonpregnant, 80 pregnant, and 29 lactating patients received the second dose. Of those who received the second dose, 4 did not provide side effect data (3 pregnant,
1 lactating). Thus, percentages are based off of 16 nonpregnant, 77 pregnant, and 28 lactating participants; d “Other” side effects reported after the vaccine dose 2: joint pain, nausea, sore throat,
dizziness/light headedness, stomach ache, night sweats, clogged ears, or swollen eyes.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

However, a boost in breastmilk IgG were detectable in 10 of 10 umbilical days from the boost dose. Interestingly,
levels was observed (Figure 2, A), cords after maternal vaccination there was a significant improvement of
concomitant with the boost observed (Figure 2, D and E). The cord with the transfer of S-, but not RBD-, specific
systemically/in maternal serum lowest spike- and RBD-specific IgG IgG1 into the cord with time from boost
(Figure 1, A). IgG1 RBD rose signifi- belonged to a mother who delivered (Figure 2, D and E), suggesting that time
cantly from V0 to V2 (3.44e3.50; between the first and second vaccine from vaccination may be an important
P¼.002) but not V0 to V1 (3.44e3.45; doses and had received her first vaccine determinant of transfer rates of specific
P¼.7) in breastmilk, and there was no dose 17 days before delivery, suggesting IgG subpopulations after immunization
significant rise in anti-RBD IgA or IgM that 2 doses may be essential to optimize in pregnancy (Supplemental Figure 5, A
in breastmilk after either dose humoral immune transfer to the and B).
(Supplemental Figure 4). Overall, these neonate. Neutralizing antibody (NAb)
data suggest that the boost may drive titers were lower in umbilical cord than Vaccine reactogenicity in
enhanced breastmilk transfer of IgG, in maternal serum, although this finding pregnancy and lactation
the setting of consistent unboosted IgA did not achieve statistical significance Composite reactogenicity score after
transfer. (Figure 2, F) (maternal sera, median, boost dose of vaccine was significantly
104.7; IQR, 61.2e188.2; cord sera, me- positively correlated with both maternal
Impact of maternal vaccination on dian, 52.3; IQR, 11.7e69.6; P¼.05). serum and breastmilk antibody titers.
placental antibody transfer Notably, 2 umbilical cords had unde- Composite symptom score after vacci-
Maternal IgG is also capable of crossing tectable NAbs: in 1 case, the mother had nation was significantly positively
the placenta to confer immunity to the not yet received vaccine 2 (17 days from correlated with maternal serum spike-
neonate. Spike- and RBD-specific IgG V1) and in the other, the mother was 7 and RBD-specific IgG1 and IgG3;

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e5


Original Research OBSTETRICS ajog.org

breastmilk anti-spike IgG1, IgG3, and


TABLE 2
IgA; and breastmilk anti-RBD IgG1
Characteristics of pregnant, delivered vaccine recipients and lactating
(Supplemental Table 2). Within the
vaccine recipients
pregnant women, medical comorbidities
Pregnant, delivered vaccine recipients (n¼13) were not significantly associated with
Characteristic
maternal serum antibody titers,
although there were relatively few med-
Gestational age at delivery, median (IQR), wk 39.3 (39e40.3) ical comorbidities in this group.
Days from the first vaccine to delivery, median (IQR) 36.5 (30e42)
Days from the second vaccine to delivery, median (IQR) a
14 (11e16) Discussion
Labor 11 (85)
Principal findings
Here, robust and comparable IgG titers
Mode of delivery were observed across pregnant, lactating,
Vaginal 10 (77) and nonpregnant controls, all of which
Cesarean 3 (23) were significantly higher than those
observed in pregnant women with pre-
Birthweight, g 3452 (563)
vious SARS-CoV-2 infection. Boosting
Adverse pregnancy outcome resulted in augmented IgG levels in the
Fetal growth restriction 0 (0) blood, translating to transfer of IgG to
Preeclampsia/gestational hypertension 0 (0) the neonate through the placenta and
breastmilk.
Preterm delivery 1 (8)
- Spontaneous 1 Results
- Medically indicated 0 The lack of boosting of IgM was likely
Composite infant morbidity b related to an expected class switching to
IgG, observed with increasing IgG titers
Supplemental oxygen/CPAP 1 (8)
observed after the boost. Conversely, the
TTN 1 (8) lack of boosting of IgA observed across
Special care nursery admission 0 all women in this study was unexpected.
NICU admission 2 (15) This lack of IgA augmentation may be
related to the intramuscular adminis-
Respiratory distress syndrome 0
tration of the vaccine, which triggers a
Necrotizing enterocolitis 0 robust induction of systemic, but not
Sepsis 0 mucosal, antibodies. However, higher
Assisted ventilation 0 levels of IgA were noted after the boost in
pregnant Moderna recipients, poten-
Seizure 0
tially attributable to enhanced class
Grade 3/4 intraventricular hemorrhage 0 switching after a longer boosting inter-
Death 0 val. Robust IgG levels were noted in all
vaccinees, and vaccine-induced IgG was
Lactating vaccine recipients (n¼31)
transferred across the placenta to the
Characteristic fetus, as has been noted in the setting of
Months after delivery, median (IQR) 7.3 (3.8e10.8) influenza, pertussis, and other vaccina-
Months after delivery tion in pregnancy.16e18 The presence of
NAb transfer in nearly all cords and
0e3 5 (16)
improved transfer with increased time
3e6 6 (19) from vaccination point to the promise of
6e>9 18 (58) mRNA vaccine-induced delivery of im-
Unknown 2 (6) munity to neonates. Transfer would
Values are expressed as number (percentage) unless indicated otherwise.
perhaps be optimized if vaccination is
CPAP, continuous positive airway pressure; IQR, interquartile range; NICU, neonatal intensive care unit; TTN, transient
administered earlier during gestation,
tachypnea of the newborn. although this needs to be directly
a
Two patients delivered before receiving the second dose (17 days after V1 and 14 days after V1, cord blood only available for the examined in future studies. Although the
patient delivering 17 days after V1); b The 1 preterm delivery accounted for the documented cases of supplemental oxygen, TTN, and
1 of the 2 NICU admissions. The other NICU admission was a term infant with growth restriction admitted for persistent hypoglycemia.
transferred levels of IgA through
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021. breastmilk did not increase with boost-
ing, IgG transfer increased significantly

303.e6 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

FIGURE 1
Maternal vaccination induces a robust SARS-CoV-2especific antibody response

AeD, Violin plots show the log10 transformed mean fluorescence intensity (MFI) for (A) IgG spike-, (B) IgG RBD-, (C) IgA spike-, and (D) IgA RBDespecific
titers across V0, V1, and V2 time points collected from nonpregnant of reproductive age (blue), pregnant (orange), or lactating (purple) participants.
Participants who received BNT 162b2 from Pfizer/BioNTech are depicted as open circles, and participants who received mRNA-1273 from Moderna/NIH
are depicted as closed circles. Differences across time points and groups were assessed by repeated measures mixed-effects model followed by post hoc
Tukey’s multiple comparisons test. The asterisk indicates P<.05, the double asterisk indicates P<.01, the triple asterisk indicates P<.001, and the
quadruple asterisk indicates P<.0001. E, Line graph showing the log10 transformed relative spike-specific titers across V0, V1, and V2 time points
collected from nonpregnant (blue), pregnant (orange), or lactating (purple) participants for IgG (circles and solid lines), IgM (open triangles and dashed
lines), and IgA (squares and dotted lines). F, Violin plots show the IgG and IgM spikeespecific titer in nonpregnant (blue), pregnant (orange), lactating
(purple), and naturally-infected pregnant (yellow) participants. Participants who received BNT 162b2 from Pfizer/BioNTech are depicted as open circles,
and participants who received mRNA-1273 from Moderna/NIH are depicted as closed circles. Differences across groups were assessed by Kruskal-
Wallis test followed by post hoc Dunn’s multiple comparisons test. The quadruple asterisk indicates P<.0001 compared with natural infection in
pregnant women.
Ig, immunoglobulin; mRNA, messenger RNA; RBD, receptor-binding domain; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

with boost, resulting in the delivery of likely reflects differences in antibody natural infection,14 qualitative changes
high levels of IgG to the neonate through profile programming across mucosally in vaccine-elicited antibodies are likely
breastmilk. Importantly, emerging data acquired natural SARS-CoV-2 infection to profoundly alter antibody transfer,
point to a critical role for breastmilk IgG vs intramuscular vaccination. Whether and immunization with a de novo anti-
in neonatal immunity against several breastmilk IgG or IgA will be more gen earlier in pregnancy is likely to in-
other vaccinatable viral pathogens critical for neonatal protection remains crease placental transfer. Understanding
including HIV, respiratory syncytial vi- unclear. vaccine-induced antibody transfer ki-
rus, and influenza.19e21 In contrast, IgA Based on what is known about other netics across all pregnancy trimesters
dominates breastmilk profiles in natural vaccines, the amount of maternal IgG will be an important direction for future
SARS-CoV-2 infection.22 The different transferred across the placenta to the research. Although timing maternal
isotype transfer profile for breastmilk cord is likely to differ by trimester of COVID-19 vaccination may not be
(IgG in vaccine, IgA in natural infection) vaccination.16,17 Based on data from possible during this phase of the

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e7


Original Research OBSTETRICS ajog.org

FIGURE 2
Placental and breastmilk transfer of vaccine-induced SARS-CoV-2 antibodies

Spike RBD Neutralizing

BNT 162b2 (Pfizer) mRNA-1273 (Moderna/NIH)

A 7
**** B 7 *
C 6
** ***
***
**
6 6
***
IgG1 Spike

IgM Spike
IgA Spike
5

5 5

4
4 4

3 3 3
V0 V1 V2 V0 V1 V2 V0 V1 V2
Breastmilk Breastmilk Breastmilk
D TR = 0.78
E TR = 0.91
F
IgG Spike Spike r=0.81, p = 0.01
IgG RBD RBD r=0.50, p = 0.17
0.5 0.4 0.15 Cord Blood IgG Titer (OD)
0.15
Cord Blood IgG Titer (OD)

0.4 0.3
0.10 0.10
0.3
0.2
0.2
0.05 0.05
0.1
0.1
0.0 0.0 0.00 0.00
M C 0 10 20 30 40 50 M C 0 10 20 30 40 50
Time from maternal V2 (days) Time from maternal V2 (days)
Cord Blood Cord Blood

AeC, Violin plots show the log10 transformed mean fluorescence intensity (MFI) for (A) IgG1-, (B) IgA-, and (C) IgM spikeespecific breastmilk titers across
V0, V1, and V2 time points. Differences across time points were assessed with repeated measures mixed-effects model followed by post hoc Tukey’s
multiple comparisons test. Participants who received BNT 162b2 from Pfizer/BioNTech are depicted as open circles, and participants who received
mRNA-1273 from Moderna/NIH are depicted as closed circles. The asterisk indicates P<.05, the double asterisk indicates P<.01, the triple asterisk
indicates P<.001, and the quadruple asterisk indicates P<.0001. DeE, Dot plots showing relative (D) spike- and (E) RBD-specific maternal blood (M)
and cord blood (C) titers of IgG1. Wilcoxon matched-pairs signed rank test was performed to determine significance. At the right of each panel, the x-axis
shows the time from the second vaccine to delivery and the y-axis shows cord blood log10 transformed titer for (D) IgG spike (purple) and (E) IgG RBD
(turquoise). Correlation was determined by Spearman correlation test. PBS background subtraction was used to determine corrected optical density (OD)
of 0.0. F, Neutralizing antibody titers (50% inhibitory dose) of maternal blood (M) and cord blood (C) are presented. Wilcoxon matched-pairs signed rank
test was performed to determine significance.
Ig, immunoglobulin; PBS, phosphate-buffer saline; RBD, receptor-binding domain; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TR, transfer ratio.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

pandemic, understanding optimal tracked by the Centers for Disease Clinical implications
timing of vaccination to augment Control and Prevention using the V-safe When considering vaccination in preg-
neonatal humoral immunity remains smartphone application. Consistent nancy, evidence regarding maternal and
important. Unlike vaccines that aim to with our observations, the V-safe data fetal benefit and potential maternal and
boost preexisting antibodies (eg, influ- indicate no significant differences in fetal harm and effects on pregnancy
enza and pertussis vaccines), optimal postvaccination reactions in pregnant vs outcomes should be weighed carefully.
timing for de novo vaccine administra- nonpregnant women at the age of 16 to Although the absolute risk of severe
tion remains unclear. Thus, as the prev- 54 years.23 Although the side effect pro- COVID-19 is low in pregnant women,
alence of SARS-CoV-2 community file of pregnant women receiving the pregnancy is a risk factor for severe dis-
spread decreases, different factors such COVID-19 vaccines was not signifi- ease.27,28 There are well-documented
as optimizing neonatal immunity via cantly different from nonpregnant maternal, neonatal, and obstetrical
placental or breastmilk transfer may be women, the relatively high incidence of risks of SARS-CoV-2 infection during
weighted more heavily to inform future fever (up to 32% after the second dose) pregnancy.29e33 These data provide a
vaccine deployment. raises a theoretical concern for pregnant compelling argument that COVID-19
After EUA for the COVID-19 mRNA recipients,24,25 although the level of risk mRNA vaccines induce similar humoral
vaccines, safety information has been remains controversial.26 immunity in pregnant and lactating

303.e8 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

women as in the nonpregnant popula- immunity similar to that observed in framework for shared decision-making. CMAJ
tion. These data do not elucidate po- nonpregnant women with similar side 2021;193:E312–4.
12. Sahin U, Muik A, Derhovanessian E, et al.
tential risks to the fetus. effect profiles. Although humoral im- COVID-19 vaccine BNT162b1 elicits human
mune response and side effects are only 2 antibody and T H 1 T cell responses. Nature
Research implications of many considerations for pregnant 2020;586:594–9.
Future studies, in larger populations women and their care providers in 13. Jackson LA, Anderson EJ, Rouphael NG,
spanning vaccine administration across weighing whether or not to be vacci- et al. An mRNA vaccine against SARS-CoV-2 -
preliminary report. N Engl J Med 2020;383:
all 3 trimesters and evaluating associated nated against COVID-19 in pregnancy, 1920–31.
fetal/neonatal transfer of IgG via cord these data confirm that the COVID-19 14. Atyeo C, Pullen KM, Bordt EA, et al.
and breastmilk, may enhance our ability mRNA vaccines result in comparable Compromised SARS-CoV-2-specific placental
to develop evidence-based recommen- humoral immune responses in pregnant antibody transfer. Cell 2021;184:628–42.e10.
dations for the administration of vac- and lactating women with those 15. Edlow AG, Li JZ, Collier A-RY, et al.
Assessment of maternal and neonatal SARS-
cines and particularly different platforms observed in nonpregnant populations.n CoV-2 viral load, transplacental antibody trans-
during pregnancy. Although limited ev- fer, and placental pathology in pregnancies
idence of antibody-dependent enhance- Acknowledgments During the COVID-19 pandemic. JAMA Netw
ment has been observed in the context of We thank Dr Anjali Kaimal and Dr Jeff Ecker for Open 2020;3:e2030455.
preexisting natural or vaccine immunity their assistance starting the COVID-19 Preg- 16. Palmeira P, Quinello C, Silveira-Lessa AL,
nancy Biorepository at Massachusetts General Zago CA, Carneiro-Sampaio M. IgG placental
in adults, future studies should carefully transfer in healthy and pathological pregnancies.
Hospital.
examine the impact of transferred im- Clin Dev Immunol 2012;2012:985646.
munity on infant immune response and 17. Wilcox CR, Holder B, Jones CE. Factors
should define the optimal window for References affecting the FcRn-mediated transplacental
immunization to empower infants with 1. CDC COVID-19 Response Team; Food and transfer of antibodies and implications for
Drug Administration. Allergic reactions including vaccination in pregnancy. Front Immunol
robust immunity.
anaphylaxis after receipt of the first dose of 2017;8:1294.
Moderna COVID-19 vaccine—United States, 18. Fouda GG, Martinez DR, Swamy GK,
Strengths and limitations December 21, 2020-January 10, 2021. MMWR Permar SR. The Impact of IgG transplacental
This study was limited by the select Morb Mortal Wkly Rep 2021;70:125–9. transfer on early life immunity. Immunohorizons
population of primarily healthcare 2. Zambrano LD, Ellington S, Strid P, et al. Up- 2018;2:14–25.
date: characteristics of symptomatic women of 19. Fouda GG, Yates NL, Pollara J, et al. HIV-
workers from 1 city in the United States, specific functional antibody responses in breast
reproductive age with laboratory-confirmed
the focused time frame with limited SARS-CoV-2 infection by pregnancy status - milk mirror those in plasma and are primarily
number of delivered participants, United States, January 22-October 3, 2020. mediated by IgG antibodies. J Virol 2011;85:
inability to assess persistent immunity, MMWR Morb Mortal Wkly Rep 2020;69: 9555–67.
and the exclusive focus on antibody titers 1641–7. 20. Mazur NI, Horsley NM, Englund JA, et al.
3. Krammer F. SARS-CoV-2 vaccines in devel- Breast milk Prefusion F immunoglobulin G as a
rather than T celledriven or other correlate of protection against respiratory syn-
opment. Nature 2020;586:516–27.
functional immunity. Future work 4. Creech CB, Walker SC, Samuels RJ. SARS- cytial virus acute respiratory illness. J Infect Dis
examining T cells and other immune CoV-2 vaccines. JAMA 2021 [Epub ahead of 2019;219:59–67.
functions may provide additional in- print]. 21. Demers-Mathieu V, Huston RK,
sights on mRNA vaccineeinduced im- 5. Bianchi DW, Kaeser L, Cernich AN. Involving Markell AM, McCulley EA, Martin RL,
pregnant individuals in clinical research on Dallas DC. Impact of pertussis-specific IgA,
munity in pregnancy and lactation. The IgM, and IgG antibodies in mother’s own
COVID-19 vaccines. JAMA 2021;325:1041–2.
strengths of this work include the pro- 6. Riley LE, Jamieson DJ. Inclusion of pregnant breast milk and donor breast milk during
vision of longitudinal data profiling and lactating persons in COVID-19 vaccination preterm infant digestion. Pediatr Res 2020.
vaccine-induced immune response efforts. Ann Intern Med 2021 [Epub ahead of https://doi.org/10.1038/s41390-020-1031-2.
across contemporaneously-recruited print]. 22. Pace RM, Williams JE, Järvinen KM, et al.
7. Beigi RH, Krubiner C, Jamieson DJ, et al. The Characterization of SARS-CoV-2 RNA, anti-
pregnant, lactating, and nonpregnant bodies, and neutralizing capacity in milk pro-
need for inclusion of pregnant women in COVID-
women; the ability to compare vaccine- 19 vaccine trials. Vaccine 2021;39:868–70. duced by women with COVID-19. mBio
induced IgG titers to those from previ- 8. Klein SL, Creisher PS, Burd I. COVID-19 2021;12:e03192e20.
ous SARS-CoV-2 infection; and the in- vaccine testing in pregnant females is neces- 23. Centers for Disease Control and Prevention.
clusion of 10 maternal/neonatal dyads, sary. J Clin Invest 2021;131:e147553. Vaccine safety. V-safe after vaccination health
9. Minkoff H, Ecker J. Balancing risks: making checker. 2021. Available at: https://www.cdc.
demonstrating transfer of vaccine- gov/vaccinesafety. Accessed March 6, 2021.
decisions for maternal treatment without data on
induced IgG (including NAbs) to the fetal safety. Am J Obstet Gynecol 2021 [Epub 24. Graham JM Jr, Edwards MJ, Edwards MJ.
neonate, with improved cord titers ach- ahead of print]. Teratogen update: gestational effects of
ieved as interval from vaccination 10. Stafford IA, Parchem JG, Sibai BM. The maternal hyperthermia due to febrile illnesses
increased. coronavirus disease 2019 vaccine in pregnancy: and resultant patterns of defects in humans.
risks, benefits, and recommendations. Am J Teratology 1998;58:209–21.
Obstet Gynecol 2021 [Epub ahead of print]. 25. Dreier JW, Andersen AM, Berg-Beckhoff G.
Conclusions 11. Zipursky JS, Greenberg RA, Maxwell C, Systematic review and meta-analyses: fever in
COVID-19 vaccination in pregnancy Bogler T. Pregnancy, breastfeeding and the pregnancy and health impacts in the offspring.
and lactation generated robust humoral SARS-CoV-2 vaccine: an ethics-based Pediatrics 2014;133:e674–88.

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e9


Original Research OBSTETRICS ajog.org

26. Sass L, Urhoj SK, Kjærgaard J, Dreier JW, pandemic and pregnancy. Am J Obstet Gynecol Received March 10, 2021; revised March 21, 2021;
Strandberg-Larsen K, Nybo Andersen AM. Fe- 2020;222:521–31. accepted March 22, 2021.
ver in pregnancy and the risk of congenital K.J.G., E.A.B., and C.A. contributed equally to this
malformations: a cohort study. BMC Pregnancy work.
Childbirth 2017;17:413. Author and article information G.A. and A.G.E. contributed equally to this work.
27. Ellington S, Strid P, Tong VT, et al. Charac- From the Department of Obstetrics and Gynecology, K.J.G. has consulted for Illumina, BillionToOne, and
teristics of women of reproductive age with Brigham and Women’s Hospital, Harvard Medical School, Aetion outside the submitted work. A.F. reported serving
laboratory-confirmed SARS-CoV-2 infection by Boston, MA (Drs Gray, Akinwunmi, and Diouf); Lurie as a cofounder of and owning stock in Alba Therapeutics
pregnancy status - United States, January 22- Center for Autism, Department of Pediatrics, Massa- and serving on scientific advisory boards for NextCure
June 7, 2020. MMWR Morb Mortal Wkly Rep chusetts General Hospital, Harvard Medical School, and Viome outside the submitted work. G.A. reported
2020;69:769–75. Boston, MA (Dr Bordt); Ragon Institute of MGH, MIT, and serving as a founder of Systems SeromYx. M.A.E. re-
28. Douedi S, Miskoff J. Novel coronavirus 2019 Harvard, Cambridge, MA (Ms Atyeo and Drs Deriso and ported serving as medical advisor for Mirvie. The
(COVID-19): a case report and review of treat- Alter); PhD Program in Virology, Division of Medical Sci- remaining authors report no conflict of interest.
ments. Medicine (Baltimore) 2020;99:e20207. ences, Graduate School of Arts & Sciences, Harvard This work was supported by the National Institutes of
29. DeBolt CA, Bianco A, Limaye MA, et al. University, Boston, MA (Ms Atyeo); Department of Ob- Health, including the Eunice Kennedy Shriver National
Pregnant women with severe or critical coronavirus stetrics and Gynecology, Massachusetts General Hospi- Institute of Child Health and Human Development (grants
disease 2019 have increased composite morbidity tal, Harvard Medical School, Boston, MA (Mss Young, R01HD100022 and 3R01HD100022-02S20 to A.G.E.)
compared with nonpregnant matched controls. Baez, Cvrk, and Brigida and Drs Shook, James, Guzman, and the National Heart, Lung, and Blood Institute (grants
Am J Obstet Gynecol 2020 [Epub ahead of print]. Goldfarb and Edlow); Vincent Center for Reproductive K08HL1469630-02 and 3K08HL146963-02S1 to
30. Hantoushzadeh S, Shamshirsaz AA, Biology, Massachusetts General Hospital, Boston, MA (Dr K.J.G.). Additional support was provided by the National
Aleyasin A, et al. Maternal death due to COVID- Shook and Edlow); Division of Infectious Diseases, Institute of Allergy and Infectious Diseases
19. Am J Obstet Gynecol 2020;223:109.e1–16. Department of Medicine, Massachusetts General Hospi- (3R37AI080289-11S1, R01AI146785, U19AI42790-01,
31. Pierce-Williams RAM, Burd J, Felder L, et al. tal, Harvard Medical School, Boston, MA (Dr Bebell); MGH U19AI135995-02, U19AI42790-01, and
Clinical course of severe and critical coronavirus Center for Global Health, and Harvard Medical School, 1U01CA260476-01 to G.A.; R01A1145840 supplement
disease 2019 in hospitalized pregnancies: a Boston, MA (Dr Bebell); Mucosal Immunology and Biology to M.A.E.); the Gates Foundation; the Massachusetts
United States cohort study. Am J Obstet Research Center, Massachusetts General Hospital, Bos- Consortium on Pathogen Readiness; the Musk Founda-
Gynecol MFM 2020;2:100134. ton, MA (Drs Yonker and Fasano); Department of Pedi- tion; the Ragon Institute of Massachusetts General Hos-
32. Juan J, Gil MM, Rong Z, Zhang Y, Yang H, atrics, Massachusetts General Hospital, Harvard Medical pital, Massachusetts Institute of Technology, and the
Poon LC. Effect of coronavirus disease 2019 School, Boston, MA (Drs Yonker and Fasano); Department Massachusetts General Hospital; and Brigham and
(COVID-19) on maternal, perinatal and neonatal of Cardiothoracic Surgery, Massachusetts General Hos- Women’s Hospital Departments of Obstetrics and
outcome: systematic review. Ultrasound Obstet pital, Harvard Medical School, Boston, MA (Dr Rabi); and Gynecology.
Gynecol 2020;56:15–27. Maternal and Child Health Research Center, Perelman Corresponding authors: Galit Alter, PhD. galter@mgh.
33. Dashraath P, Wong JLJ, Lim MXK, et al. School of Medicine, University of Pennsylvania, Phila- harvard.edu; Andrea G. Edlow, MD, MSc. aedlow@mgh.
Coronavirus disease 2019 (COVID-19) delphia, PA (Dr Elovitz). harvard.edu

303.e10 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

GLOSSARY
SARS-CoV-2: a single-stranded RNA virus that causes COVID-19
SARS-CoV-2 spike protein: a virus surface protein that mediates viral entry into cells and is composed of S1 and S2 subunits
SARS-CoV-2 receptor-binding domain (RBD): a region of the spike protein that binds to the angiotensin-converting enzyme 2 receptor on
human cells for viral entry into cells.
SARS-CoV-2 nucleocapsid (N) antigen: an antigen important for eliciting antibodies against SARS-CoV-2 during infection. A critical protein
in many parts of the viral life cycle.
COVID-19 mRNA vaccine: a vaccine designed by packaging messenger RNA (mRNA) that encodes for the SARS-CoV-2 spike protein into an
injection. The mRNA elicits an immune response against the spike protein which allows a vaccinated individual’s immune system to become
trained to recognize the spike protein and prevent infection with SARS-CoV-2.
Antibody titers: a measurement of the antibody levels generated in response to exposure to an antigen.
Immunoglobulins (IgG, IgM, IgA): antibodies are referred to by immunoglobulin type, including IgG, IgM and IgA. IgG is the most abundant
type of immunoglobulin– it is found in all body fluids and can cross the placenta. IgM is primarily found in blood and lymph and is the first
type of antibody to be generated in response to a new infection. IgA is found in mucous membranes including the respiratory and
gastrointestinal tracts, saliva, and tears. IgA is the main type of antibody found in breastmilk.
Prime vaccine dose: the first dose of a vaccine that “primes” the body to respond to a subsequent exposure.
Boost vaccine dose: an additional dose of vaccine given to “boost” the immune system. A boost dose is currently given for both approved
COVID-19 mRNA vaccines 3 to 4 weeks after the prime vaccine dose.
Immunogenicity: the ability of a foreign substance (eg, antigen or vaccine) to elicit an immune response in an individual.
Reactogenicity: the degree of physical effects following vaccination owing to the body’s immune response. These include the adverse
reaction of fever and injection site soreness.

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e11


Original Research OBSTETRICS ajog.org

Supplemental Methods SARS-CoV-2 spike, or SARS-CoV-2 N. and replaced with the HA tag
Luminex-based antibody Plates were incubated for 30 minutes at (YPYDVPDYA). The D614G mutation is
quantification room temperature and washed in wash then made in this vector using the
Antibody quantification was performed buffer (0.05% Tween-20, 400 mM Quickchange XL Site-directed Muta-
as described previously.14 Briefly, a NaCl, 50 mM Tris, pH 8.0). Plates were genesis (Agilent Technologies, Inc, Santa
multiplexed Luminex assay was used to blocked with a 1% BSA solution then Clara, CA).
determine relative titer of antigen- washed again with wash buffer. Serum
specific isotypes and subclasses using samples were diluted at 1:100 and Pseudotyped virus production and
the following antigens: severe acute res- added to the plates. Plates were incu- quantification
piratory syndrome coronavirus 2 (SARS- bated at 37 C for 30 minutes. After HEK293T cells were plated in T150
CoV-2) receptor-binding domain incubation, plates were washed, and flasks 1 night before transfection at a
(RBD), S1, S2 (all Sino Biologic), and antihuman IgG or antihuman IgM confluency of approximately 50%. The
SARS-CoV-2 spike (LakePharma). An- coupled to horseradish peroxidase next day, the cells were cotransfected
tigens were covalently linked to (Bethyl Laboratories, Montgomery, TX) with the abovementioned 3 plasmids at
carboxyl-modified Magplex Luminex was added for detection. Plates were 1:1:1 molar ratio for a total DNA con-
beads using Sulfo-NHS (N-hydrox- incubated for 30 minutes at room centration of 40 mg using the TransIT-
ysulfosuccinimide, Pierce) and ethyl temperature and washed. The ELISA LT1 Transfection Reagent (Mirus Bio,
dimethylaminopropyl carbodiimide hy- was developed with 3,30 ,5,50 -tetrame- Madison, WI). Three days later, the su-
drochloride. Antigen-coupled micro- thylbenzidine and stopped with sulfuric pernatant was collected and the virus
spheres were blocked, washed, acid. The signal was read at 450 nm and was pelleted by ultracentrifugation
resuspended in phosphate-buffer saline background corrected from a reference (100,000g over a 20% sucrose cushion,
(PBS), and stored at 4 C. wavelength of 570 nm. Units had an for 2 hours). The virus was then quan-
To form immune complexes, appro- optical density of 450 to 570. tified using the Lenti-X p24 Rapid Titer
priately diluted plasma (1:100 for Kit (Takara Bio Mountain View, CA) and

immunoglobulin (Ig) G2/3, IgA1, IgM; Neutralization assay aliquots were frozen at 80 C for future
1:500 for IgG1) or breastmilk (1:5 for Cell lines use.
IgG1, IgA1, and IgM) was added to the HEK-ACE2 are clonal cells expressing
antigen-coupled microspheres, and ACE2 receptor and are generously pro- Neutralization assay
plates were incubated overnight at 4C, vided by Michael Farzan. On the morning of the experiment,
shaking at 700 rpm. The following day, 17,000 ACE2 cells were plated in each
plates were washed with 0.1% BSA Plasmids and viral constructs well of a flat-bottom 96-well plate in
0.02% Tween-20. PE-coupled mouse The lentiviruses pseudotyped with the 100 mL of D10 (Dulbecco’s Modified
antihuman detection antibodies spike protein of SARS-CoV-2 are made Eagle Mediumþ10% fetal bovine
(Southern Biotech, Birmingham, AL) by cotransfecting HEK293T cells with 3 serum). A total of 6 hours later, the
were used to detect antigen-specific plasmids: The psPAX2 was generously serum samples were heat inactivated

antibody binding. Fluorescence was provided by Didlier Trono (Addgene by incubation at 56 C for 1 hour. A
acquired using an Intellicyt iQue, and Plasmid #12260, Addgene, Watertown, solution containing virus at 1.9 ng
relative antigen-specific antibody titer MA) and is a second-generation lenti- equivalent of p24 per mL was prepared
was log10 transformed for time course virus packaging vector. The pSin-DsRed- in D10. The heat-inactivated serum
blood and breastmilk analyses. PBS IRES-Puro are a modification of the was diluted in this virus-containing
background intensity was reported for pSin-EF-Sox2-Puro, which was gener- media 1:5-fold, and then 3-fold serial
each antigen as a threshold for ously provided by James Thomson dilutions were done in the same virus-
positivity. (Addgene plasmid # 16577). The sox2 containing media. The virus and

ORF was replaced with DsRed using serum samples were incubated at 37 C
Antibody quantification using standard cloning techniques. The vec- for 2 hours. Notably, 50 mL of the
enzyme-linked immunosorbent tors expressing the spike are made from virus-serum mix was then added to
assay PiggyBac (PB) vector generously pro- the ACE2 cells. Therefore, the lowest
Antibodies against SARS-CoV-2 RBD vided by Sahand Hormoz. The codon- final dilution of each serum sample is
and spike were quantified using optimized spike gene was amplified 15-fold. The cells were incubated at

enzyme-linked immunosorbent assay from a plasmid obtained from Sino 37 C for 48 hours, and the red fluo-
(ELISA) as previously described.15 Biologic (VG40589-UT) and cloned into rescent protein was quantified using
Briefly, plates were coated with 500 the PB vector. The C-terminal 19 amino the flow cytometer (BD Accuri C6, BD
ng/mL per well of SARS-CoV-2 RBD, acids of the spike protein were deleted Biosciences, San Jose, CA).

303.e12 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

SUPPLEMENTAL FIGURE 1
Maternal vaccination induces robust SARS-CoV-2especific antibodies in maternal serum

AeB, Violin plots show the log10 transformed mean fluorescence intensity (MFI) for (A) IgM spikee and (B) IgM RBDespecific titers across V0, V1, and V2
time points collected from nonpregnant controls (blue), pregnant (orange), or lactating (purple) patients. Participants injected with BNT 162b2 from Pfizer/
BioNTech are depicted as open circles, and participants injected with mRNA-1273 from Moderna/NIH are depicted as closed circles. Differences across
time points and groups were assessed by repeated measures mixed-effects model followed by post hoc Tukey’s multiple comparisons test. The asterisk
indicates P<.05, the double asterisk indicates P<.01, the triple asterisk indicates P<.001, and the quadruple asterisk indicates P<.0001. The dotted
line depicts PBS background level. C, Line graph showing the log10 transformed relative spike-specific titers across V0, V1, and V2 time points collected
from nonpregnant controls (blue), pregnant (orange), or lactating (purple) patients for IgG (circles and solid lines) and IgA (squares and dotted lines). The
dotted line depicts PBS background level. DeI, Violin plots show the log10 transformed (D) IgM S1e, (E) IgM S2e, (F) IgG S1e, (G), IgG S2e, (H) IgA
S1e, and (I) IgA S2especific titers across V0, V1, and V2 time points collected from nonpregnant controls (blue), pregnant (orange), or lactating (purple)
patients. Participants injected with BNT 162b2 from Pfizer/BioNTech are depicted as open circles, and participants injected with mRNA-1273 from
Moderna/NIH are depicted as closed circles. Differences across time points and groups were assessed by repeated measures mixed-effects model
followed by post hoc Tukey’s multiple comparisons test. The asterisk indicates P<.05, the double asterisk indicates P<.01, the triple asterisk indicates
P<.001, the quadruple asterisk indicates P<.0001.
Ig, immunoglobulin; NIH, National Institutes of Health; PBS, phosphate-buffer saline; SARS-CoV-2, severe acute respiratory syndrome coronavirus.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e13


Original Research OBSTETRICS ajog.org

SUPPLEMENTAL FIGURE 2
mRNA-1273 (Moderna/NIH) induces a greater IgA response than does BNT 162b2 (Pfizer/BioNTech)

AeC, Violin plots show the log10 transformed mean fluorescence intensity (MFI) for (A) IgA spikee, (B) IgG spikee, and (C) IgM spikeespecific titers
across V1 and V2 time points collected from nonpregnant (blue), pregnant (orange), or lactating (purple) participants receiving either mRNA-1273
(Moderna) or BNT 162b2 (Pfizer). Differences across time points and groups were assessed by repeated measures mixed-effects model followed by
post hoc Tukey’s multiple comparisons test. The dotted line depicts PBS background level. The asterisk indicates P<.05, the double asterisk indicates
P<.01, the triple asterisk indicates P<.001, the quadruple asterisk indicates P<.0001.
Ig, immunoglobulin; mRNA, messenger RNA; NIH, National Institutes of Health; PBS, phosphate-buffer saline.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

SUPPLEMENTAL FIGURE 3
Neither trimester of infection nor vaccination affect SARS-CoV-2 antibody
production

A, Violin plots show the IgG spikeespecific titer induced by vaccination during the first trimester
(red), second trimester (orange), or third trimester (yellow). Participants who received BNT 162b2
from Pfizer/BioNTech are depicted as open circles, and participants who received mRNA-1273 from
Moderna/NIH are depicted as closed circles. Differences across groups were assessed by Kruskal-
Wallis test. Kruskal-Wallis P¼.48. B, Violin plots show the IgG spikeespecific of naturally-infected
pregnant women infected during the second trimester (yellow) or third trimester (brown). Differences
across groups were assessed by Mann-Whitney test. Mann-Whitney P¼.48.
Ig, immunoglobulin; mRNA, messenger RNA; OD, optical density; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

303.e14 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

SUPPLEMENTAL FIGURE 4
Maternal vaccination induces SARS-CoV-2especific antibodies in breastmilk

AeI, Violin plots show the log10 transformed mean fluorescence intensity (MFI) for (A) IgG1 RBDe, (B) IgA RBDe, (C) IgM RBD, (D) IgG1 S1e, (E) IgA
S1e, and (F) IgM S1e, (G) IgG1 S2e, (H) IgA S2e, and (I) IgM S2especific breastmilk titers across V0, V1, and V2 time points. Differences across time
points were assessed with repeated measures mixed-effects model followed by post hoc Tukey’s multiple comparisons test. Participants injected with
BNT 162b2 from Pfizer/BioNTech are depicted as open circles, and participants injected with mRNA-1273 from Moderna/NIH are depicted as closed
circles. The dotted line depicts PBS background level. The asterisk indicates P<.05, the double asterisk indicates P<.01, the triple asterisk indicates
P<.001, and the quadruple asterisk indicates P<.0001.
Ig, immunoglobulin; PBS, phosphate-buffer saline; RBD, receptor-binding domain; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e15


Original Research OBSTETRICS ajog.org

SUPPLEMENTAL FIGURE 5
Transfer of SARS-CoV-2especific antibodies from maternal to umbilical
cord blood following maternal vaccination

AeB, The x-axis shows the time from V2 until delivery and the y-axis shows cord blood log10
transformed titer for (A) IgG3 spike (purple) and (B) IgG3 RBD (turquoise). Significance and rho were
determined by Spearman’s correlation test.
Ig, immunoglobulin; MFI, mean fluorescence intensity; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

SUPPLEMENTAL TABLE 1
Characteristics of pregnant women with natural COVID-19 infection
Characteristic Natural COVID-19 infection in pregnancya (N¼37)
Participant age, mean (SD) 32.5 (5.3)
Race
White 14 (39)
Black 5 (14)
Asian 1 (3)
Multiracial 1 (3)
Other 14 (39)
Unknown 1 (3)
Ethnicity
Hispanic or Latino 16 (44)
Not Hispanic or Latino 19 (53)
Unknown or not reported 1 (3)
Gravidity (including current pregnancy), median (IQR) 2 (2e3)
Parity (excluding current delivery), median (IQR) 1 (0e1)
COVID severity
Mild 18 (50)
Moderate 11 (31)
Severe 7 (19)
Gestational age at COVID diagnosis in wk, median (IQR) 30.1 (26.9e33.8)
Days from symptom onset to blood draw in d, median 62.5 (39.5e84)
(IQR)
COVID-19, coronavirus disease 2019; IQR, interquartile range; RT-PCR, reverse transcription-polymerase chain reaction; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; SD,
standard deviation.
a
Only symptomatic women or those with clinical COVID-19 included for the timing of infection from symptom onset. All women had a positive RT-PCR test result for SARS-CoV-2 by nasopharyngeal
swab.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

303.e16 American Journal of Obstetrics & Gynecology SEPTEMBER 2021


ajog.org OBSTETRICS Original Research

SUPPLEMENTAL TABLE 2
SARS-CoV-2especific antibody titer correlation with composite participant symptom score after vaccine dose 2
V2 time point antibodies (2e6 wk after vaccination)
Spearman rho P value
Maternal serum
Spike IgG1 0.25 .05
Spike IgG3 0.45 .0003
RBD IgG1 0.29 .02
RBD IgG3 0.36 .005
Breastmilk
Spike IgG1 0.49 .04
Spike IgG3 0.42 .04
Spike IgA 0.42 .04
RBD IgG1 0.42 .04
Ig, immunoglobulin; RBD, receptor-binding domain; SARS-CoV-2, severe acute respiratory syndrome coronavirus.
Gray et al. Coronavirus disease 2019 vaccination in pregnancy and lactation. Am J Obstet Gynecol 2021.

SEPTEMBER 2021 American Journal of Obstetrics & Gynecology 303.e17

You might also like