Abstract

Background

Pregnant people with coronavirus disease 2019 (COVID-19) experience higher risk for severe disease and adverse pregnancy outcomes, but no pharmacokinetic (PK) data exist to support dosing of COVID-19 therapeutics during pregnancy. We report PK and safety data for intravenous remdesivir in pregnancy.

Methods

IMPAACT 2032 was a phase 4 prospective, open-label, nonrandomized opportunistic study of hospitalized pregnant and nonpregnant women receiving intravenous remdesivir as part of clinical care. Intensive PK sampling was performed on infusion days 3, 4, or 5 with collection of plasma and peripheral blood mononuclear cells (PBMCs). Safety data were recorded from first infusion through 4 weeks after last infusion and at delivery. Geometric mean ratios (GMR) (90% confidence intervals [CI]) of PK parameters between pregnant and nonpregnant women were calculated.

Results

Fifty-three participants initiated remdesivir (25 pregnant; median gestational age, 27.6 weeks; interquartile range, 24.9–31.0 weeks). Plasma exposures of remdesivir, its 2 major metabolites (GS-704277 and GS-441524), and the free remdesivir fraction were similar between pregnant and nonpregnant participants. Concentrations of the active triphosphate (GS-443902) in PBMCs increased 2.04-fold (90% CI, 1.35–3.03) with each additional infusion in nonpregnant versus pregnant participants. Three adverse events in nonpregnant participants were related to treatment (1 grade 3; 2 grade 2 resulting in treatment discontinuation). There were no treatment-related adverse pregnancy outcomes or congenital anomalies detected.

Conclusions

Plasma remdesivir PK parameters were comparable between pregnant and nonpregnant women, and no safety concerns were identified based on our limited data. These findings suggest no dose adjustments are indicated for intravenous remdesivir during pregnancy.

Clinical Trials Registration

NCT04582266.

Pregnant people are at high risk of severe coronavirus disease 2019 (COVID-19) disease and adverse pregnancy outcomes [1–3]. Several studies have shown higher risks of hospitalization, intensive care unit admissions, mechanical ventilation, and death in comparison to nonpregnant persons [1, 2], and these risks are higher in unvaccinated persons [4] and those with comorbidities [2]. Higher rates of hypertensive disorders, cesarean delivery, preterm birth, and stillbirth have also been observed in comparison to pregnant people without COVID-19 [1–3, 5]. These poor outcomes demonstrate a critical need for safe and effective COVID-19 therapeutics in pregnant people, but data supporting their use in pregnancy continue to lag data in nonpregnant persons.

Remdesivir is an intravenously administered nucleoside prodrug analog indicated for treatment of COVID-19. Early in the pandemic, remdesivir was used for the treatment of hospitalized patients with severe COVID-19 infection [6], with clinical use later expanding to those at risk for severe disease in the outpatient setting [7]. Remdesivir is moderately protein bound (88%–94%) and hydrolyzed to the alanine intermediate, GS-704277, and then the nucleotide monophosphate (MP) analog, GS-441524-MP [8]. GS-441524-MP can either be dephosphorylated to the main metabolite, GS-441524, or phosphorylated to the active triphosphate form, GS-443902, which blocks severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication through competitive inhibition of RNA-dependent RNA polymerase. Pregnancy-specific remdesivir data are limited to clinical and birth outcomes from case series, cohort, and compassionate use studies [9, 10]. The potential impact of pregnancy-related physiologic changes on the pharmacokinetics (PK) of remdesivir and its metabolites have not been studied and need to be examined.

We report findings from IMPAACT 2032, which examined the PK and safety of intravenous remdesivir in pregnant and nonpregnant women with COVID-19. The primary objectives of this study were to describe the PK of remdesivir and its plasma metabolites and to describe clinical and safety outcomes through 4 weeks after last infusion in pregnant women. Secondary objectives were to describe these same outcomes in nonpregnant women. Other objectives included comparisons of the PK of remdesivir and its metabolites between pregnant and nonpregnant women, in addition to describing and comparing GS-443902 in peripheral blood mononuclear cells (PBMCs) and remdesivir protein binding. Birth and infant outcomes were also summarized.

METHODS

Study Design

IMPAACT 2032 was a phase 4 prospective, open-label, nonrandomized opportunistic study of hospitalized pregnant and nonpregnant women with COVID-19 prescribed remdesivir as part of clinical care in the United States (NCT04582266). The decision to prescribe remdesivir and provision of all clinical care was under the direction of the clinicians at each site who were not members of the site research team. Enrollment could occur through the day of the fourth remdesivir infusion. The study enrolled pregnant and nonpregnant women in separate arms and comprised 3 data collection periods: preinfusion, infusion, and safety follow-up (Figure 1). Written informed consent was obtained from either participants or a legally authorized representative in cases where participants of legal age were unable to directly provide written informed consent. Consent was also obtained to collect limited information on birth and infant outcomes. The study protocol (Supplementary material) was reviewed and approved by the Division of AIDS (DAIDS), National Institute of Allergy and Infectious Diseases, US National Institutes of Health, and site institutional review boards.

Study design. The preinfusion period provided baseline data collection for 48 hours prior to initiating remdesivir. The infusion period included the initiation of RDV (200 mg on day 1 and 100 mg every 24 hours thereafter) for up to 5 or 10 days as clinically indicated. The safety follow-up period was inclusive of the time from discontinuing or completing remdesivir through 4 weeks after the last infusion. Among pregnant women (Arm 1), safety and birth outcomes were collected from the onset of labor or start of the cesarean delivery through 24 hours after delivery. The delivery visit could occur during remdesivir treatment, safety follow-up, or afterward. Except for PK sampling, study procedures and data collections were largely performed by medical chart abstraction or remote contact. Abbreviations: EOI, end of infusion; IV, intravenous; PBMC, peripheral blood mononuclear cell; PK, pharmacokinetics; RDV, remdesivir.
Figure 1.

Study design. The preinfusion period provided baseline data collection for 48 hours prior to initiating remdesivir. The infusion period included the initiation of RDV (200 mg on day 1 and 100 mg every 24 hours thereafter) for up to 5 or 10 days as clinically indicated. The safety follow-up period was inclusive of the time from discontinuing or completing remdesivir through 4 weeks after the last infusion. Among pregnant women (Arm 1), safety and birth outcomes were collected from the onset of labor or start of the cesarean delivery through 24 hours after delivery. The delivery visit could occur during remdesivir treatment, safety follow-up, or afterward. Except for PK sampling, study procedures and data collections were largely performed by medical chart abstraction or remote contact. Abbreviations: EOI, end of infusion; IV, intravenous; PBMC, peripheral blood mononuclear cell; PK, pharmacokinetics; RDV, remdesivir.

Eligibility Criteria

Inclusion criteria for both study arms included hospitalization with confirmed or suspected COVID-19, and either receiving or expected to receive remdesivir as part of clinical care. Pregnant women with a viable intrauterine pregnancy of any gestational age were eligible. Nonpregnant women were required to be 18 to 45 years of age, female sex at birth, not taking gender-affirming hormone therapy, and not suspected to be pregnant. Exclusion criteria for both arms included starting or having received the fourth remdesivir dose; contraindications to remdesivir treatment; receipt of disallowed medications within 48 hours prior to entry (eg, chloroquine, hydroxychloroquine); and any other condition that would compromise participant safety or interpretation of study outcomes.

Pharmacokinetic Assessments

Intensive PK assessments were performed on a single day between infusion days 3 and 5 (Figure 1 and Supplementary Methods). PBMC cell counts were determined based on cellular DNA content [11]. Remdesivir, GS-704277, GS-441524, and GS-443902 were quantified using validated liquid chromatography tandem mass spectrometry (LC-MS/MS) methods as previously described [12, 13]. The remdesivir free fraction was determined ex vivo and α-1-acid glycoprotein (AAG) was quantified using an enzyme-linked immunosorbent assay (ELISA) method (Supplementary Methods). Plasma PK parameters were determined using noncompartmental methods (Phoenix WinNonlin version 8.3; Certara).

Clinical and Safety Outcomes

Clinical management of participants was overseen by each participant's health care team. The protocol team's clinical management committee (CMC) monitored safety reports monthly, including listings of adverse events (AEs). Safety assessments in pregnant and nonpregnant participants were inclusive of vital signs, respiratory status, and clinical laboratory results (hepatic, renal, hematologic, and any other grade 3 or 4 abnormalities). Only the most abnormal results were recorded during the 48 hours prior to initiating remdesivir (baseline), daily over each 24-hour dosing interval during the infusion period, and through the last data collection time point during the safety period. AEs were assessed by the site investigator with guidance from the treating physician, where possible, with further review and confirmation of AE assessments by the CMC. In cases where AE relationships could not be determined by the site or discordant assessments between the site and CMC existed and could not be resolved, the CMC assessment was used. The CMC adjudication was used for AE summaries. AE relationship assessments to the study drug were categorized as either related or not related, with the former being defined as a reasonable possibility that the AE was related to remdesivir, and the latter being defined as there not being a reasonable possibility. AE severity was graded according to the DAIDS Table for Grading the Severity of Adult and Pediatric Adverse Events (corrected version 2.1, July 2017) [14]. For pregnant participants, serum creatinine, creatinine clearance, and estimated glomerular filtration rate (eGFR) were graded based on measured values and not change from baseline.

Statistical Analysis

Two populations were defined for the purposes of analyses: the safety-evaluable population, inclusive of enrolled participants who received any amount of remdesivir, and the PK-evaluable population. PK-evaluable participants received the full remdesivir dose on the day of intensive sampling and had both adequate sample collection—defined as meeting the following criteria: (1) at least 3 of 4 samples from end of infusion through 3 hours postdose, (2) either the 5- or 7-hour sample, and (3) either the predose or 23-hour after end of infusion sample—and concentration results for remdesivir and GS-441524 (ie, at least 2 drug concentrations greater than the lower limit of quantitation). The target sample size of 20 PK-evaluable participants per arm was chosen to assess whether a greater than 30% difference in PK parameters was detected between pregnant and nonpregnant participants and provided sufficiently narrow confidence intervals (CIs) for a coefficient of variation up to 40%.

PK, clinical, safety, and birth outcomes were summarized using descriptive statistics by study arm, with further stratification of PK results by trimester in pregnant participants. Plasma PK parameters were compared using geometric mean ratios (GMR) with 90% CIs between pregnant and nonpregnant participants. Sparse GS-443902 concentrations in PBMCs were analyzed using generalized linear mixed effects models to account for repeated measures across multiple infusions. Safety outcomes were summarized through the end of the infusion period; 48 hours, 7 days, and 4 weeks after last infusion; and at delivery. The primary safety outcomes were any renal, hepatic, or hematologic abnormality through 7 days after last infusion, and grade 3 or higher AEs, drug-related AEs, and serious AEs through 4 weeks after last infusion. Analyses of AEs with missing data used an available case approach, wherein participants who discontinued follow-up before the time point of interest and had a relevant event were included. Birth outcomes were summarized using a complete case approach for missing data, wherein only participants with delivery data available were included in analyses. Statistical analyses were performed using SAS version 9.4 software (SAS Institute, Inc).

RESULTS

Study Population

Fifty-four participants were enrolled (26 pregnant and 28 nonpregnant) between March 2021 and December 2021. Participant disposition is outlined in Supplementary Figure 1. One pregnant participant withdrew prior to receiving remdesivir. The remaining 53 participants comprised the safety population. Baseline demographics and clinical characteristics are presented in Table 1. Most (98%) had confirmed COVID-19 by diagnostic polymerase chain reaction testing prior to initiation of remdesivir. Among pregnant participants, median (interquartile range; IQR) gestational age was 27.6 weeks (interquartile range [IQR], 24.9–31.0 weeks); 13 (52%) were enrolled during the second trimester, and 12 (48%) were enrolled during the third trimester. Compared to nonpregnant participants at baseline, pregnant participants were slightly younger and had lower weights/body mass indices. Most were on low- (64%) or high-flow oxygen therapy (30%), and none were receiving vasopressor or inotropic support at baseline.

Table 1.

Baseline Demographics and Clinical Characteristics

CharacteristicPregnant Women
(n = 25)
Nonpregnant Women
(n = 28)
Total
(n = 53)
Demographics
 Age, y33.0 (27.0–37.0)37.5 (31.5–40.5)35.0 (27.0–39.0)
 Weight, kg76.5 (71.1–93.4)100.2 (77.3–144.0)89.0 (72.6–117.9)
 BMI, kg/m230.2 (27.8–39.2)37.4 (29.4–51.6)33.2 (28.0–47.1)
Race and ethnicity
 Black and not Hispanic/Latina6 (24.0)15 (53.6)21 (39.6)
 White and Hispanic/Latina6 (24.0)4 (14.3)10 (18.9)
 Other and Hispanic/Latina6 (24.0)1 (3.6)7 (13.2)
 White and not Hispanic/Latina4 (16.0)2 (7.1)6 (11.3)
 Other and not Hispanic/Latina2 (8.0)0 (0.0)2 (3.8)
 Black and Hispanic/Latina0 (0.0)1 (3.6)1 (1.9)
 Unknown1 (4.0)5 (17.9)6 (11.3)
Baseline clinical characteristics
 Duration of symptoms prior to hospitalization, d7.0 (6.0–9.0)8.0 (6.0–9.5)8.0 (6.0–9.0)
 Duration of symptoms prior to 1st remdesivir infusion, d8.0 (6.0–9.0)8.0 (7.0–10.0)8.0 (6.0–10.0)
 Respiratory support type
  Ambient air1 (4.5)0 (0.0)1 (2.1)
  Low-flow oxygen therapy12 (54.5)18 (72.0)30 (63.8)
  High-flow oxygen therapy9 (40.9)5 (20.0)14 (29.8)
  NIPPV0 (0.0)2 (8.0)2 (4.3)
Baseline clinical laboratory tests
 Creatinine, mg/dL0.50 (0.44–0.60)0.70 (0.63–0.89)0.62 (0.50–0.74)
 ALT, U/L25.1 (14.5–44.6)26.1 (16.0–58.1)26.1 (16.0–49.1)
 AST, U/L41.1 (21.0–48.1)33.1 (29.1–61.1)38.1 (28.1–50.1)
 Lymphocytes, 109/L1.00 (0.60–1.23)0.97 (0.60–1.50)1.00 (0.60–1.36)
 Hemoglobin, g/dL11.2 (10.4–12.1)11.7 (10.5–12.4)11.5 (10.4–12.2)
 Ferritin, µg/L107 (51.7–169)237 (139–471)150 (62.6–341)
 C-reactive protein, mg/L62.0 (33.4–134.0)63.9 (25.3–104.0)63.0 (28.2–109.0)
CharacteristicPregnant Women
(n = 25)
Nonpregnant Women
(n = 28)
Total
(n = 53)
Demographics
 Age, y33.0 (27.0–37.0)37.5 (31.5–40.5)35.0 (27.0–39.0)
 Weight, kg76.5 (71.1–93.4)100.2 (77.3–144.0)89.0 (72.6–117.9)
 BMI, kg/m230.2 (27.8–39.2)37.4 (29.4–51.6)33.2 (28.0–47.1)
Race and ethnicity
 Black and not Hispanic/Latina6 (24.0)15 (53.6)21 (39.6)
 White and Hispanic/Latina6 (24.0)4 (14.3)10 (18.9)
 Other and Hispanic/Latina6 (24.0)1 (3.6)7 (13.2)
 White and not Hispanic/Latina4 (16.0)2 (7.1)6 (11.3)
 Other and not Hispanic/Latina2 (8.0)0 (0.0)2 (3.8)
 Black and Hispanic/Latina0 (0.0)1 (3.6)1 (1.9)
 Unknown1 (4.0)5 (17.9)6 (11.3)
Baseline clinical characteristics
 Duration of symptoms prior to hospitalization, d7.0 (6.0–9.0)8.0 (6.0–9.5)8.0 (6.0–9.0)
 Duration of symptoms prior to 1st remdesivir infusion, d8.0 (6.0–9.0)8.0 (7.0–10.0)8.0 (6.0–10.0)
 Respiratory support type
  Ambient air1 (4.5)0 (0.0)1 (2.1)
  Low-flow oxygen therapy12 (54.5)18 (72.0)30 (63.8)
  High-flow oxygen therapy9 (40.9)5 (20.0)14 (29.8)
  NIPPV0 (0.0)2 (8.0)2 (4.3)
Baseline clinical laboratory tests
 Creatinine, mg/dL0.50 (0.44–0.60)0.70 (0.63–0.89)0.62 (0.50–0.74)
 ALT, U/L25.1 (14.5–44.6)26.1 (16.0–58.1)26.1 (16.0–49.1)
 AST, U/L41.1 (21.0–48.1)33.1 (29.1–61.1)38.1 (28.1–50.1)
 Lymphocytes, 109/L1.00 (0.60–1.23)0.97 (0.60–1.50)1.00 (0.60–1.36)
 Hemoglobin, g/dL11.2 (10.4–12.1)11.7 (10.5–12.4)11.5 (10.4–12.2)
 Ferritin, µg/L107 (51.7–169)237 (139–471)150 (62.6–341)
 C-reactive protein, mg/L62.0 (33.4–134.0)63.9 (25.3–104.0)63.0 (28.2–109.0)

Continuous data presented as median (interquartile range) unless otherwise indicated; categorical data presented as count (percentage).

Abbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; BMI, body mass index; NIPPV, noninvasive positive pressure ventilation.

Table 1.

Baseline Demographics and Clinical Characteristics

CharacteristicPregnant Women
(n = 25)
Nonpregnant Women
(n = 28)
Total
(n = 53)
Demographics
 Age, y33.0 (27.0–37.0)37.5 (31.5–40.5)35.0 (27.0–39.0)
 Weight, kg76.5 (71.1–93.4)100.2 (77.3–144.0)89.0 (72.6–117.9)
 BMI, kg/m230.2 (27.8–39.2)37.4 (29.4–51.6)33.2 (28.0–47.1)
Race and ethnicity
 Black and not Hispanic/Latina6 (24.0)15 (53.6)21 (39.6)
 White and Hispanic/Latina6 (24.0)4 (14.3)10 (18.9)
 Other and Hispanic/Latina6 (24.0)1 (3.6)7 (13.2)
 White and not Hispanic/Latina4 (16.0)2 (7.1)6 (11.3)
 Other and not Hispanic/Latina2 (8.0)0 (0.0)2 (3.8)
 Black and Hispanic/Latina0 (0.0)1 (3.6)1 (1.9)
 Unknown1 (4.0)5 (17.9)6 (11.3)
Baseline clinical characteristics
 Duration of symptoms prior to hospitalization, d7.0 (6.0–9.0)8.0 (6.0–9.5)8.0 (6.0–9.0)
 Duration of symptoms prior to 1st remdesivir infusion, d8.0 (6.0–9.0)8.0 (7.0–10.0)8.0 (6.0–10.0)
 Respiratory support type
  Ambient air1 (4.5)0 (0.0)1 (2.1)
  Low-flow oxygen therapy12 (54.5)18 (72.0)30 (63.8)
  High-flow oxygen therapy9 (40.9)5 (20.0)14 (29.8)
  NIPPV0 (0.0)2 (8.0)2 (4.3)
Baseline clinical laboratory tests
 Creatinine, mg/dL0.50 (0.44–0.60)0.70 (0.63–0.89)0.62 (0.50–0.74)
 ALT, U/L25.1 (14.5–44.6)26.1 (16.0–58.1)26.1 (16.0–49.1)
 AST, U/L41.1 (21.0–48.1)33.1 (29.1–61.1)38.1 (28.1–50.1)
 Lymphocytes, 109/L1.00 (0.60–1.23)0.97 (0.60–1.50)1.00 (0.60–1.36)
 Hemoglobin, g/dL11.2 (10.4–12.1)11.7 (10.5–12.4)11.5 (10.4–12.2)
 Ferritin, µg/L107 (51.7–169)237 (139–471)150 (62.6–341)
 C-reactive protein, mg/L62.0 (33.4–134.0)63.9 (25.3–104.0)63.0 (28.2–109.0)
CharacteristicPregnant Women
(n = 25)
Nonpregnant Women
(n = 28)
Total
(n = 53)
Demographics
 Age, y33.0 (27.0–37.0)37.5 (31.5–40.5)35.0 (27.0–39.0)
 Weight, kg76.5 (71.1–93.4)100.2 (77.3–144.0)89.0 (72.6–117.9)
 BMI, kg/m230.2 (27.8–39.2)37.4 (29.4–51.6)33.2 (28.0–47.1)
Race and ethnicity
 Black and not Hispanic/Latina6 (24.0)15 (53.6)21 (39.6)
 White and Hispanic/Latina6 (24.0)4 (14.3)10 (18.9)
 Other and Hispanic/Latina6 (24.0)1 (3.6)7 (13.2)
 White and not Hispanic/Latina4 (16.0)2 (7.1)6 (11.3)
 Other and not Hispanic/Latina2 (8.0)0 (0.0)2 (3.8)
 Black and Hispanic/Latina0 (0.0)1 (3.6)1 (1.9)
 Unknown1 (4.0)5 (17.9)6 (11.3)
Baseline clinical characteristics
 Duration of symptoms prior to hospitalization, d7.0 (6.0–9.0)8.0 (6.0–9.5)8.0 (6.0–9.0)
 Duration of symptoms prior to 1st remdesivir infusion, d8.0 (6.0–9.0)8.0 (7.0–10.0)8.0 (6.0–10.0)
 Respiratory support type
  Ambient air1 (4.5)0 (0.0)1 (2.1)
  Low-flow oxygen therapy12 (54.5)18 (72.0)30 (63.8)
  High-flow oxygen therapy9 (40.9)5 (20.0)14 (29.8)
  NIPPV0 (0.0)2 (8.0)2 (4.3)
Baseline clinical laboratory tests
 Creatinine, mg/dL0.50 (0.44–0.60)0.70 (0.63–0.89)0.62 (0.50–0.74)
 ALT, U/L25.1 (14.5–44.6)26.1 (16.0–58.1)26.1 (16.0–49.1)
 AST, U/L41.1 (21.0–48.1)33.1 (29.1–61.1)38.1 (28.1–50.1)
 Lymphocytes, 109/L1.00 (0.60–1.23)0.97 (0.60–1.50)1.00 (0.60–1.36)
 Hemoglobin, g/dL11.2 (10.4–12.1)11.7 (10.5–12.4)11.5 (10.4–12.2)
 Ferritin, µg/L107 (51.7–169)237 (139–471)150 (62.6–341)
 C-reactive protein, mg/L62.0 (33.4–134.0)63.9 (25.3–104.0)63.0 (28.2–109.0)

Continuous data presented as median (interquartile range) unless otherwise indicated; categorical data presented as count (percentage).

Abbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; BMI, body mass index; NIPPV, noninvasive positive pressure ventilation.

Clinical Outcomes

Median length of hospitalization was slightly longer in pregnant versus nonpregnant participants (10 [IQR, 7–12] versus 7 [IQR, 5–10] days). Most participants remained on low- (range, 41%–58%) or high-flow (range, 36%–53%) oxygen therapy throughout treatment. Four participants worsened during this period: 3 pregnant participants, 1 requiring mechanical ventilation on infusion days 2–4 and the other 2 requiring vasopressor/inotropic support on infusion days 2–5; and 1 nonpregnant participant requiring nasal intermittent positive pressure ventilation. Most participants (70%) completed remdesivir through 5 days of therapy, 1 (2%) each completed 6 and 10 days, and 9 (17%) were discontinued in alignment with treatment guidelines at their clinical provider's discretion. All participants were on at least 1 other concomitant medication during remdesivir treatment, with several on dexamethasone, oral or intravenous antibiotics, immune modulators, and various other medications to manage COVID-19 symptoms (Supplementary Table 1). Of remaining participants, 2 (4%) discontinued due to AEs, 2 (4%) withdrew, and 1 (2%) was lost to follow-up.

Pharmacokinetics

Concentration-time profiles for all 3 plasma analytes were comparable between pregnant and nonpregnant women (Figure 2AD). Remdesivir area under the concentration-time curve over the 24-hour dosing interval (AUC0-24h) and volume of distribution did not significantly differ in pregnant versus nonpregnant participants, but the lower bounds of the GMR 90% CIs were not contained within the 30% equivalence margin of 70%–143% (Table 2). All other GMR 90% CIs of plasma PK parameters for remdesivir, GS-704277, and GS-441524 were contained within the predefined equivalence margin. Albumin and AAG were 30% and 27% lower in pregnant versus nonpregnant participants, respectively (Table 2). However, the free fraction of remdesivir was similar between arms (Table 2). Further stratification of plasma PK and protein binding results by trimester did not identify significant differences within or between arms (Supplementary Table 2), but plasma AUCs of remdesivir and its metabolites were numerically lower in the second trimester compared to the third trimester and nonpregnant women. GS-443902 concentrations in PBMCs appeared to plateau in pregnant participants over time, whereas concentrations in nonpregnant participants increased by 2.04-fold with each additional infusion relative to pregnant participants (Figure 3).

Concentration-time profiles for total remdesivir (A), free remdesivir (B) GS-704277 (C), and GS-441524 (D). Data presented as median concentrations for each nominal time point by arm assuming a 1-hour infusion duration. Total remdesivir plot overlaid with half maximal effective concentration (EC50) in human airway epithelial (HAE) cells [15].
Figure 2.

Concentration-time profiles for total remdesivir (A), free remdesivir (B) GS-704277 (C), and GS-441524 (D). Data presented as median concentrations for each nominal time point by arm assuming a 1-hour infusion duration. Total remdesivir plot overlaid with half maximal effective concentration (EC50) in human airway epithelial (HAE) cells [15].

GS-443902 concentrations in PBMCs. Data presented as geometric mean (geometric SD) by study arm across nominal time points and overall. Gray shaded area shows 25th and 75th percentile concentrations measured in healthy volunteers on days 5 and 10 following administration of a 200-mg dose on day 1 with 100-mg daily thereafter for 5 or 10 days (6.7 and 12.4 µM, respectively). The GMR for GS-443902 in nonpregnant versus pregnant participants increased by 2.04-fold (90% CI, 1.35–3.03) with each additional infusion. Comparisons at each time point reflect GMR (90% CI) for nonpregnant versus pregnant participants from the same generalized linear mixed effects model. Pooled results reflect up to 2 observations per participant but were overlaid to show comparisons by arm. Abbreviations: CI, confidence interval; GMR, geometric mean ratio; PBMC, peripheral blood mononuclear cell.
Figure 3.

GS-443902 concentrations in PBMCs. Data presented as geometric mean (geometric SD) by study arm across nominal time points and overall. Gray shaded area shows 25th and 75th percentile concentrations measured in healthy volunteers on days 5 and 10 following administration of a 200-mg dose on day 1 with 100-mg daily thereafter for 5 or 10 days (6.7 and 12.4 µM, respectively). The GMR for GS-443902 in nonpregnant versus pregnant participants increased by 2.04-fold (90% CI, 1.35–3.03) with each additional infusion. Comparisons at each time point reflect GMR (90% CI) for nonpregnant versus pregnant participants from the same generalized linear mixed effects model. Pooled results reflect up to 2 observations per participant but were overlaid to show comparisons by arm. Abbreviations: CI, confidence interval; GMR, geometric mean ratio; PBMC, peripheral blood mononuclear cell.

Table 2.

Plasma Pharmacokinetic Parameters for Remdesivir, GS-704277, and GS-441524 and Protein Binding by Study Arm

PK ParametersPregnant (n = 21)Nonpregnant (n = 22)GMR (90% CI)
Infusion duration, ha1.0 (0.5–1.28)1.0 (0.5–2.0)
Remdesivir
 AUClast, ng · h/mL1132 (85.7)1190 (82.7)0.95 (0.65–1.38)
 AUC0-24h, ng · h/mL1247 (87.2)1303 (49.2)0.96 (0.67–1.37)
 Cmax, ng/mL1360 (108)1237 (111)1.10 (0.70–1.73)
 Tmax, h1.17 (0.60–1.31)1.08 (0.66–1.53)0.86 (0.64–1.15)
 t1/2, h1.01 (36.0)1.09 (36.4)0.92 (0.75–1.13)
 CL, L/h80.2 (87.2)79.6 (47.4)1.01 (0.70–1.45)
 Vd, L118 (89.8)125 (57.4)0.94 (0.63–1.39)
GS-704277
 AUClast, ng · h/mL408 (47.8)424 (35.5)0.96 (0.78–1.18)
 AUC0-24h, ng · h/mL454 (29.8)437 (36.2)1.04 (0.88–1.23)
 Cmax, ng/mL217 (41.4)213 (34.1)1.02 (0.85–1.23)
 Tmax, h1.18 (0.62–1.36)1.08 (0.66–1.46)0.99 (0.79–1.24)
 t1/2, h1.33 (19.9)1.23 (30.0)1.08 (0.95–1.23)
GS-441524
 AUClast, ng · h/mL1842 (31.9)2065 (36.0)0.89 (0.75–1.06)
 AUC0-24h, ng · h/mL1836 (32.2)2046 (37.7)0.90 (0.75–1.07)
 Cmax, ng/mL113 (27.8)121 (32.0)0.93 (0.80–1.09)
 Tmax, h2.1 (2.0–3.4)2.9 (2.0–4.7)0.77 (0.61–.97)
 t1/2, h20.0 (28.6)20.3 (30.3)0.96 (0.82–1.18)
 C24h, ng/mL51.6 (38.5)57.1 (44.1)0.90 (0.73–1.12)
Protein binding
 Remdesivir free fraction, %3.43 (22.8)3.22 (26.2)1.06 (0.95–1.19)
 Albumin, g/L28.0 (26.0–30.0)36.0 (33.5–38.0)0.70 (0.56–.86)
 AAG, mg/dL94.6 (77.2–144.3)124.4 (98.9–172.4)0.73 (0.60–.88)
PK ParametersPregnant (n = 21)Nonpregnant (n = 22)GMR (90% CI)
Infusion duration, ha1.0 (0.5–1.28)1.0 (0.5–2.0)
Remdesivir
 AUClast, ng · h/mL1132 (85.7)1190 (82.7)0.95 (0.65–1.38)
 AUC0-24h, ng · h/mL1247 (87.2)1303 (49.2)0.96 (0.67–1.37)
 Cmax, ng/mL1360 (108)1237 (111)1.10 (0.70–1.73)
 Tmax, h1.17 (0.60–1.31)1.08 (0.66–1.53)0.86 (0.64–1.15)
 t1/2, h1.01 (36.0)1.09 (36.4)0.92 (0.75–1.13)
 CL, L/h80.2 (87.2)79.6 (47.4)1.01 (0.70–1.45)
 Vd, L118 (89.8)125 (57.4)0.94 (0.63–1.39)
GS-704277
 AUClast, ng · h/mL408 (47.8)424 (35.5)0.96 (0.78–1.18)
 AUC0-24h, ng · h/mL454 (29.8)437 (36.2)1.04 (0.88–1.23)
 Cmax, ng/mL217 (41.4)213 (34.1)1.02 (0.85–1.23)
 Tmax, h1.18 (0.62–1.36)1.08 (0.66–1.46)0.99 (0.79–1.24)
 t1/2, h1.33 (19.9)1.23 (30.0)1.08 (0.95–1.23)
GS-441524
 AUClast, ng · h/mL1842 (31.9)2065 (36.0)0.89 (0.75–1.06)
 AUC0-24h, ng · h/mL1836 (32.2)2046 (37.7)0.90 (0.75–1.07)
 Cmax, ng/mL113 (27.8)121 (32.0)0.93 (0.80–1.09)
 Tmax, h2.1 (2.0–3.4)2.9 (2.0–4.7)0.77 (0.61–.97)
 t1/2, h20.0 (28.6)20.3 (30.3)0.96 (0.82–1.18)
 C24h, ng/mL51.6 (38.5)57.1 (44.1)0.90 (0.73–1.12)
Protein binding
 Remdesivir free fraction, %3.43 (22.8)3.22 (26.2)1.06 (0.95–1.19)
 Albumin, g/L28.0 (26.0–30.0)36.0 (33.5–38.0)0.70 (0.56–.86)
 AAG, mg/dL94.6 (77.2–144.3)124.4 (98.9–172.4)0.73 (0.60–.88)

Data presented as geometric mean (CV%), except infusion duration which is reported as median (range), and Tmax, albumin, and AAG which are reported as median (IQR). Intensive PK results analyzed using noncompartmental analysis with linear up-log down trapezoidal rule (Phoenix WinNonlin; Certara, Inc).

Abbreviations: AAG, α-1 acid glycoprotein; AUClast, area under the concentration-time curve through the last measurable time point; AUC0-24h, area under the concentration-time curve from time 0 through 24 hours; Cmax, maximum concentration; C24h: concentration at 24 hours postdose; CI, confidence interval; CL, clearance; CV, coefficient of variation; GMR, geometric mean ratio; IQR, interquartile range; PK, pharmacokinetics; t1/2, half-life; Tmax, time to maximum concentration; Vd, volume of distribution.

aOn day of intensive PK assessment.

Table 2.

Plasma Pharmacokinetic Parameters for Remdesivir, GS-704277, and GS-441524 and Protein Binding by Study Arm

PK ParametersPregnant (n = 21)Nonpregnant (n = 22)GMR (90% CI)
Infusion duration, ha1.0 (0.5–1.28)1.0 (0.5–2.0)
Remdesivir
 AUClast, ng · h/mL1132 (85.7)1190 (82.7)0.95 (0.65–1.38)
 AUC0-24h, ng · h/mL1247 (87.2)1303 (49.2)0.96 (0.67–1.37)
 Cmax, ng/mL1360 (108)1237 (111)1.10 (0.70–1.73)
 Tmax, h1.17 (0.60–1.31)1.08 (0.66–1.53)0.86 (0.64–1.15)
 t1/2, h1.01 (36.0)1.09 (36.4)0.92 (0.75–1.13)
 CL, L/h80.2 (87.2)79.6 (47.4)1.01 (0.70–1.45)
 Vd, L118 (89.8)125 (57.4)0.94 (0.63–1.39)
GS-704277
 AUClast, ng · h/mL408 (47.8)424 (35.5)0.96 (0.78–1.18)
 AUC0-24h, ng · h/mL454 (29.8)437 (36.2)1.04 (0.88–1.23)
 Cmax, ng/mL217 (41.4)213 (34.1)1.02 (0.85–1.23)
 Tmax, h1.18 (0.62–1.36)1.08 (0.66–1.46)0.99 (0.79–1.24)
 t1/2, h1.33 (19.9)1.23 (30.0)1.08 (0.95–1.23)
GS-441524
 AUClast, ng · h/mL1842 (31.9)2065 (36.0)0.89 (0.75–1.06)
 AUC0-24h, ng · h/mL1836 (32.2)2046 (37.7)0.90 (0.75–1.07)
 Cmax, ng/mL113 (27.8)121 (32.0)0.93 (0.80–1.09)
 Tmax, h2.1 (2.0–3.4)2.9 (2.0–4.7)0.77 (0.61–.97)
 t1/2, h20.0 (28.6)20.3 (30.3)0.96 (0.82–1.18)
 C24h, ng/mL51.6 (38.5)57.1 (44.1)0.90 (0.73–1.12)
Protein binding
 Remdesivir free fraction, %3.43 (22.8)3.22 (26.2)1.06 (0.95–1.19)
 Albumin, g/L28.0 (26.0–30.0)36.0 (33.5–38.0)0.70 (0.56–.86)
 AAG, mg/dL94.6 (77.2–144.3)124.4 (98.9–172.4)0.73 (0.60–.88)
PK ParametersPregnant (n = 21)Nonpregnant (n = 22)GMR (90% CI)
Infusion duration, ha1.0 (0.5–1.28)1.0 (0.5–2.0)
Remdesivir
 AUClast, ng · h/mL1132 (85.7)1190 (82.7)0.95 (0.65–1.38)
 AUC0-24h, ng · h/mL1247 (87.2)1303 (49.2)0.96 (0.67–1.37)
 Cmax, ng/mL1360 (108)1237 (111)1.10 (0.70–1.73)
 Tmax, h1.17 (0.60–1.31)1.08 (0.66–1.53)0.86 (0.64–1.15)
 t1/2, h1.01 (36.0)1.09 (36.4)0.92 (0.75–1.13)
 CL, L/h80.2 (87.2)79.6 (47.4)1.01 (0.70–1.45)
 Vd, L118 (89.8)125 (57.4)0.94 (0.63–1.39)
GS-704277
 AUClast, ng · h/mL408 (47.8)424 (35.5)0.96 (0.78–1.18)
 AUC0-24h, ng · h/mL454 (29.8)437 (36.2)1.04 (0.88–1.23)
 Cmax, ng/mL217 (41.4)213 (34.1)1.02 (0.85–1.23)
 Tmax, h1.18 (0.62–1.36)1.08 (0.66–1.46)0.99 (0.79–1.24)
 t1/2, h1.33 (19.9)1.23 (30.0)1.08 (0.95–1.23)
GS-441524
 AUClast, ng · h/mL1842 (31.9)2065 (36.0)0.89 (0.75–1.06)
 AUC0-24h, ng · h/mL1836 (32.2)2046 (37.7)0.90 (0.75–1.07)
 Cmax, ng/mL113 (27.8)121 (32.0)0.93 (0.80–1.09)
 Tmax, h2.1 (2.0–3.4)2.9 (2.0–4.7)0.77 (0.61–.97)
 t1/2, h20.0 (28.6)20.3 (30.3)0.96 (0.82–1.18)
 C24h, ng/mL51.6 (38.5)57.1 (44.1)0.90 (0.73–1.12)
Protein binding
 Remdesivir free fraction, %3.43 (22.8)3.22 (26.2)1.06 (0.95–1.19)
 Albumin, g/L28.0 (26.0–30.0)36.0 (33.5–38.0)0.70 (0.56–.86)
 AAG, mg/dL94.6 (77.2–144.3)124.4 (98.9–172.4)0.73 (0.60–.88)

Data presented as geometric mean (CV%), except infusion duration which is reported as median (range), and Tmax, albumin, and AAG which are reported as median (IQR). Intensive PK results analyzed using noncompartmental analysis with linear up-log down trapezoidal rule (Phoenix WinNonlin; Certara, Inc).

Abbreviations: AAG, α-1 acid glycoprotein; AUClast, area under the concentration-time curve through the last measurable time point; AUC0-24h, area under the concentration-time curve from time 0 through 24 hours; Cmax, maximum concentration; C24h: concentration at 24 hours postdose; CI, confidence interval; CL, clearance; CV, coefficient of variation; GMR, geometric mean ratio; IQR, interquartile range; PK, pharmacokinetics; t1/2, half-life; Tmax, time to maximum concentration; Vd, volume of distribution.

aOn day of intensive PK assessment.

Safety Outcomes

Most participants (60%) had at least 1 AE of any grade and the majority of these occurred during the infusion period (Table 3 and Supplementary Table 3). Through 7 days after last infusion, 26% (95% CI, 10%–48%) and 20% (95% CI, 7%–41%) of pregnant and nonpregnant participants, respectively, had hematologic abnormalities comprising grade 3 or higher lymphocyte, leukocyte, or hemoglobin decreases. Several pregnant and nonpregnant participants had grade 3 or higher AEs (24% [95% CI, 8%–47%] and 17% [95% CI, 5.0%–38.8%], respectively) and serious AEs (SAE) (63% [95% CI, 41%–81%] and 54% [95% CI, 33%–74%], respectively) through 4 weeks after last infusion (Table 3 and Supplementary Table 4). No SAEs were deemed related to remdesivir. Three AEs deemed related to remdesivir occurred in nonpregnant participants. These included 1 grade 3 eGFR decrease based on actual values that later resolved, and 2 grade 2 bradycardia AEs (1 after dose 2 and the other after dose 4) resulting in treatment discontinuation.

Table 3.

Safety Outcomes Through the Infusion, Primary Safety Follow-up, and Delivery Periods

Outcome MeasurePregnant
(N = 25)
Nonpregnant
(N = 28)
n/Na (%)(95% CI)bn/Na (%)(95% CI)b
During infusion period
 Adverse event of any grade15/24 (62.5)(40.6–81.2)15/26 (57.7)(36.9–76.6)
  Renal adverse event of any grade0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
  Hepatic adverse event of any grade0/23 (0.0)(.0–14.8)1/26 (3.8)(.1–19.6)
  Hematologic adverse event of any grade6/23 (26.1)(10.2–48.4)5/25 (20.0)(6.8–40.7)
 Grade 3 or higher adverse event14/24 (58.3)(36.6–77.9)13/26 (50.0)(29.9–70.1)
 Serious adverse event3/23 (13.0)(2.8–33.6)3/25 (12.0)(2.5–31.2)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
First infusion through 7 d after last infusion
 Adverse event of any grade16/24 (66.7)(44.7–84.4)15/25 (60.0)(38.7–78.9)
  Renal adverse event of any grade0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
  Hepatic adverse event of any grade1/19 (5.3)(.1–26.0)1/24 (4.2)(.1–21.1)
  Hematologic adverse event of any grade7/22 (31.8)(13.9–54.9)6/23 (26.1)(10.2–48.4)
First infusion through 4 wk after last infusion
 Grade 3 or higher adverse event15/24 (62.5)(40.6–81.2)13/24 (54.2)(32.8–74.4)
 Serious adverse event5/21 (23.8)(8.2–47.2)4/23 (17.4)(5.0–38.8)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
During delivery period
 Maternal grade 3 or higher adverse event2/17 (11.8)(1.5–36.4)
 Serious maternal adverse event2/17 (11.8)(1.5–36.4)
 Maternal grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/17 (0.0)(.0–19.5)
Outcome MeasurePregnant
(N = 25)
Nonpregnant
(N = 28)
n/Na (%)(95% CI)bn/Na (%)(95% CI)b
During infusion period
 Adverse event of any grade15/24 (62.5)(40.6–81.2)15/26 (57.7)(36.9–76.6)
  Renal adverse event of any grade0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
  Hepatic adverse event of any grade0/23 (0.0)(.0–14.8)1/26 (3.8)(.1–19.6)
  Hematologic adverse event of any grade6/23 (26.1)(10.2–48.4)5/25 (20.0)(6.8–40.7)
 Grade 3 or higher adverse event14/24 (58.3)(36.6–77.9)13/26 (50.0)(29.9–70.1)
 Serious adverse event3/23 (13.0)(2.8–33.6)3/25 (12.0)(2.5–31.2)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
First infusion through 7 d after last infusion
 Adverse event of any grade16/24 (66.7)(44.7–84.4)15/25 (60.0)(38.7–78.9)
  Renal adverse event of any grade0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
  Hepatic adverse event of any grade1/19 (5.3)(.1–26.0)1/24 (4.2)(.1–21.1)
  Hematologic adverse event of any grade7/22 (31.8)(13.9–54.9)6/23 (26.1)(10.2–48.4)
First infusion through 4 wk after last infusion
 Grade 3 or higher adverse event15/24 (62.5)(40.6–81.2)13/24 (54.2)(32.8–74.4)
 Serious adverse event5/21 (23.8)(8.2–47.2)4/23 (17.4)(5.0–38.8)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
During delivery period
 Maternal grade 3 or higher adverse event2/17 (11.8)(1.5–36.4)
 Serious maternal adverse event2/17 (11.8)(1.5–36.4)
 Maternal grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/17 (0.0)(.0–19.5)

All adverse events were coded under medical dictionary for regulatory activities (MedDRA) version 25.0 grade: 1 = mild, 2 = moderate, 3 = severe, 4 = potentially life-threatening, 5 = death. According to protocol, from the start of first remdesivir infusion through 28 days after the last infusion, all infusion reactions of any grade, all SAEs, and all grade 3 or higher events were reported as adverse events. During the delivery period (onset of labor through 24 hours after delivery) for arm 1, all infant congenital anomalies, all SAEs, and all grade 3 or higher events were also reported as adverse events.

Abbreviations: CI, confidence interval; CMC, Clinical Management Committee; n, number of participants who experienced event(s); N, number of participants in arm 1 in each trimester at 48 hours before first infusion; SAE, serious adverse event.

aExcludes women who discontinued follow-up before the end of the time frame and did not have an event of interest.

bExact binomial confidence interval (Clopper-Pearson).

Table 3.

Safety Outcomes Through the Infusion, Primary Safety Follow-up, and Delivery Periods

Outcome MeasurePregnant
(N = 25)
Nonpregnant
(N = 28)
n/Na (%)(95% CI)bn/Na (%)(95% CI)b
During infusion period
 Adverse event of any grade15/24 (62.5)(40.6–81.2)15/26 (57.7)(36.9–76.6)
  Renal adverse event of any grade0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
  Hepatic adverse event of any grade0/23 (0.0)(.0–14.8)1/26 (3.8)(.1–19.6)
  Hematologic adverse event of any grade6/23 (26.1)(10.2–48.4)5/25 (20.0)(6.8–40.7)
 Grade 3 or higher adverse event14/24 (58.3)(36.6–77.9)13/26 (50.0)(29.9–70.1)
 Serious adverse event3/23 (13.0)(2.8–33.6)3/25 (12.0)(2.5–31.2)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
First infusion through 7 d after last infusion
 Adverse event of any grade16/24 (66.7)(44.7–84.4)15/25 (60.0)(38.7–78.9)
  Renal adverse event of any grade0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
  Hepatic adverse event of any grade1/19 (5.3)(.1–26.0)1/24 (4.2)(.1–21.1)
  Hematologic adverse event of any grade7/22 (31.8)(13.9–54.9)6/23 (26.1)(10.2–48.4)
First infusion through 4 wk after last infusion
 Grade 3 or higher adverse event15/24 (62.5)(40.6–81.2)13/24 (54.2)(32.8–74.4)
 Serious adverse event5/21 (23.8)(8.2–47.2)4/23 (17.4)(5.0–38.8)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
During delivery period
 Maternal grade 3 or higher adverse event2/17 (11.8)(1.5–36.4)
 Serious maternal adverse event2/17 (11.8)(1.5–36.4)
 Maternal grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/17 (0.0)(.0–19.5)
Outcome MeasurePregnant
(N = 25)
Nonpregnant
(N = 28)
n/Na (%)(95% CI)bn/Na (%)(95% CI)b
During infusion period
 Adverse event of any grade15/24 (62.5)(40.6–81.2)15/26 (57.7)(36.9–76.6)
  Renal adverse event of any grade0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
  Hepatic adverse event of any grade0/23 (0.0)(.0–14.8)1/26 (3.8)(.1–19.6)
  Hematologic adverse event of any grade6/23 (26.1)(10.2–48.4)5/25 (20.0)(6.8–40.7)
 Grade 3 or higher adverse event14/24 (58.3)(36.6–77.9)13/26 (50.0)(29.9–70.1)
 Serious adverse event3/23 (13.0)(2.8–33.6)3/25 (12.0)(2.5–31.2)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/23 (0.0)(.0–14.8)1/25 (4.0)(.1–20.4)
First infusion through 7 d after last infusion
 Adverse event of any grade16/24 (66.7)(44.7–84.4)15/25 (60.0)(38.7–78.9)
  Renal adverse event of any grade0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
  Hepatic adverse event of any grade1/19 (5.3)(.1–26.0)1/24 (4.2)(.1–21.1)
  Hematologic adverse event of any grade7/22 (31.8)(13.9–54.9)6/23 (26.1)(10.2–48.4)
First infusion through 4 wk after last infusion
 Grade 3 or higher adverse event15/24 (62.5)(40.6–81.2)13/24 (54.2)(32.8–74.4)
 Serious adverse event5/21 (23.8)(8.2–47.2)4/23 (17.4)(5.0–38.8)
 Grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/19 (0.0)(.0–17.6)1/23 (4.3)(.1–21.9)
During delivery period
 Maternal grade 3 or higher adverse event2/17 (11.8)(1.5–36.4)
 Serious maternal adverse event2/17 (11.8)(1.5–36.4)
 Maternal grade 3 or higher adverse event assessed as related to remdesivir by the CMC0/17 (0.0)(.0–19.5)

All adverse events were coded under medical dictionary for regulatory activities (MedDRA) version 25.0 grade: 1 = mild, 2 = moderate, 3 = severe, 4 = potentially life-threatening, 5 = death. According to protocol, from the start of first remdesivir infusion through 28 days after the last infusion, all infusion reactions of any grade, all SAEs, and all grade 3 or higher events were reported as adverse events. During the delivery period (onset of labor through 24 hours after delivery) for arm 1, all infant congenital anomalies, all SAEs, and all grade 3 or higher events were also reported as adverse events.

Abbreviations: CI, confidence interval; CMC, Clinical Management Committee; n, number of participants who experienced event(s); N, number of participants in arm 1 in each trimester at 48 hours before first infusion; SAE, serious adverse event.

aExcludes women who discontinued follow-up before the end of the time frame and did not have an event of interest.

bExact binomial confidence interval (Clopper-Pearson).

Labor and Delivery Outcomes

Delivery outcomes were available for 17 of 25 (68%) participants (Supplementary Table 5). Three maternal participants experienced AEs of any grade (17.6% [95% CI, 3.8%–43.4%]), of which 2 were hematologic abnormalities (grade 3 hemoglobin decreases). Two maternal participants experienced grade 3 or higher SAEs, including fetal heart deceleration (grade 3) and respiratory failure (grade 3), the latter of which overlapped with the safety follow-up period. One participant experienced an intrauterine fetal demise at 26.7 weeks (5.9% [95% CI, .1–28.7%]). Of the remaining live births, the median gestational age at birth was 38.5 weeks (range, 34–41 weeks). Four infants (25% [95% CI, 7.3%–52.4%]) were delivered preterm (before 37 weeks of gestation) and 2 (12.5% [95% CI, 1.6%–38.3%]) were small for gestational age (<10th percentile birth weight for gestational age). No AEs or adverse pregnancy outcomes were deemed related to remdesivir. No maternal deaths occurred, and no congenital anomalies were reported.

DISCUSSION

The plasma PK of remdesivir and its major metabolites were similar between hospitalized pregnant and nonpregnant women with COVID-19. GS-443902 concentrations in PBMCs increased by 2-fold in nonpregnant versus pregnant participants. Albumin and AAG were lower in pregnancy, but the free remdesivir fraction was similar between pregnant and nonpregnant women. Based on our limited data, no safety concerns were identified. Our PK findings collectively support use of standard adult dosing of remdesivir in pregnant individuals when clinically indicated.

Plasma PK parameters for remdesivir and its metabolites were largely comparable between arms. The AUC of remdesivir was not contained within the 70%–143% bounds, which may be due in part to the greater variability with differing infusion durations and a sicker patient population. Post hoc comparisons by trimester initially suggested some PK differences, but these did not reach statistical significance, likely due to limited sample sizes. Separate PK analyses in persons with COVID-19 have shown a variety of factors can influence the PK of remdesivir and its metabolites [16–18]. Further evaluation of demographic and clinical factors that may affect remdesivir and metabolite PK is underway, and continued refinements of physiologically based PK modeling in pregnancy should be pursued [19]. Plasma exposures were comparable between study arms and to other PK studies in nonpregnant persons with COVID-19 [16, 17]. Consistent with other PK studies in pregnancy, albumin and AAG were lower in pregnancy, but no differences in the remdesivir free fraction were detected. The free fraction measured in this study was lower than what has been reported (6%–12%) [20]. Binding preference for albumin versus AAG is not known, but a molecular docking study has suggested moderate binding potential for remdesivir by albumin [21]. AAG concentrations in our participants were also high, which may be explained in part by AAG being an acute phase reactant [22, 23]. Median measures during pregnancy in this study were elevated compared to historical data in pregnant persons without COVID-19 (approximately 95 vs approximately 45–60 mg/dL) [24–26].

The accumulation of GS-443902 concentrations in PBMCs differed between arms, appearing to plateau in pregnant participants while nonpregnant participants continued to increase with each infusion. The continued accumulation in nonpregnant women was consistent with the previously published half-life of approximately 43 hours in PBMCs [8]. The underlying mechanisms and clinical relevance behind the differing accumulation is unclear. GS-443902 concentrations in PBMCs serve as a measure of drug activation within cells [8]. Compared with healthy volunteers, GS-443902 concentrations were comparable but more variable in nonpregnant women [8], and lower in pregnant women. However, all GS-443902 concentrations in this study were above the in vitro half maximal effective concentration (EC50) of approximately 0.01 µM in human airway and lung epithelial cells [15]. Additionally, it is unknown how reflective PBMCs are of drug activation in other tissue types. Multiple factors have been shown to affect cellular concentrations of other nucleos(t)ide analogs in PBMCs, many of which overlap with the changes seen in pregnancy and COVID-19 infection [27–30]. Further investigation of factors that may affect GS-443902 concentrations, relationships between plasma and cellular concentrations, and understanding the clinical significance of this finding are warranted [19, 31].

No safety concerns were identified, and few participants discontinued remdesivir due to an AE. There were several grade 3 or higher SAEs unrelated to remdesivir use. These were largely consistent with outcomes in severe COVID-19 infection, and our enrolled population reflected known demographic and clinical risk factors for progression to severe disease [1, 2, 9, 32]. Of events related to remdesivir, bradycardia has been reported [33], which may be due in part to its adenosine analog structure and ability to bind to adenosine/P2Y purinoreceptors receptors [18]. However, COVID-19 infection has also been associated with several other adverse cardiovascular outcomes [34, 35]. No adverse pregnancy outcomes related to remdesivir were identified, and no congenital abnormalities were detected. However, pregnancy outcomes were unknown for 8 of 25 (32%) participants, and our study did not include any pregnant people in the first trimester of pregnancy. The proportion of adverse birth outcomes based on our limited data were consistent with prior literature in pregnant people with COVID-19, particularly the risk of preterm birth [1, 2]. Furthermore, collective data have not identified a higher risk of adverse pregnancy outcomes, major birth defects, or miscarriage when remdesivir is initiated in the second or third trimester [20, 36, 37]. However, there are currently insufficient data to evaluate the risk of remdesivir use when initiated during the first trimester [20].

There were strengths and limitations in this study. At the start of the pandemic, PK data with remdesivir were limited to healthy volunteers who were primarily male [8, 13]. This necessitated enrolling nonpregnant women for direct comparison of drug exposures in persons with COVID-19. However, to ensure that enrollment proceeded in a timely manner, participants were not matched based on demographic or clinical factors. Differences in baseline factors, such as weight and race/ethnicity, were apparent between arms. Given the opportunistic design with enrollment permitted through the fourth remdesivir dose, this design may have selected for participants who were already tolerating therapy compared to individuals who had poorer outcomes at treatment initiation. Alternatively, potential patients who recovered quickly may have stopped drug early and not been enrolled, thereby leading to underestimation of positive outcomes. This study was also not designed to compare clinical, safety, or adverse pregnancy outcomes. The median time to remdesivir initiation was 8 days across both arms, and earlier treatment initiation is associated with improved clinical outcomes in nonpregnant [7] and pregnant populations [10, 36]. The longer hospitalization duration in pregnant women may have contributed additional bias in reporting of safety follow-up outcomes. Overall outcomes in our study were consistent with the known disease course of severe COVID-19 infection. Pregnancy outcome data were also missing for 8 participants. As adverse outcomes occurred earlier in the safety observation period, missing outcomes may be more representative of uncomplicated births, but this is unknown.

The COVID-19 pandemic has continued to highlight the exclusion of pregnant people from treatment and vaccine studies [38]. Pregnancy PK and safety studies are key to determining drug dosing, safety, and efficacy of medications in this population. Unfortunately, pregnancy studies for new drugs are often not performed or are delayed for multiple years after initial regulatory approvals [39]. These exclusions can result in effective drugs not being used during pregnancy or used at suboptimal doses. There is a critical need to generate pregnancy PK and safety data in a timely manner, particularly during public health emergencies. IMPAACT 2032 was rapidly developed in response to the known delays in drug development that can occur in pregnant people. Ultimately, PK and safety results from this study were included in recent labeling updates for use of intravenous remdesivir during pregnancy [20], illustrating the critical role these studies have in informing appropriate treatment of pregnant people with new medications.

In conclusion, the plasma PK of remdesivir, its metabolites, and the free fraction of remdesivir were comparable between hospitalized pregnant and nonpregnant women with COVID-19, suggesting remdesivir does not require dose adjustment in pregnancy. This study addressed a significant knowledge gap in the pharmacology of COVID-19 therapeutics in pregnant people.

Supplementary Data

Supplementary materials are available at The Journal of Infectious Diseases online (http://jid.oxfordjournals.org/). Supplementary materials consist of data provided by the author that are published to benefit the reader. The posted materials are not copyedited. The contents of all supplementary data are the sole responsibility of the authors. Questions or messages regarding errors should be addressed to the author.

Notes

Acknowledgments. We thank the study participants and families, clinical providers, the IMPAACT 2032 Protocol Team, and the following study sites and personnel: Lurie Children's Hospital of Chicago (Emanuela Lartey, RN; Rohit Kalra, MS, CCRP; Lynn Yee, MD, MPH; James Etta Stewart); Bronx-Lebanon Hospital Center (Martha Cavallo, PNP; Mirza Baig, MD); Johns Hopkins University Baltimore (Aleisha Collinson-Streng, BSN, RN; Thuy Anderson, BSN, RN; Bonnie Addison, BA); SUNY Stony Brook (Barsha Chakraborty, MPH; Cecilia Avila, MD, MPH; Giuseppe Caso, MD, PhD); David Geffen School of Medicine at UCLA (Carla Janzen, MD, PhD; Michele F. Carter, BS, RN); Baylor College of Medicine/Texas Children's Hospital (Mary Paul, MD; Ruth Eser-Jose, MSN, RN, CPN; Mariam Pontifes, CCRP; Chivon McMullen Jackson, RN); Pediatric Perinatal HIV Clinical Trials Unit, Miami Fl (Nicolette Gomez, CCRP; Grace Alvarez FMD, CCRP; Charles Mitchell, MD; JoNell Potter, PhD); Emory University School of Medicine (Martina L. Badell, MD; Sierra Jordan-Thompson, MPH; Riaun Floyd, BA; LaTeshia Thomas-Seaton, MS, APRN, CCRC); University of Colorado Denver (Adriana Weinberg, MD; Shane Curran-Hays, MS; Christine Kwon, BS; Carrie Glenny, RN, BSN; ); Rush University Cook County Hospital Chicago (Mariam Aziz, MD; Maureen McNichols, RN CCRC). We also thank Gilead Sciences, Inc and the following representatives: Rich Clark, Heather Maxwell, Gina Brown, Chris Blair, and James Rooney. The University of Colorado is a Certara Center of Excellence. The Center of Excellence program supports leading institutions with Certara's state-of-the-art model-informed drug development software.

Author contributions. K. M. B., K. B., D. F. C., S. N., K. M. P., A. M. S., A. C. E., D. E. S., E. C., E. G., K. G., D. E. Y., P. J. P., N. C., B. M. B., M. M., and J. D. M. contributed to the conception, design, and oversight of the study. K. M. B., J. D. M., E. C., and B. M. B. had access to, analyzed, verified, and interpreted pharmacokinetic data. J. J., M. U. P., A. A., C. B., and J. G. D. were involved in implementation, recruitment, and oversight of study conduct at their respective study sites. K. B., D. S., F. B., K. B., B. J., and C. R. had access to data and were involved in data management and oversight. K. B. and D. E. S. performed statistical analyses and interpreted results. K. M. B., K. B., D. F. C., S. N., K. M. P., A. M. S., A. C. E., D. E. S., E. C., E. G., K. G., D. E. Y., P. J. P., N. C., K. K., R. H., B. M. B., M. M., and J. D. M. reviewed and interpreted data. All authors contributed to manuscript development and approved the final manuscript version.

Disclaimer. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health (NIH).

Data availability. The data cannot be made publicly available due to the ethical restrictions in the study's informed consent documents and in the International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) Network's approved human subjects protection plan; public availability may compromise participant confidentiality. However, data are available to all interested researchers upon request to the IMPAACT Statistical and Data Management Center's data access committee (email address: sdac.data@fstrf.org) with the agreement of the IMPAACT Network.

Financial support. This work was supported by the National Institute of Allergy and Infectious Diseases, the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), and the National Institute of Mental Health, all components of the NIH (grant numbers UM1AI068632 to International Maternal Pediatric Adolescent AIDS Clinical Trials Network (IMPAACT) Leadership and Operations Center (LOC), UM1AI068616 to IMPAACT Statistical and Data Management Center (SDMC), and UM1AI106716 to IMPAACT Laboratory Center (LC); and NICHD contract number HHSN275201800001I). Additional support was provided by Gilead Sciences, Inc.

References

1

Smith
 
ER
,
Oakley
 
E
,
Grandner
 
GW
, et al.   
Adverse maternal, fetal, and newborn outcomes among pregnant women with SARS-CoV-2 infection: an individual participant data meta-analysis
.
BMJ Glob Health
 
2023
;
8
:
e009495
.

2

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

3

Matsuo
 
K
,
Green
 
JM
,
Herrman
 
SA
,
Mandelbaum
 
RS
,
Ouzounian
 
JG
.
Severe maternal morbidity and mortality of pregnant patients with COVID-19 infection during the early pandemic period in the US
.
JAMA Netw Open
 
2023
;
6
:
e237149
.

4

Dagan
 
N
,
Barda
 
N
,
Biron-Shental
 
T
, et al.   
Effectiveness of the BNT162b2 mRNA COVID-19 vaccine in pregnancy
.
Nat Med
 
2021
;
27
:
1693
5
.

5

Smith
 
LH
,
Dollinger
 
CY
,
VanderWeele
 
TJ
,
Wyszynski
 
DF
,
Hernandez-Diaz
 
S
.
Timing and severity of COVID-19 during pregnancy and risk of preterm birth in the International Registry of Coronavirus Exposure in Pregnancy
.
BMC Pregnancy Childbirth
 
2022
;
22
:
775
.

6

Beigel
 
JH
,
Tomashek
 
KM
,
Dodd
 
LE
, et al.   
Remdesivir for the treatment of COVID-19—preliminary report.
 
N Engl J Med
 
2020
;
383
:
1813
26
.

7

Gottlieb
 
RL
,
Vaca
 
CE
,
Paredes
 
R
, et al.   
Early remdesivir to prevent progression to severe COVID-19 in outpatients
.
N Engl J Med
 
2022
;
386
:
305
15
.

8

Humeniuk
 
R
,
Mathias
 
A
,
Kirby
 
BJ
, et al.   
Pharmacokinetic, pharmacodynamic, and drug-interaction profile of remdesivir, a SARS-CoV-2 replication inhibitor
.
Clin Pharmacokinet
 
2021
;
60
:
569
83
.

9

Burwick
 
RM
,
Yawetz
 
S
,
Stephenson
 
KE
, et al.   
Compassionate use of remdesivir in pregnant women with severe coronavirus disease 2019
.
Clin Infect Dis
 
2021
;
73
:
e3996
4004
.

10

Nasrallah
 
S
,
Nguyen
 
AQ
,
Hitchings
 
L
, et al.   
Pharmacological treatment in pregnant women with moderate symptoms of coronavirus disease 2019 (COVID-19) pneumonia
.
J Matern Fetal Neonatal Med
 
2022
;
35
:
5970
7
.

11

Xiao
 
D
,
Ling
 
KHJ
,
Tarnowski
 
T
, et al.   
The determination of human peripheral blood mononuclear cell counts using a genomic DNA standard and application in tenofovir diphosphate quantitation
.
Anal Biochem
 
2019
;
585
:
113399
.

12

Xiao
 
D
,
John Ling
 
KH
,
Tarnowski
 
T
, et al.   
Validation of LC-MS/MS methods for determination of remdesivir and its metabolites GS-441524 and GS-704277 in acidified human plasma and their application in COVID-19 related clinical studies
.
Anal Biochem
 
2021
;
617
:
114118
.

13

Humeniuk
 
R
,
Mathias
 
A
,
Cao
 
H
, et al.   
Safety, tolerability, and pharmacokinetics of remdesivir, an antiviral for treatment of COVID-19, in healthy subjects
.
Clin Transl Sci
 
2020
;
13
:
896
906
.

14

US Department of Health and Human Services, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Division of AIDS
. Division of AIDS (DAIDS) table for grading the severity of adult and pediatric adverse events, corrected version 2.1, July 2017. https://rsc.niaid.nih.gov/sites/default/files/daidsgradingcorrectedv21.pdf. Accessed 1 July 2023.

15

Pruijssers
 
AJ
,
George
 
AS
,
Schafer
 
A
, et al.   
Remdesivir inhibits SARS-CoV-2 in human lung cells and chimeric SARS-CoV expressing the SARS-CoV-2 RNA polymerase in mice
.
Cell Rep
 
2020
;
32
:
107940
.

16

Humeniuk
 
R
,
Deng
 
R
,
Sorribes
 
IC
, et al.   
Pharmacokinetics inform remdesivir dosing in patients with severe renal impairment
,
abstract 514
.
Conference on Retroviruses and Opportunistic Infections
,
Seattle, WA
,
2023
.

17

Humeniuk
 
R
,
Deng
 
R
,
Sorribes
 
IC
, et al.   
Dosing recommendation for remdesivir in pediatric COVID-19 patients
,
poster 113
.
American College of Clinical Pharmacology Annual Meeting
,
Bethesda, MD
,
2022
.

18

Center for Drug Evaluation Research, U.S. Food and Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/214787Orig1s000ClinpharmR.pdf. Accessed 1 July 2023.

19

Liu
 
XI
,
Dallmann
 
A
,
Brooks
 
K
, et al.   
Physiologically-based pharmacokinetic modeling of remdesivir and its metabolites in pregnant women with COVID-19
.
CPT Pharmacometrics Syst Pharmacol
 
2023
;
12
:
148
53
.

20

Gilead Sciences
.
 
Veklury
 
[package insert]
.
Foster City, CA
:
Gilead Sciences
,
2023
.

21

Gurung
 
AB
,
Ali
 
MA
,
Lee
 
J
,
Farah
 
MA
,
Al-Anazi
 
KM
,
Sami
 
H
.
Molecular modelling studies unveil potential binding sites on human serum albumin for selected experimental and in silico COVID-19 drug candidate molecules
.
Saudi J Biol Sci
 
2022
;
29
:
53
64
.

22

Bartelink
 
IH
,
Bet
 
PM
,
Widmer
 
N
, et al.   
Elevated acute phase proteins affect pharmacokinetics in COVID-19 trials: lessons from the CounterCOVID-imatinib study
.
CPT Pharmacometrics Syst Pharmacol
 
2021
;
10
:
1497
511
.

23

Boffito
 
M
,
Back
 
DJ
,
Flexner
 
C
, et al.   
Toward consensus on correct interpretation of protein binding in plasma and other biological matrices for COVID-19 therapeutic development
.
Clin Pharmacol Ther
 
2021
;
110
:
64
8
.

24

Zhao
 
S
,
Gockenbach
 
M
,
Grimstein
 
M
, et al.   
Characterization of plasma protein alterations in pregnant and postpartum individuals living with HIV to support physiologically-based pharmacokinetic model development
.
Front Pediatr
 
2021
;
9
:
721059
.

25

Abduljalil
 
K
,
Furness
 
P
,
Johnson
 
TN
,
Rostami-Hodjegan
 
A
,
Soltani
 
H
.
Anatomical, physiological and metabolic changes with gestational age during normal pregnancy: a database for parameters required in physiologically based pharmacokinetic modelling
.
Clin Pharmacokinet
 
2012
;
51
:
365
96
.

26

Dallmann
 
A
,
Ince
 
I
,
Meyer
 
M
,
Willmann
 
S
,
Eissing
 
T
,
Hempel
 
G
.
Gestation-specific changes in the anatomy and physiology of healthy pregnant women: an extended repository of model parameters for physiologically based pharmacokinetic modeling in pregnancy
.
Clin Pharmacokinet
 
2017
;
56
:
1303
30
.

27

Anderson
 
PL
,
Kakuda
 
TN
,
Kawle
 
S
,
Fletcher
 
CV
.
Antiviral dynamics and sex differences of zidovudine and lamivudine triphosphate concentrations in HIV-infected individuals
.
AIDS
 
2003
;
17
:
2159
68
.

28

Anderson
 
PL
,
King
 
T
,
Zheng
 
JH
,
MaWhinney
 
S
.
Cytokine and sex hormone effects on zidovudine- and lamivudine-triphosphate concentrations in vitro
.
J Antimicrob Chemother
 
2008
;
62
:
738
45
.

29

Castillo-Mancilla
 
JR
,
Meditz
 
A
,
Wilson
 
C
, et al.   
Reduced immune activation during tenofovir-emtricitabine therapy in HIV-negative individuals
.
J Acquir Immune Defic Syndr
 
2015
;
68
:
495
501
.

30

Seifert
 
SM
,
Chen
 
X
,
Meditz
 
AL
, et al.   
Intracellular tenofovir and emtricitabine anabolites in genital, rectal, and blood compartments from first dose to steady state
.
AIDS Res Hum Retroviruses
 
2016
;
32
:
981
91
.

31

Fan
 
J
,
Yang
 
Y
,
Grimstein
 
M
, et al.   
Whole body PBPK modeling of remdesivir and its metabolites to aid in estimating active metabolite exposure in the lung and liver in patients with organ dysfunction
.
Clin Pharmacol Ther
 
2022
;
111
:
624
34
.

32

Ko
 
JY
,
Danielson
 
ML
,
Town
 
M
, et al.   
Risk factors for coronavirus disease 2019 (COVID-19)-associated hospitalization: COVID-19-associated hospitalization surveillance network and behavioral risk factor surveillance system
.
Clin Infect Dis
 
2021
;
72
:
e695
703
.

33

Devgun
 
JM
,
Zhang
 
R
,
Brent
 
J
, et al.   
Identification of bradycardia following remdesivir administration through the US food and drug administration American College of Medical Toxicology COVID-19 toxic pharmacovigilance project
.
JAMA Netw Open
 
2023
;
6
:
e2255815
.

34

Clerkin
 
KJ
,
Fried
 
JA
,
Raikhelkar
 
J
, et al.   
COVID-19 and cardiovascular disease
.
Circulation
 
2020
;
141
:
1648
55
.

35

Xie
 
Y
,
Xu
 
E
,
Bowe
 
B
,
Al-Aly
 
Z
.
Long-term cardiovascular outcomes of COVID-19
.
Nat Med
 
2022
;
28
:
583
90
.

36

Budi
 
DS
,
Pratama
 
NR
,
Wafa
 
IA
,
Putra
 
M
,
Wardhana
 
MP
,
Wungu
 
CDK
.
Remdesivir for pregnancy: a systematic review of antiviral therapy for COVID-19
.
Heliyon
 
2022
;
8
:
e08835
.

37

Gutierrez
 
R
,
Mendez-Figueroa
 
H
,
Biebighauser
 
JG
,
Bhalwal
 
A
,
Pineles
 
BL
,
Chauhan
 
SP
.
Remdesivir use in pregnancy during the SARS-CoV-2 pandemic
.
J Matern Fetal Neonatal Med
 
2022
;
35
:
9445
51
.

38

Smith
 
DD
,
Pippen
 
JL
,
Adesomo
 
AA
,
Rood
 
KM
,
Landon
 
MB
,
Costantine
 
MM
.
Exclusion of pregnant women from clinical trials during the coronavirus disease 2019 pandemic: a review of international registries
.
Am J Perinatol
 
2020
;
37
:
792
9
.

39

Colbers
 
A
,
Mirochnick
 
M
,
Schalkwijk
 
S
,
Penazzato
 
M
,
Townsend
 
C
,
Burger
 
D
.
Importance of prospective studies in pregnant and breastfeeding women living with human immunodeficiency virus
.
Clin Infect Dis
 
2019
;
69
:
1254
8
.

Author notes

Presented in part: Conference on Retroviruses and Opportunistic Infections, February 12–16 2022.

Potential conflicts of interest. K. M. B. has received consulting fees from ViiV Healthcare. K. B. has received research support from Gilead Sciences, Inc and ViiV Healthcare paid to her institution. D. S. has received research support from Gilead Sciences, Inc, Merck, and ViiV Healthcare paid to his institution. A. A. serves on scientific advisory boards for Gilead Sciences, Inc and ViiV Healthcare; is a site principal investigator for a multisite and investigator-initiated studies with Gilead Sciences, Inc; and is a consultant for Merck. E. V. C. is serving on a data and safety monitoring board for Melinta Pharmaceuticals. D. E. Y. was previously an unpaid technical advisor for the nonprofit Cover the Globe and Maipelo Trust. M. M. has received research support from Gilead Sciences, Inc, Merck, and ViiV Healthcare paid to his institution. R. H. and K. K. are employees of Gilead Sciences, Inc and hold stock in the company. J. D. M. has received research support from Gilead Sciences, Inc paid to his institution. All other authors report no potential conflicts.

All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)

Supplementary data