Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2020 Jun 22;2(3):100159. doi: 10.1016/j.ajogmf.2020.100159

Placental transfer and safety in pregnancy of medications under investigation to treat coronavirus disease 2019

Margaux Louchet a, Jeanne Sibiude a,b,c, Gilles Peytavin d, Olivier Picone a,b,c, Jean-Marc Tréluyer c,e, Laurent Mandelbrot a,b,c,
PMCID: PMC7308040  PMID: 32838264

Abstract

Objective

Treatment of coronavirus disease 2019 is mostly symptomatic, but a wide range of medications are under investigation against severe acute respiratory syndrome coronavirus 2. Although pregnant women are excluded from clinical trials, they will inevitably receive therapies whenever they seem effective in nonpregnant patients and even under compassionate use.

Methods

We conducted a review of the literature on placental transfer and pregnancy safety data of drugs under current investigation for coronavirus disease 2019.

Results

Regarding remdesivir, there are no data in pregnant women. Several other candidates already have safety data in pregnant women, because they are repurposed drugs already used for their established indications. Thus, they may be used in pregnancy, although their safety in the context of coronavirus disease 2019 may differ from conventional use. These include HIV protease inhibitors such as lopinavir/ritonavir that have low placental transfer, interferon that does not cross the placental barrier, and hydroxychloroquine or chloroquine that has high placental transfer. There are also pregnancy safety and placental transfer data for colchicine, steroids, oseltamivir, azithromycin, and some monoclonal antibodies. However, some drugs are strictly prohibited in pregnancy because of known teratogenicity (thalidomide) or fetal toxicities (renin-angiotensin system blockers). Other candidates including tocilizumab, other interleukin 6 inhibitors, umifenovir, and favipiravir have insufficient data on pregnancy outcomes.

Conclusion

In life-threatening cases of coronavirus disease 2019, the potential risks of therapy to the fetus may be more than offset by the benefit of curing the mother. Although preclinical and placental transfer studies are required for a number of potential anti-severe acute respiratory syndrome coronavirus 2 drugs, several medications can already be used in pregnant women.

Key words: coronavirus disease 2019, placenta, pregnancy, severe acute respiratory syndrome coronavirus 2

Introduction

The current coronavirus disease 2019 (COVID-19) pandemic is a global health emergency that affects all populations, including pregnant women.1 , 2 COVID-19 can result in maternal morbidity and mortality from pneumonia and acute respiratory distress syndrome (ARDS),3 similar to severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) infections and influenza.4 , 5 Studies on pregnancy complications are still lacking, although a high preterm birth rate has been reported. This is mostly caused by iatrogenic preterm birth owing to the diagnosis of COVID-196 principally preterm cesarean deliveries.7, 8, 9 Whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly contributes to spontaneous preterm birth or medical complications such as preeclampsia that require iatrogenic preterm birth is less clear. Perinatal transmission may occur but seems rare.6 There is little evidence of in utero or intrapartum exposure, because most amniotic fluid, cord blood, neonatal plasma, and oropharyngeal and placental specimens have been reported to indicate negative results,7, 8, 9 but a case has been reported of a positive result for a reverse transcription polymerase chain reaction (RT-PCR) in a nasopharyngeal swab from a neonate born by elective cesarean delivery and immediately isolated from the mother.10 Postnatal exposure is possible through respiratory and skin contact, but breast milk samples reported negative results in most studies. Anti–SARS-CoV-2 immunoglobulin M was reported in 8 newborns of infected mothers in 2 studies,11 , 12 but these may be false-positive results for immunoglobulins10 because the RT-PCR results were negative. In a Chinese report of 33 neonates born to women with COVID-19, 3 positive PCR test results were reported.13

AJOG MFM at a Glance.

Why was this study conducted?

Although pregnant women can be severely affected by coronavirus disease 2019 (COVID-19), they are generally excluded from clinical trials because of concern about fetal safety. We have data on transplacental transfer of drugs that are currently under investigation to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.

Key findings

The medications considered to treat COVID-19 are repurposed drugs that are in use for other indications, most of which have data on placental transfer and pregnancy safety. Lopinavir and ritonavir, hydroxychloroquine or chloroquine, colchicine, steroids, oseltamivir, azithromycin, and some monoclonal antibodies can be used in pregnant women. Renin-angiotensin system blockers should not be used. Data are lacking for interleukin 6 (IL-6) inhibitors and remdesivir.

What does this add to what is known?

Some of the therapies considered for COVID-19 can be used in pregnant women, but there is a crucial need for research on placental transfer and safety of important investigational drugs including remdesivir.

There is currently no specific antiviral treatment recommended for COVID-19 in general or specifically for pregnant women.3 , 14, 15, 16 Pregnant women remain excluded from all clinical trials to date. Remdesivir, lopinavir/ritonavir, interferon, and hydroxychloroquine or chloroquine are under investigation in several large phase 3 trials, including the Discovery trial of the REACTing consortium17 and the World Health Organization worldwide open-label trial.18 These studies are multicenter, adaptive, randomized, open clinical trials on the safety and efficacy of treatments for hospitalized adults with COVID-19. Data on their use in pregnant women are already available for several of these drugs, including lopinavir/ritonavir studied in women living with HIV,19 , 20 interferon beta (in inflammatory diseases), chloroquine (in malaria prophylaxis), or hydroxychloroquine (in inflammatory diseases). However, for other drugs, there are no data of their use during pregnancy. This is the case with remdesivir, which is generally not considered in pregnant women because of its renal and liver toxicity profile.

Given the potential severity of COVID-19 in pregnant women, there is an urgent need to know which therapies may be used. Placental transfer for each medication is key background information, because the placenta is an active barrier21 that lets variable amounts of drugs cross into the fetal circulation22. The objective of this study was to review the toxicity profiles in pregnancy and the placental transfer of medications under consideration as therapy against SARS-CoV-2.

Materials and methods

We conducted a review of the literature concerning the drugs investigated against SARS-CoV-2 in published or ongoing clinical studies worldwide, with a total of 502 clinical studies registered on the ClinicalTrials.gov website from March 01, 2020 to May 30, 2020. We described the characteristics of drugs based on the French national drug safety agency (ANSM) database (https://www.ansm.sante.fr/) and the pharmacokinetic parameters using the PubChem database and the Human Metabolome Database. We searched pregnancy safety and placental passage data from the PubMed database with keywords pregnancy, placenta, fetus, or newborn for each individual study drug. In vitro efficacy of drugs was defined by their 50% effective concentration (EC50).23, 24, 25 Promising drugs with in vitro antiviral effect against SARS-CoV-2 of EC50<10 μM have been reported.

Results

For the principal medications currently under investigation against SARS-CoV-2, safety data in pregnant women, placental transfer, and neonatal outcome are summarized in the Table .

Table.

Transplacental drug transfer in humans and safety data in women and neonates exposed to medications used for the treatment of patients with coronavirus disease 2019

Transplacental drug transfer
Safety in pregnant women and neonates
Study design Main results Study design Main results
Azithromycin
Sutton et al26 In vivoa n=30 women
Result: detectable in cord blood
Fan et al27 Type of study: retrospective cohort study
Population: exposed to macrolides, n=104 604 children
Control group: exposed to penicillin
Exposed during the first trimester:
  • Major malformation (RR, 1.55; 95% CI, 1.19–2.03)

  • Cardiovascular malformation (RR, 1.62; 95% CI, 1.05–2.51)

Exposed in any trimester:
  • Major malformation (RR, 1.13; 95% CI, 0.94–1.36)

  • Genital malformation (RR, 1.58; 95% CI, 1.14–2.19)

Heikkinen et al28 Ex vivob
Open circuit
n=21 term placentas
Transplacental transfer rated: 2.6%
Lin et al29 Type of study: retrospective cohort study
Population: congenital malformation, heart defect (n=4132) and pyloric stenosis (n=735)
Control group: infants without any malformation
Macrolides (without erythromycin):
  • Cardiac malformation (OR, 0.7; 95% CI, 0.4–1.3)

  • pyloric stenosis (OR, 1.7; 95% CI, 0.6–4.6)

Almaramhy and Al-Zalabani30 Type of study: meta-analysis of cohort studies
Population: exposed to macrolides, n=739 children
Control group: general population or unexposed women
  • Pyloric stenosis (OR,  1.47; 95% CI, 1.03–2.09; I2,  29.3%)

Almaramhyand Al-Zalabani30 Type of study: meta-analysis of case-control studies
Population: exposed to macrolides, n=9000
Control group: general population or unexposed women
  • Pyloric stenosis (OR, 1.02; 95% CI, 0.66–1.58; I2,  51.2%)

Bérard et al31 Type of study: retrospective cohort study
Population: exposed to azithromycin at first trimester, n=120
Control group: children of unexposed women
  • Major malformation (RR, 1.19; 95% CI, 0.98, 1.44)

Cooper et al32 Type of study: retrospective cohort study
Population: exposed to macrolides without erythromycin, n=621
Control group: general population
  • Pyloric stenosis (OR, 2.77; 95% CI, 1.22–6.30)

Chloroquine
Law et al33 In vivoa n=19 women
F/M concentration ratioc:
Chloroquine: 110%
DECQ: 120%
Osadchy et al34 Type of study: analysis of 12 studies
Population: exposed to chloroquine or hydroxychloroquine, n=588
Control group: rheumatic disease unexposed patients
Notably, 5 studies with a total of 251 exposed children reported no clinical visual abnormalities in any case
Silva et al35 Type of study: retrospective cohort study
Population: exposed women to chloroquine for malaria, n=20
Control group: unexposed malarial patient
  • Risk for failing in the hearing screening (RR, 5.64; 95% CI, 1.17–27.3)

Colchicine
Amoura et al36 In vivoa n=1 woman
F/M concentration ratioc: 15%
Diav-Citrin et al37 Type of study: prospective cohort study
Population: Mediterranean fever/Behcet’s exposed women, n=238
Control group: unexposed women
  • Major congenital anomalies: 4.5% vs 3.9% (P=.648)

  • Preterm birth: higher, 15.0% vs 5.9% (P<.001)

  • Birthweight: lower, 3000 g vs 3330 g (P<.001)

Hydroxychloroquine
Costedoat-Chalumeau et al38 In vivoa n=11 women
F/M concentration ratioc: 104% (51–182)
Costedoat-Chalumeau et al39 Type of study: retrospective cohort study
Population: connectivitis exposed patients, n=133
Control group: connectivitis unexposed patients
  • Abortion: 11.3% vs 10% (P=.78)

  • Fetal death: 0.8% vs 2.9% (P=.27)

  • Live birth: 88% vs 84.3% (P=.46)

  • Premature birth: 28% vs 17% (P=.21)

  • Full-term birth 72% vs 67% (P=.21)

Cimaz et al40 Type of study: retrospective cohort study
Population: exposed children, n=6
Outcome: electroretinography results
Results were normal in all cases.
Interferon beta (IFN-β)
Waysbort et al41 Ex vivob
Open circuit
n=4 placentas
No transplacental transfer rated was observed
Hellwig et al42 Type of study: retrospective cohort study
Population: IFN-β exposed women, n=1348
Control group: EUROCAT and MACDP database
Among known pregnancies outcomes:
  • Spontaneous abortion: 11.9% (n=160)

  • Stillbirth: 1.5% (n=20)

Among known live birth outcomes (82%):
  • Congenital birth defects: 1.4% (n=14)

(RR, 0.61; 95% CI, 0.31, 0.94)
Duma et al43 Ex vivob
Open circuit
n=1 placenta
No transplacental transfer rated was observed
Coyle et al44 Type of study: retrospective cohort study
Population: IFN-β exposed women, n=96
Control group: unexposed women
  • Spontaneous abortion: 11.5% (n=11; P=.86)

Among 96 birth outcomes (3 twins):
  • Spontaneous abortion: 11.1% (n=11)

  • Congenital birth defects: 5.8% (n=5; P=.092)

Pons et al45 In vivoa n=2 women
Result: undetectable in cord blood
Interleukin inhibitors
Tocilizumab
Tada et al46 In vivoa n=1 woman
F/M concentration ratioc: 89% in cord blood and 78% in newborn
Nakajima et al47 Type of study: retrospective cohort study
Population: exposed neonates, n=36 neonates
Control group: none
  • Noncongenital anomalies

Hoeltzenbein et al48 Type of study: retrospective cohort study
Population: rheumatoid and juvenile idiopathic arthritis, n=288
Control group: none
Prospective data (n=180):
  • 109 live births (60.6%)

  • 39 spontaneous abortions (21.7%)

  • 1 stillbirth

  • Malformations: 4.5%

  • Preterm birth (31.2%)

Retrospective data (n=108):
  • 55 live births (50.9%)

  • 31 spontaneous abortions (28.7%)

  • Preterm birth (20%)

Interleukin-1 inhibitors
No specific data on transplacental transfer in human Youngstein et al49 Type of study: retrospective cohort study
Population: autoinflammatory disease, n=31 (8 with canakinumab and 23 with anakinra)
Control group: none
  • Miscarriage: n=2/31

  • No serious neonatal infection

  • One case of renal agenesis

  • Preterm birth (20%)

Ivermectin
Nicolas et al50 Type of study: meta-analysis
Population: mostly African women, n=893
Control group: unexposed women
  • Preterm birth: not reported

  • Spontaneous abortion or stillbirth:

  • OR of 1.15 (0.75, 1.78) in observational studies

  • OR of 0.62 (0.18, 2.14) in randomized controlled trials

  • Congenital anomalies

OR of 1.69 (0.83, 3.41) in observational studies
OR of 1.10 (0.07, 17.65) in randomized controlled trials
Leflunomide
Bérard et al51 Type of study: administrative database cohort study
Population: exposed women, n=73
Control group: unexposed women
  • Spontaneous abortion: aOR, 1.09 (0.90, 1.32)

  • Major congenital malformation: aOR, 0.97 (0.81, 1.16)

  • Prematurity: aOR, 4.03 (0.91, 17.85)

  • Low birthweight: aOR, 1.06 (0.90, 1.25)

Lopinavir/ritonavir
Gavard et al52 Ex vivob n=25 placentas
Transplacental transfer rated (albumin concentration):
23.6% (2 g/L) and 3.3% (40 g/L)
Sibiude et al19 Type of study: prospective HIV cohort study
Population: Exposed to LPV/RIT, n=3704
Control group: Exposed to other or no antiretroviral drugs
  • No association between birth defects and lopinavir or ritonavir with a power of >85% for an OR of 1.5

Ivanovic et al53 In vivoa n=11 placentas
Undetectable
Tookey et al20 Type of study: Prospective HIV cohort study
Population: exposed to LPV/RIT, n=4864 pregnancies
Control group: pregnancy as the unit of observation
  • 13% were at <37 weeks’ gestation

  • 2.5% were at <32 weeks’ gestation

  • 15% had birthweight of <2500 g

  • 2.3% had birthweight of <1500 g

  • 2.9% had ≥1 congenital abnormality

Marzolini et al54 In vivoa n=1 placenta
Undetectable
Oseltamivir
Huang et al55 Ex vivob n=20 placentas
Transplacental transfer ratesd of 12.39% (oseltamivir) and 10.17% (its metabolite)
Ehrenstein et al56 Type of study: retrospective cohort study
Population: exposed to oseltamivir, n=1898 pregnancies
Control group: general population
After first-trimester exposure (n=449 pregnancies)
  • Major malformation (OR, 0.94; 95% CI, 0.49–1.83)

  • Congenital heart defect (OR, 1.75; 95% CI, 0.51–5.98)

Chambers et al57 Type of study: prospective cohort study
Population: exposed to oseltamivir, n=112 pregnancies
Control group: general population
  • Major birth defects (RR, 0.84; 95% CI, 0.19, 2.80)

  • Spontaneous abortion: none reported

  • Preterm delivery (RR, 0.65; 95% CI, 0.26, 1.63)

Ribavirin
No specific data on transplacental transfer Sinclair et al58 Type of study: retrospective cohort study
Population: HCV-infected women treated with ribavirin in pregnancy, general population, n=272 pregnant women
Control group: MACDP
Direct exposure (n=133):
  • Live birth: 85/134 (63.4%)

  • Miscarriage: 23/134 (17.2%)

  • Birth defects: 7/85 (8.2%)

  • (95% CI, 3.4–16.2)

  • Indirect exposure (n=139)

  • Live birth: 95/134 (68.3%)

  • Miscarriage: 18/139 (12.9%)

  • Birth defects: 4/95 (4.2%)

(95% CI, 1.2–10.4)
Sildenafil
Russo et al59 Ex vivob
Closed circuit
n=6 placentas
F/M concentration ratioc: 91% to 95%
Dunn et al60 Type of study: meta-analysis
Population: exposed women, n=165
Control group: unexposed
Maternal effects:
  • Headache: 49/107 (46%)

  • Visual trouble: 14/81 (17%)

  • Hypotension: 0/39 (0%)

  • Cesarean delivery: 55/66 (83%)

Neonatal outcome:
  • Nursery admission: 35/52 (67.3%)

  • Stillbirth: 3/69 (4.3%)

  • Neonatal death: 5/129 (4%)

  • Congenital malformations: 0/35 (0%)

Russo et al61 In vivoa Undergoing Ferreira et al62 Type of study: meta-analysis
Population: exposed women with preeclampsia (n=139) or fetal intrauterine growth restriction (n=275)
Control group: unexposed
  • Birthweight: +223 g with high heterogeneity, I=96%

  • No difference in gestational age at birth, I=93%

  • No significant difference in neonatal death

  • No difference in headache, I=69%

aOR, adjusted odds ratio; CI, confidence interval; COVID-19, coronavirus disease 2019; DECQ, desethylchloroquine; EUROCAT, European Registration of Congenital Anomalies and Twins; F/M, fetal-to-maternal; HCV, hepatitis C virus; IFN-β, interferon beta; LPV/RIT, lopinavir/ritonavir; MACDP, Metropolitan Atlanta Congenital Defects Program; OR, odds ratio; RR, risk ratio.

Louchet. Medication for coronavirus disease 2019 in pregnancy. AJOG MFM 2020.

a

Corresponds to blood samples in mothers and neonates.

b

Corresponds to perfused placenta model.

c

Corresponds to comparison of fetal and maternal concentrations, expressed as ratio of concentration or transfer rate (%).

d

Transplacental transfer rate corresponds to ratio of fetal-to-maternal molecule. It includes parameters such as flow rate or compartment volumes (concentration x flow rate/concentration x flow rate). This formula is often used in open cotyledon circuits.200

Azithromycin

Azithromycin is an antibiotic of the macrolide class. It has a molecular mass of 749 g/mol, is sparsely soluble in water, and has 20% protein binding. It has in vitro activity against Zika virus21 , 22 but has not proved significant efficiency against Ebola virus63 nor in decreasing the 90-day mortality in MERS infection in a retrospective cohort of 136 infected patients.64 Nevertheless, azithromycin has an in vitro EC50 of 2.12 μM25 and has been used in patients infected by SARS-CoV-2.65 The combination of azithromycin with hydroxychloroquine seemed to be more effective than hydroxychloroquine alone.66 Currently, several studies are underway to conclude on its effectiveness against COVID-19. The main toxicity in adults is QT interval prolongation and torsade de pointe.

Regarding pregnancy, azithromycin crosses the placental barrier in sheep67 and nonhuman primate68 models, with a maternal-fetal plasma concentration ratio of 3.2 after intravenous administration. Placental transfer in humans was lower in a clinical study26 and in the human ex vivo cotyledon perfusion model.28

Regarding safety, most studies found no increase in major congenital malformations in the event of exposure during the first trimester (risk ratio [RR], 1.19; 95% confidence interval [CI], 0.98–1.44; 120 cases exposed)31 and no cardiac malformation (RR, 0.7; 95% CI, 0.4–1.3) if any macrolide is used during the first trimester.29 An association between prenatal macrolide exposure and pyloric stenosis has been reported, but remains controversial.29 , 30 , 32 A recent English cohort study reported an increased risk for fetal malformations when using macrolides during the first trimester of pregnancy (compared with the use of penicillin: RR, 1.55; 95% CI, 1.19–2.03) and in particular cardiovascular anomalies (RR, 1.62; 95% CI, 1.05–2.51) and genital anomalies, regardless of the trimester of exposure (RR, 1.58; 95% CI, 1.14–2.19).27

Chloroquine and hydroxychloroquine

Chloroquine is an antimalarial drug derived from 4-aminoquinoline. Its molecular mass is 319.9 g/mol; it is lipophilic and has a protein binding of 50%. It has been reported to have antiviral properties in vitro. Chloroquine is able to inhibit influenza A in vitro69 but previous randomized trials in humans with influenza reported no efficacy vs placebo.70 Chloroquine has in vitro activity against coronaviruses by interfering with the glycosylation of the SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2) and reduces viral replication71, 72, 73 with an EC50 of 9.12 μM.24 It inhibits the replication of SARS-CoV-1 at the plasma concentrations conventionally used for malaria treatment.74 Recently, chloroquine has proved in vitro efficacy against SARS-CoV-2, blocking infection75 and virus replication with an EC50 of 9.12 μM.24 Chloroquine is under investigation in several clinical studies for the treatment of COVID-19.76

Chloroquine cardiac toxicities include restrictive cardiomyopathy reported in several case reports77, 78, 79, 80 and, in case of an overdose, the risk for hypotension and cardiac conduction disorder (prolongation of the QT space and enlargement of the QRS).

Concerning pregnant women, pharmacokinetic studies report an increase in clearance of chloroquine and its active metabolite desethylchloroquine (DECQ), especially in the second and third trimesters.81 , 82 Coadministration of azithromycin modestly decreases exposure to DECQ without modifying exposure to chloroquine.81 Its use in pregnant women could be responsible for ototoxicity reported in a small study (RR, 5.64; 95% CI, 1.17–27.3).35 Chloroquine crosses the placenta33 , 83, 84, 85, 86 with a mean ratio of cord-to-maternal concentrations of 1.1 for chloroquine and 1.2 for DECQ. With regard to pregnancy outcomes including miscarriages, no increased risk has been reported.87 In humans, studies have not reported any teratogenic or fetotoxic effect in the event after in utero exposure; in particular, no risk for ocular damage in newborns was found in a meta-analysis of 588 infants.34

Hydroxychloroquine is also an antimalarial drug derived from 4-aminoquinoline with an anti-inflammatory action. It has a molecular mass of 335.9 g/mol, is lipophilic, and has a protein binding of 45%. It has indicated antiviral activity against SARS-CoV-2 in small in vitro studies88 , 89 and some studies have reported that hydroxychloroquine reduces viral excretion.66 , 90 Compassionate use has been authorized in patients with COVID-19,91 and a large number of observational studies that have been performed worldwide published conflicting conclusions on its efficacy and safety on COVID-19,92, 93, 94, 95, 96, 97, 98, 99, 100 and randomized clinical trials are currently underway (53 studies with randomized statement criteria are available in ClinicalTrials.gov website, including NCT04329923, NCT04307693, ChiCTR2000029559, ChiCTR2000029898-99, and ChiCTR2000029992).

Hydroxychloroquine is recommended for patients with rheumatoid arthritis and lupus. It obtained marketing authorization in these indications in 1997.101 Hydroxychloroquine toxicities are retinopathy in case of long-term treatment at high doses (prevalence from 2%–8%),102 , 103 heart conduction disorders,104 and more rarely ventricular tachyarrhythmia or QT prolongation.

Hydroxychloroquine has been reported to be safe for use in pregnant women at doses of 200 mg once or twice a day. It crosses the placenta by passive diffusion with a strong correlation between maternal and fetal concentrations and a fetal-to-maternal concentrations ratio of 1.04 (range, 0.51–1.82 ).38 Pregnancy outcomes in the event of exposure are similar to those of untreated women with the same conditions (number of miscarriages and rate of malformation in particular).39 , 105 , 106 There has been no reported increase in visual disturbances in newborns of exposed women.40

Lopinavir/ritonavir

The fixed combination lopinavir/ritonavir has been widely used for the treatment of adults living with HIV. Lopinavir has a molecular mass of 1349.7 g/mol, is practically insoluble in water, and has a protein-binding rate of 98% to 99%. Lopinavir is a protease inhibitor prescribed in combination with ritonavir, an inhibitor of the isoform CYP3A4 of cytochrome p450 which is used as a booster to obtain therapeutic plasma concentrations. It has in vitro and in vivo efficiency against SARS-CoV-1 and MERS infections.107 , 108 It is considered as an option for COVID-19 treatment in several trials, although the first randomized, open-label, controlled study to be published reported no significant benefits of lopinavir/ritonavir.109

The tolerability of lopinavir/ritonavir is poorer than of other HIV protease inhibitors, especially regarding digestive disorders and lipid abnormalities.85 In the randomized controlled trial from China in adults with severe COVID-19 infection,109 treatment (400 mg/100 mg twice a day) had to be stopped early for 13.8% (n=13) of patients because of adverse events (primarily gastrointestinal adverse events). This incidence of severe adverse events was higher than what was reported in HIV infection at the same dose. Lopinavir/ritonavir exposure increases plasma concentrations of CYP3A4-metabolized drug taken concurrently.

In pregnant women living with HIV, lopinavir/ritonavir was for several years the most widely used antiretroviral therapy, with a nucleoside reverse transcriptase backbone,86 and used as monotherapy (at 400 mg/100 mg, twice daily).110 During pregnancy, the clearance of the free form of lopinavir/ritonavir has not changed and experts do not recommend increasing the dosage of the drug (generally 400 mg of lopinavir/100 mg of ritonavir twice a day).111 Lopinavir/ritonavir crosses the placenta but poorly. Fetal transfer rates with perfused cotyledon model are around 20% to 25%52 and the level in cord blood is undetectable in studies with cord-to-mother ratios.53 , 54 This is caused by strong protein binding for protease inhibitors and to the efflux mechanism of the placental membrane transporters and in particular to P-glycoprotein.112, 113, 114

Pregnancy outcomes were reported to be similar to those of control women living with HIV.19 , 20 There was no increase in the rate of congenital malformations after first-trimester exposure. A large randomized trial has reported a higher rate of preterm births in the groups treated with lopinavir/ritonavir-based antiretroviral treatment (ART) than monotherapy of zidovudine.115 This has already been described in a randomized trial comparing lopinavir/ritonavir-based ART with a combination of 3 nucleoside reverse transcriptase inhibitors.116

In addition, several cases of lopinavir/ritonavir toxicity have been described in treated preterm infants, in particular signs suggesting adrenal insufficiency with hyponatremia and hyperkalemia.117 These undesirable effects led to an alert by the Food and Drug Administration (FDA) in 2011 to contraindicate this drug in premature babies.85

Remdesivir

Remdesivir (GS-5734) is a prodrug of a nucleoside analog antiviral. It has a molecular mass of 602.9 g/mol with a protein-binding rate of 85% to 92% in animal studies. It is metabolized by the liver and has a renal and biliary excretion. Its water solubility is not yet available on the PubChem website. It is metabolized into its active form GS-441524. It competes with adenosine triphosphate and inhibits the viral RNA polymerase.118 Remdesivir was initially developed against Ebola virus, with efficacy in a clinical phase 3 trial.119 It has also been studied to treat MERS-CoV and SARS-CoV-1 infections.120 Its efficacy against SARS-CoV-2 has been proven in vitro75 , 121 with EC50 of 11.41 or 6.25 μM associated with emetine23 , 24 and in vivo in animals.122 Its mechanisms of action (shortening of the DNA chain or modification of excision activity) remain to be explained in detail.120 , 123 A number of clinical trials are underway to determine the efficacy and safety of remdesivir to treat COVID-19 (NCT04252664, NCT04257656, NCT04280705, NCT04292730, and NCT04292899). Notably, 2 double-blinded randomized clinical trials have been published using an intravenous loading dose of 200 mg followed by 100 mg daily for 10 days.124 , 125 Both indicated a reduction of time to clinical improvement.124 Similar results have been reported in its compassionate use for severely ill patients.126

To date, tolerance data in humans are sparse. There is a risk for renal dysfunction127 , 128 and elevated liver enzymes, a possible effect on mitochondrial functions,127 and a case of cardiac arrest in a clinical trial on Ebola infection.119 Thus, investigators recommended biological monitoring of liver and kidney functions. Clinical trials on COVID-19 disease also report adverse effects with an incidence ranging from 21% to 66%.124, 125, 126

In a clinical study on the Ebola disease, 6 pregnant women who were treated with remdesivir had positive pregnancy tests,119 but these cases were not described. We found no study addressing the safety of remdesivir in pregnant women or its placental transfer. Considering its small molecular weight and its high protein-binding rate, we may expect that remdesivir crosses the placenta.

Other antiviral drugs

Several antiviral drugs commonly used in clinical practice have been proposed,129 although some in vitro studies of antiviral effects on SARS-CoV-2 replication failed to indicate an effect of oseltamivir, favipiravir, ribavirin or darunavir.23 , 130

Oseltamivir is widely recommended to treat influenza in children or adults who are considered at risk for severe manifestations, including pregnant women. According to the ex vivo human placental perfusion, oseltamivir and its metabolite oseltamivir carboxylate cross poorly the placenta with transfer rates of 12.39% and 10.17%, respectively.55 Retrospective studies with large population do not provide evidence of risk for any major congenital malformation or heart defect.57 No evidence of increased risk for preterm birth nor small for gestational age infants was reported in another prospective study.57 , 131 We did not find any information about pregnancy and zanamivir, which is another neuraminidase inhibitor.

Favipiravir is an antiviral drug approved in Japan and China for influenza which inhibits the RNA polymerase enzyme.132 , 133 It has indicated efficacy against Ebola virus in animals and humans.134 , 135 A potential synergistic benefit of favipiravir associated with oseltamivir has been described in influenza.136 , 137 Favipiravir is under investigation for treating SARS-CoV-2 in several clinical trials (NCT04310228, NCT04358549). We found no information about the use of favipiravir in pregnant women but caution is needed because of a warning by the Japanese drug agency on a teratogenic risk in preclinical animal studies.132 , 138

The combination of interferon alpha and ribavirin (which is an antiviral mainly used to treat hepatitis C) is also under investigation in COVID-19, even though it has not been proven to reduce mortality for the treatment of MERS-CoV.139 , 140 In pregnant women, ribavirin is usually contraindicated because of its possible teratogenic role in the event of direct or indirect exposure (preconception or in the event of father’s treatment). There are some reassuring data,141 but the number of exposures is too small to conclude on safety.

Some HIV protease inhibitors are under consideration (NCT04252274), in addition to lopinavir discussed previously. Darunavir with a ritonavir boost has been the most widely used therapy for HIV in pregnant women in Europe and America for nearly a decade, and there are data from prospective cohort studies. There is no increase in the risk for congenital malformations. However, the use of boosted protease inhibitors has been associated with an increased risk for preterm delivery, although it is not clear whether the effect is related to any specific drug or other confounding factors.142 Nelfinavir is a selective inhibitor of HIV protease which has been reported to inhibit SARS-CoV replication in vitro.143 The placental transfer of nelfinavir is low.54 There is no increase in the risk for congenital malformations.

Arbidol hydrochloride (umifenovir) is an immunomodulator and a broad-spectrum antiviral drug. Arbidol is able to block viral fusion against influenza viruses and has been reported to have antiviral activity in vitro against SARS-CoV.144 It has been reported to increase the effect of lopinavir/ritonavir treatment on SARS-CoV-2 excretion at 7 and 14 days in a retrospective cohort study (n=33 patients),145 but clinical outcomes have not yet been assessed.146 Comparison of arbidol with lopinavir/ritonavir or oseltamivir is under investigation in a randomized controlled phase 4 study (NCT04255017). A study reports its use, in association with other antiviral drugs, in 7 pregnant women with SARS-CoV-2 infection.147 We found no information on safety of umifenovir in pregnancy.

Clevudine (NCT04347915) is used in hepatitis B infection in Asian countries. We found no information on its safety profile in pregnancy or its placental transfer.

Other antiinfective drugs

Ivermectin, usually used to treat parasite infections, is currently under investigation in COVID-19 clinical trials (NCT04390022 and NCT04381884). Transplacental transfer has been proven in sheep with limited fetal exposure.148 Data on safety for pregnant women and neonates are sparse, and a recent meta-analysis has concluded to insufficient evidence for its safety profile in pregnancy.50 Nitazoxanide is another antiparasitic drug under consideration (NCT04343989) for which we found no specific data on specific use in pregnant women.

The FDA-approved antihelminthic drug niclosamide has indicated in vitro antiviral efficacy against SARS-CoV-2.24 As mentioned previously, the amebicide emetine inhibits SARS-CoV-2 replication23 , 24 and has been studied in association with remdesivir.

Interferon

Interferons beta and lambda are both studied as COVID-19 therapy but most information concerns interferon beta. Interferon beta is a cytokine with antiviral, antiproliferative, and immunomodulatory activities. It is commonly used to treat multiple sclerosis. It is a glycoprotein that works after it has attached to the cell membrane by triggering a cascade of intracellular signaling with, in particular, the induction of enzymes that could inhibit viral replication in infected cells. Interferon beta has been proposed as immune-modulatory treatment to control the massive inflammation resulting in lung injury in SARS-CoV-2 infection.

There is a long experience of safety and tolerability of interferon in nonpregnant adults and the most frequently reported adverse events are injection site reactions, asthenia, flu-like symptoms, headache, and lymphopenia.149

Pregnancy outcomes in women treated with interferon beta have been documented in prospective and retrospective pharmacovigilance studies on 1348 and 99,948 pregnant women, respectively.42 , 44 , 150 The prescribed subcutaneous doses ranged from 30 μg/0.5 mL per week to 30 μg/0.5 mL 3 times a week. These studies did not find an increased risk for miscarriage or congenital malformation associated with treatment with interferon beta. Other perinatal outcomes were not described.

We did not find any study on the transplacental passage of interferon beta. However, interferons have high molecular weights and it has been reported that interferon alpha does not cross the placenta in humans, according to data from the ex vivo cotyledon perfusion model41 , 43 and neonatal cord blood samples.45 By analogy, we can assume that interferon beta does not cross the placental barrier.

Convalescent plasma

Transfusion of immunoglobulins from patients who have recovered from the disease is a passive immunotherapy. Convalescent plasma transfusion has been previously described as a therapeutic option in the management of Ebola and previous coronavirus infections.151 , 152 It was not associated with a significant improvement in survival in Ebola patients153 but was useful in the treatment of SARS-CoV infection with a higher discharge rate (control group with usual care represented with 418 patients previously treated in the same center), especially if given at the early phase of the disease (n=80).154 There have been case reports on the use of convalescent plasma transfusion in association with supportive care and antiviral drugs.145 , 146 Recently, convalescent plasma has been approved by the FDA to treat critically ill patients with COVID-1944 , 149 , 150 and clinical trials are currently being conducted (NCT0432142, NCT04321421 [ongoing], and NCT04333355 [ongoing]). Previous studies included pregnant women with SARS-CoV-2 (n=1)155 and Ebola infection (n=8)153 but the authors recognized that pregnancy was incompletely recorded in the control group. Concerning transplacental transfer of immunoglobulins, it is well known that transfer of maternal immunoglobulin G (IgG) to the fetus plays an important role in preventing neonatal infections. Antibodies are transported across the placenta with mediation of the neonatal Fc receptor156 and transfer increases with gestational age, mostly in the third trimester.

Inactivated vaccines are also currently under investigation (NCT04352608). Safety in pregnant women has obviously not been assessed.

Immune modulators

The leading cause of mortality from COVID-19 is respiratory failure from severe acute respiratory syndrome. Accumulating evidence suggests that patients with severe COVID-19 have a cytokine storm, which is a hyperinflammatory syndrome characterized by a fulminant and fatal hypercytokinemia and multiorgan failure and previously described with SARS-CoV infection.157 Other approaches to anti-inflammatory therapy include cytokine blockers, steroids, and other potent anti-inflammatory agents.

Interleukin 6 (IL-6) is a proinflammatory cytokine that participates in the cytokine storm which characterizes ARDS in COVID-19. Therefore, the following anti–IL-6 therapies are being considered in COVID-19 pneumonia: tocilizumab (NCT04317092, NCT04331795, NCT04332913); and other antiinterleukin molecules such as clazakizumab (NCT04343989), levilimab (NCT04397562), olokizumab (NCT04380519), sarilumab (NCT04315298, NCT04321993, NCT04357808), and siltuximab (NCT04330638, NCT04329650). Tocilizumab is a humanized monoclonal antibody that binds to IL-6 receptors and inhibits the signal transmitted by the cytokine. Anti–IL-6 therapy is usually used as an immunosuppressant in rheumatoid arthritis and its use requires monitoring of liver function. Tocilizumab crosses the placental barrier and is found in cord blood assays (89% of maternal plasma dosage), plasma assays in newborns (78%), and breast milk (11%)46 with a peak in breast milk reached on the third day after injection.158 There does not seem to be an increased risk for congenital anomalies47 , 48 but the interpretation of the data is difficult because there is often coexposure with methotrexate. In 2016, at the European Congress of Rheumatology, specialists recommended against tocilizumab in pregnancy judging the data insufficient in terms of fetal safety.159 Even fewer data are available regarding sarilumab/siltuximab. Similar to other IgG antibodies of the IgG1 subclass, they cross the placenta. Immune response may be impaired in infants with in utero exposure. A registry has been established to monitor outcomes after exposure to anti–IL-6 monoclonal antibodies during pregnancy. Other antiinterleukin inhibitors (anti–IL-1) that are also under investigation are canakinumab (NCT04362813) and anakinra (NCT04362813). A retrospective study on 31 exposed women has reported global sparse reassuring data with no neonatal infection but renal agenesis in 1 fetus after anakinra exposure.49

The sphingosine-1-phosphate receptor regulator fingolimod (FTY720) is an immune modulator which prevents lymphocytic contribution to autoimmune reaction. It also has been proposed in COVID-19 therapy (NCT04280588). Fingolimod has been widely used in severe multiple sclerosis. It is generally well tolerated and has a long-term safety profile (including infection or malignancy rates)160 , 161 but serious cardiac adverse effects have been reported in patients at risk for heart failure. Data on the safety in pregnant women do not indicate any increase in major malformation rates among live births in comparison with the general population and the multiple sclerosis population (n=91 pregnancies and 75 live births).162 We found no data on placental transfer of fingolimod.

Baricitinib, a Janus kinase (JAK) and adaptor-associated kinase inhibitor, was found capable to reduce or interrupt the passage of the virus into target cells (endocytosis into pulmonary cell) and to inhibit the JAK1- and JAK2-mediated cytokine release, constituting the rationale to perform a trial on patients with mild to moderate COVID-19.2 , 163 The drug was licensed for the treatment of rheumatoid arthritis at the daily dose of 4 mg orally, with excellent results in terms of clinical response and a good safety profile.163 Because baricitinib does not interact with antivirals owing to its prevalent renal elimination, it may be used in combination with antiviral therapy. As a biological and small molecule, baricitinib is contraindicated in pregnancy, awaiting larger studies to assess its safety.164 , 165 Another anti-JAK molecule is also tested in current studies, that is, ruxolitinib (NCT04377620, NCT04334044, NCT04334044).

Infliximab is also under consideration for severe COVID-19 pneumonia (NCT04333420). There are data from retrospective cohorts on the safety of infliximab in pregnant women treated for Crohn’s disease.166 It crosses the placenta in the second and third trimesters and has been detected in newborn samples 6 months after delivery.167 , 168

Leflunomide is an immune suppressor used in rheumatoid arthritis that is also under investigation for COVID-19 (NCT04361214). It is teratogenic in an animal model and thus is to be avoided if possible, although a small retrospective study36 reported no association with major congenital malformation after 5 cases of first-trimester exposure or adverse pregnancy outcomes after 15-second and third-trimester exposures. We found no data on placental transfer of leflunomide.

Additional immune therapies under investigation to treat the cytokine storm in COVID-19 include lenzilumab, which is a humanized monoclonal antibody that blocks the granulocyte-macrophage colony–stimulating factor (NCT04351152), and the human IgG1 monoclonal antibody gimsilumab (NCT04351243).

Colchicine is a powerful anti-inflammatory drug. It is under investigation in a simple, pragmatic randomized controlled trial for its immunomodulatory and anti-inflammatory effects, which may reduce the cytokine storm and inflammatory cascade activation in severe forms of COVID-19 (NCT04328480, NCT04326790). To date, it has not been proven to be effective as a treatment or prevention of COVID-19 disease.169, 170, 171 Colchicine has been approved for the treatment of various inflammatory diseases such as gout, familial Mediterranean fever, Behcet’s disease, and some dermatologic indications (chronic urticaria, cutaneous vasculitis, and psoriasis). Regarding pregnant women, colchicine is not contraindicated during pregnancy.159 Colchicine crosses the placenta.36 A meta-analysis reported no increase in the incidence of miscarriage or major fetal malformations in exposed women compared with nonexposed women with familial Mediterranean fever (4 studies and 554 pregnancies with colchicine exposure).172 However, in 1 retrospective study, there was an increased risk for preterm deliveries at ≤36 weeks’ gestation (15% [n=32/214] vs 5.9% [n=51/867]; P<.01) and a lower median birthweight in singleton full-term children (3090 g vs 3315 g; P<.01).37

The use of steroids is still controversial for the treatment of SARS-CoV-2 infection. Methylprednisolone and dexamethasone are being studied in prospective randomized controlled trials (NCT04273321, NCT04325061, and NCT04329650). In addition, the glucocorticosteroid ciclesonide has indicated in vitro antiviral efficacy against SARS-CoV-2.24 In pregnant women, fluorinated glucocorticosteroids (usually betamethasone with 2 doses of 12 mg) are recommended in case of a high risk for preterm delivery to accelerate fetal lung maturation. Their placental transfer is high.173 In contrast, prednisone is inactivated by placental 11β-hydroxysteroid dehydrogenase.174 , 175 Betamethasone and dexamethasone increase blood glucose level but are considered safe for pregnant women. Their clinical benefits have been proven in numerous studies including a meta-analysis of 18 trials and more than 3700 babies.176 Steroids reduce mortality (odds ratio [OR], 0.60; 95% CI, 0.48–0.75) and respiratory distress syndrome (OR, 0.53; 95% CI, 0.44–0.63), and they may also reduce the risk for severe intraventricular hemorrhage.

Medications modifying the renin-angiotensin-aldosterone system

Drugs that regulate the renin-angiotensin-aldosterone system have important potential to treat COVID-19.177 ACE was identified as a SARS-CoV receptor in 2004.178 Thus, ACE inhibitors are considered as a therapeutic approach to limit the endocytosis of the viral complex179 and thereby the number of cells infected with SARS-CoV-2. Losartan, an angiotensin II receptor blocker, is under investigation in patients with COVID-19 (NCT04318418). Another angiotensin receptor blocker, telmisartan, is also being studied (NCT04355936). In pregnancy, drugs that block the renin-angiotensin system can damage the fetal kidneys, leading to renal failure with oligohydramnios, deformations, and lung hypoplasia.180 , 181 These drugs modifying the renin-angiotensin cascade could also affect placental functions leading to growth retardation.182

Other drugs with vascular impact

Sildenafil, a phosphodiesterase type 5 inhibitor, is a vasodilator used to treat erectile dysfunction and pulmonary arterial hypertension. It is under investigation in patients with COVID-19 (NCT04304313) because pulmonary hypertension is a complication of ARDS. In pregnant women, as in nonpregnant patients, the adverse effects are headache and visual disturbance.183 It crosses the placenta at a high rate in ex vivo model,184 and thus, it is considered in antenatal care to prevent pulmonary hypertension in case of congenital diaphragmatic hernia.185 Sildenafil has also been studied for the prevention of fetal growth restriction in case of maternal vascular disease such as preeclampsia, and a meta-analysis has reported a significant increase in fetal weight at birth.186 No increases of neonatal death or congenital anomalies have been described in another meta-analysis.183 However, the Sildenafil Therapy in Dismal Prognosis Early-Onset Intrauterine Growth Restriction group which was gathered in 2011 to explore effects of sildenafil on fetal growth restriction recommended against the prescription of sildenafil.187 , 188

Bevacizumab (NCT04305106) is an anti–vascular endothelial growth factor antibody. It has been used in cases of ovarian cancer in pregnancy with no maternal and fetal adverse effects189 but it is still recommend to avoid its use.190 It has also been used during pregnancy to treat intravitreal hemorrhage with local injection191 , 192 with no pregnancy-related complications. We found no information on the placental transfer of bevacizumab.

Anticancer agents

Thalidomide is under consideration for COVID-19 (NCT04273529 and NCT04273581), but it is definitively contraindicated in pregnancy because of its proven teratogenicity.193 , 194

Another anticancer agent, plitidepsin, is also under investigation for COVID-19 (NCT04382066). There is no information on pregnancy use, but in view of its high molecular mass (1110 g/mol), we may expect that it would not cross the placenta.

The topoisomerase II blocker etoposide, which is proposed for a clinical study on COVID-19 (NCT04356690), has been reported in few case reports to treat gestational trophoblastic disease or malignant teratomas.195, 196, 197 Data on exposure during completed pregnancy are sparse198 and evidence of adverse effect is difficult to conclude because women received combinations of drugs. We found no data on placental transfer of etoposide.

Other approaches

A large variety of other drugs are also currently under consideration against SARS-CoV-2. Dapagliflozin is an antidiabetic medication of the sodium-glucose cotransporter 2 inhibitor class which is under consideration to treat COVID-19–related respiratory failure, sepsis, and multiorgan failure in a clinical trial (NCT04350593). We found no specific data on its use during pregnancy.

Another therapeutic approach under investigation is the use of fetal-derived cells to reduce inflammation and fibrosis in COVID-19–related lung infection. The study registered as NCT04319731 is a pilot study of nebulized human amniotic fluid. Umbilical cord–derived mesenchymal stromal cells are used in several studies (NCT04333368, NCT04315987, NCT04288102). There is no data available on their safety in pregnancy.

Discussion

Among the medications under investigation to treat COVID-19, some already have sufficient safety data to allow their use in pregnant women. These include the HIV protease inhibitors such as lopinavir/ritonavir, interferon, and hydroxychloroquine. In addition, there are reassuring safety data for use of colchicine, steroids, oseltamivir, azithromycin, and to some extent monoclonal antibodies such as infliximab and possibly tocilizumab. Some drugs are strictly prohibited in pregnancy because of known teratogenicity (thalidomide) or fetal toxicities (renin-angiotensin system blockers). Other candidates have insufficient pregnancy data, including IL-6 inhibitors, umifenovir, and favipiravir. Importantly, remdesivir remains an investigational drug with restricted use in the absence of preclinical data concerning pregnancy in view of its toxicity profile (hypotension during injection, renal and liver dysfunction). The range of medications under consideration to treat COVID-19 is rapidly evolving.

An unusual aspect of clinical trials on COVID-19 is the fact that most of the drugs under investigation are repurposed and thus are already used in pregnant women for their established indications. Because pregnant women are usually excluded from clinical trials, pregnancy follow-up data were obtained from off-label use. Although some follow-up data are obtained from structured cohort studies, most studies are from retrospective registers with limited endpoints and delayed and disparate collection of clinical data. Importantly, the doses used may differ and the diseases treated differ widely, so that drug toxicities may be quite different. This is of particular concern in patients with severe organ or multiorgan failure.

The use of medications in pregnancy must take into account a number of important issues. First, there is potential for increased toxicities to the patient herself and pharmacokinetic and pharmacodynamic differences with nonpregnant individuals. Physiological changes include increased volume of distribution, lower albumin concentrations, and hormone-induced susceptibility to hepatic toxicities.199 In addition, there may be an effect on placental functions and potential adverse pregnancy outcomes, such as abortion, fetal growth restriction, and preterm delivery. The most important are the potential risks for the fetus, such as teratogenicity, fetal toxicities, and long-term effect on the child’s health.

Clinical trials usually exclude pregnant women and newborns even when benefits are expected for this population type. This precautionary principle has been denounced as unjust.183 Pregnant women, according to the principle of autonomy, should be able to take responsibility and provide consent for participation in clinical trials,184 even for drugs with potential teratogenic risk.185 Excluding pregnant women from clinical trials usually prevents us from obtaining reliable safety data from the use of drugs during pregnancy. This makes it very difficult to take care of pregnant patients with the prescription of drugs without proof of efficacy or tolerance established during pregnancy. Thus, it seems that the benefits of including pregnant patients in therapeutic trials would outweigh the risks.186

Knowledge about placental transfer should be available early enough in the process of drug evaluation to be able to move on to use in pregnant women without needless delays. Most medications cross the placenta, but the amount of placental transfer differs. Passive diffusion is the major mechanism of transplacental transfer and depends on pharmacokinetic parameters such as small molecular weight, high water solubility, polarity, and important protein-binding rate (>90%). The other important mechanism is active transport which depends on import or export protein transporters.201 Three types of studies have been developed to obtain information on fetal drug exposure. Placental transfer may be estimated by the concentration ratios between the umbilical cord blood and maternal blood at birth. However, these studies are based on a small number of patients. Preclinical studies on placental transfer are performed either in animals (but their placentas differ from the human hemochorial placenta) or in experimental models,187 , 188 including the ex vivo human cotyledon perfusion model.2, 3, 4 Physiologically based pharmacokinetic models have also been developed to predict fetal exposure to drugs.202

Because the available evidence suggests that there is little risk for vertical transmission of SARS-CoV-2, there is no need for antiviral therapies to cross the placenta. There is also no benefit to be expected from fetal exposure to immunomodulatory therapies. Thus, the optimal approaches to treating pregnant women with COVID-19 would theoretically be those with minimal fetal effect or minimal placental transfer.

The management of pregnant women with COVID-19 is essentially symptomatic. Therapies must be discussed, based on rapidly evolving data on the efficacy and safety in nonpregnant adults, also taking into account obstetrical and fetal consequences of such treatment and the consequences of lack of therapy. In severe cases, therapy should not be withheld solely because of pregnancy and should be discussed between obstetricians, intensive care and infectious disease specialists, and the patient and her partner.

Footnotes

This paper is part of a supplement that represents a collection of COVID-related articles selected for publication by the editors of AJOG MFM without additional financial support.

The authors report no conflict of interest.

References

  • 1.World Health Organization Déclaration sur la deuxième réunion du Comité d’urgence du Règlement sanitaire international 2005 concernant la flambée de nouveau coronavirus 2019 (2019-nCoV). 2020. https://www.who.int/fr/news-room/detail/30-01-2020-statement-on-the-second-meeting-of-the-international-health-regulations-(2005)-emergency-committee-regarding-the-outbreak-of-novel-coronavirus-(2019-ncov) Available at: Accessed April 1, 2020.
  • 2.Dashraath P., Wong J.L.J., Lim M.X.K., et al. Coronavirus disease 2019 (COVID-19) pandemic and pregnancy. Am J Obstet Gynecol. 2020;222:521–531. doi: 10.1016/j.ajog.2020.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rasmussen S.A., Smulian J.C., Lednicky J.A., Wen T.S., Jamieson D.J. Coronavirus disease 2019 (COVID-19) and pregnancy: what obstetricians need to know. Am J Obstet Gynecol. 2020;222:415–426. doi: 10.1016/j.ajog.2020.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Schwartz D.A., Graham A.L. Potential maternal and infant outcomes from (Wuhan) coronavirus 2019-nCoV infecting pregnant women: lessons from SARS, MERS, and other human coronavirus infections. Viruses. 2020;12:194. doi: 10.3390/v12020194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Siston A.M., Rasmussen S.A., Honein M.A., et al. Pandemic 2009 influenza A(H1N1) virus illness among pregnant women in the United States. JAMA. 2010;303:1517–1525. doi: 10.1001/jama.2010.479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Egloff C., Vauloup-Fellous C., Picone O., Mandelbrot L., Roques P. Evidence and possible mechanisms of rare maternal-fetal transmission of SARS-CoV-2. J Clin Virol. 2020;128:104447. doi: 10.1016/j.jcv.2020.104447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chen H., Guo J., Wang C., et al. Clinical characteristics and intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a retrospective review of medical records. Lancet. 2020;395:809–815. doi: 10.1016/S0140-6736(20)30360-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schwartz D.A. An analysis of 38 pregnant women with COVID-19, their newborn infants, and maternal-fetal transmission of SARS-CoV-2: maternal coronavirus infections and pregnancy outcomes. Arch Pathol Lab Med. 2020 doi: 10.5858/arpa.2020-0901-SA. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 9.Chen S., Huang B., Luo D.J., et al. [Pregnancy with new coronavirus infection: clinical characteristics and placental pathological analysis of three cases] Zhonghua Bing Li Xue Za Zhi. 2020;49:418–423. doi: 10.3760/cma.j.cn112151-20200225-00138. [DOI] [PubMed] [Google Scholar]
  • 10.Kimberlin D.W., Stagno S. Can SARS-CoV-2 infection be acquired in utero?: more definitive evidence is needed. JAMA. 2020 doi: 10.1001/jama.2020.4868. [Epub head of print] [DOI] [PubMed] [Google Scholar]
  • 11.Zeng H., Xu C., Fan J., et al. Antibodies in infants born to mothers with COVID-19 pneumonia. JAMA. 2020;323:1848–1849. doi: 10.1001/jama.2020.4861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dong L., Tian J., He S., et al. Possible vertical transmission of SARS-CoV-2 from an infected mother to her newborn. JAMA. 2020;323:1846–1848. doi: 10.1001/jama.2020.4621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zeng L., Xia S., Yuan W., et al. Neonatal early-onset infection with SARS-CoV-2 in 33 neonates born to mothers with COVID-19 in Wuhan, China. JAMA Pediatr. 2020 doi: 10.1001/jamapediatrics.2020.0878. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Peyronnet V., Sibiude J., Deruelle P., et al. [SARS-CoV-2 infection during pregnancy. Information and proposal of management care. CNGOF] Gynecol Obstet Fertil Senol. 2020;48:436–443. doi: 10.1016/j.gofs.2020.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chen S., Liao E., Cao D., Gao Y., Sun G., Shao Y. Clinical analysis of pregnant women with 2019 novel coronavirus pneumonia. J Med Virol. 2020 doi: 10.1002/jmv.25789. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sanders J.M., Monogue M.L., Jodlowski T.Z., Cutrell J.B. Pharmacologic treatments for coronavirus disease 2019 (COVID-19): a review. JAMA. 2020 doi: 10.1001/jama.2020.6019. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 17.Inserm. Covid-19: démarrage de l’essai clinique discovery. 2020. https://www.inserm.fr/actualites-et-evenements/actualites/covid-19-demarrage-essai-clinique-discovery Available at: Accessed April 1, 2020.
  • 18.Henao Restrepo A.M., Moorthy V., Preziosi M.-P. Public health emergency SOLIDARITY trial of treatments for COVID-19 infection in hospitalized patients. ISRCTN. http://www.isrctn.com/ISRCTN83971151 Available at:
  • 19.Sibiude J., Mandelbrot L., Blanche S., et al. Association between prenatal exposure to antiretroviral therapy and birth defects: an analysis of the French perinatal cohort study (ANRS CO1/CO11) PLoS Med. 2014;11 doi: 10.1371/journal.pmed.1001635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tookey P.A., Thorne C., van Wyk J., Norton M. Maternal and foetal outcomes among 4118 women with HIV infection treated with lopinavir/ritonavir during pregnancy: analysis of population-based surveillance data from the national study of HIV in pregnancy and childhood in the United Kingdom and Ireland. BMC Infect Dis. 2016;16:65. doi: 10.1186/s12879-016-1400-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Retallack H., Di Lullo E., Arias C., et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci USA. 2016;113:14408–14413. doi: 10.1073/pnas.1618029113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bosseboeuf E., Aubry M., Nhan T., et al. Azithromycin inhibits the replication of Zika virus. J Antivir Antiretrovir. 2018;10:6–11. [Google Scholar]
  • 23.Choy K.T., Wong A.Y.-L., Kaewpreedee P., et al. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antiviral Res. 2020;178:104786. doi: 10.1016/j.antiviral.2020.104786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jeon S., Ko M., Lee J., et al. Identification of antiviral drug candidates against SARS-CoV-2 from FDA-approved drugs. Antimicrob Agents Chemother. 2020 doi: 10.1128/AAC.00819-20. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Touret F., Gilles M., Barral K., et al. In vitro screening of a FDA approved chemical library reveals potential inhibitors of SARS-CoV-2 replication. 2020. http://biorxiv.org/lookup/doi/10.1101/2020.04.03.023846 Available at: [DOI] [PMC free article] [PubMed]
  • 26.Sutton A.L., Acosta E.P., Larson K.B., Kerstner-Wood C.D., Tita A.T., Biggio J.R. Perinatal pharmacokinetics of azithromycin for cesarean prophylaxis. Am J Obstet Gynecol. 2015;212:812.e1–812.e6. doi: 10.1016/j.ajog.2015.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fan H., Gilbert R., O’Callaghan F., Li L. Associations between macrolide antibiotics prescribing during pregnancy and adverse child outcomes in the UK: population based cohort study. BMJ. 2020;368:m331. doi: 10.1136/bmj.m331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Heikkinen T., Laine K., Neuvonen P.J., Ekblad U. The transplacental transfer of the macrolide antibiotics erythromycin, roxithromycin and azithromycin. BJOG. 2000;107:770–775. doi: 10.1111/j.1471-0528.2000.tb13339.x. [DOI] [PubMed] [Google Scholar]
  • 29.Lin K.J., Mitchell A.A., Yau W.P., Louik C., Hernández-Díaz S. Safety of macrolides during pregnancy. Am J Obstet Gynecol. 2013;208:221.e1–221.e8. doi: 10.1016/j.ajog.2012.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Almaramhy H.H., Al-Zalabani A.H. The association of prenatal and postnatal macrolide exposure with subsequent development of infantile hypertrophic pyloric stenosis: a systematic review and meta-analysis. Ital J Pediatr. 2019;45:20. doi: 10.1186/s13052-019-0613-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bérard A., Sheehy O., Zhao J.P., Nordeng H. Use of macrolides during pregnancy and the risk of birth defects: a population-based study. Pharmacoepidemiol Drug Saf. 2015;24:1241–1248. doi: 10.1002/pds.3900. [DOI] [PubMed] [Google Scholar]
  • 32.Cooper W.O., Ray W.A., Griffin M.R. Prenatal prescription of macrolide antibiotics and infantile hypertrophic pyloric stenosis. Obstet Gynecol. 2002;100:101–106. doi: 10.1016/s0029-7844(02)02001-x. [DOI] [PubMed] [Google Scholar]
  • 33.Law I., Ilett K.F., Hackett L.P., et al. Transfer of chloroquine and desethylchloroquine across the placenta and into milk in Melanesian mothers. Br J Clin Pharmacol. 2008;65:674–679. doi: 10.1111/j.1365-2125.2008.03111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Osadchy A., Ratnapalan T., Koren G. Ocular toxicity in children exposed in utero to antimalarial drugs: review of the literature. J Rheumatol. 2011;38:2504–2508. doi: 10.3899/jrheum.110686. [DOI] [PubMed] [Google Scholar]
  • 35.Silva V.B., Sousa M.A., Kuniyoshi I.C., Aurélio F.S., Sampaio A.L., Oliveira C.A. Risk of hearing alterations in newborns of mothers treated for malaria. Int Tinnitus J. 2015;19:20–25. doi: 10.5935/0946-5448.20150004. [DOI] [PubMed] [Google Scholar]
  • 36.Amoura Z., Schermann J.M., Wechsler B., Zerah X., Goodeau P. Transplacental passage of colchicine in familial Mediterranean fever. J Rheumatol. 1994;21:383. [PubMed] [Google Scholar]
  • 37.Diav-Citrin O., Shechtman S., Schwartz V., et al. Pregnancy outcome after in utero exposure to colchicine. Am J Obstet Gynecol. 2010;203:144.e1–144.e6. doi: 10.1016/j.ajog.2010.02.063. [DOI] [PubMed] [Google Scholar]
  • 38.Costedoat-Chalumeau N., Amoura Z., Aymard G., et al. Evidence of transplacental passage of hydroxychloroquine in humans. Arthritis Rheum. 2002;46:1123–1124. doi: 10.1002/art.10150. [DOI] [PubMed] [Google Scholar]
  • 39.Costedoat-Chalumeau N., Amoura Z., Duhaut P., et al. Safety of hydroxychloroquine in pregnant patients with connective tissue diseases: a study of one hundred thirty-three cases compared with a control group. Arthritis Rheum. 2003;48:3207–3211. doi: 10.1002/art.11304. [DOI] [PubMed] [Google Scholar]
  • 40.Cimaz R., Brucato A., Meregalli E., Muscará M., Sergi P. Electroretinograms of children born to mothers treated with hydroxychloroquine during pregnancy and breast-feeding: comment on the article by Costedoat-Chalumeau et al. Arthritis Rheum. 2004;50:3056–3057. doi: 10.1002/art.20648. author reply 3057–8. [DOI] [PubMed] [Google Scholar]
  • 41.Waysbort A., Giroux M., Mansat V., Teixeira M., Dumas J.C., Puel J. Experimental study of transplacental passage of alpha interferon by two assay techniques. Antimicrob Agents Chemother. 1993;37:1232–1237. doi: 10.1128/aac.37.6.1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hellwig K., Duarte Caron F., Wicklein E.M., Bhatti A., Adamo A. Pregnancy outcomes from the global pharmacovigilance database on interferon beta-1b exposure. Ther Adv Neurol Disord. 2020;13 doi: 10.1177/1756286420910310. 1756286420910310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dumas J.C., Giroux M., Teixeira M.G., et al. Study of the transplacental transfer of interferon alfa-2a on the model of isolated perfused cotyledon [in French] Therapie. 1993;48:73–75. [PubMed] [Google Scholar]
  • 44.Coyle P.K., Sinclair S.M., Scheuerle A.E., Thorp J.M., Albano J.D., Rametta M.J. Final results from the Betaseron (interferon β-1b) Pregnancy Registry: a prospective observational study of birth defects and pregnancy-related adverse events. BMJ Open. 2014;4 doi: 10.1136/bmjopen-2013-004536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pons J.C., Lebon P., Frydman R., Delfraissy J.F. Pharmacokinetics of interferon-alpha in pregnant women and fetoplacental passage. Fetal Diagn Ther. 1995;10:7–10. doi: 10.1159/000264183. [DOI] [PubMed] [Google Scholar]
  • 46.Tada Y., Sakai M., Nakao Y., Maruyama A., Ono N., Koarada S. Placental transfer of tocilizumab in a patient with rheumatoid arthritis. Rheumatology (Oxford) 2019;58:1694–1695. doi: 10.1093/rheumatology/kez155. [DOI] [PubMed] [Google Scholar]
  • 47.Nakajima K., Watanabe O., Mochizuki M., Nakasone A., Ishizuka N., Murashima A. Pregnancy outcomes after exposure to tocilizumab: a retrospective analysis of 61 patients in Japan. Mod Rheumatol. 2016;26:667–671. doi: 10.3109/14397595.2016.1147405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hoeltzenbein M., Beck E., Rajwanshi R., et al. Tocilizumab use in pregnancy: analysis of a global safety database including data from clinical trials and post-marketing data. Semin Arthritis Rheum. 2016;46:238–245. doi: 10.1016/j.semarthrit.2016.05.004. [DOI] [PubMed] [Google Scholar]
  • 49.Youngstein T., Hoffmann P., Gül A., et al. International multi-centre study of pregnancy outcomes with interleukin-1 inhibitors. Rheumatology (Oxford) 2017;56:2102–2108. doi: 10.1093/rheumatology/kex305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Nicolas P., Maia M.F., Bassat Q., et al. Safety of oral ivermectin during pregnancy: a systematic review and meta-analysis. Lancet Glob Health. 2020;8:e92–e100. doi: 10.1016/S2214-109X(19)30453-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bérard A., Zhao J.P., Shui I., Colilla S. Leflunomide use during pregnancy and the risk of adverse pregnancy outcomes. Ann Rheum Dis. 2018;77:500–509. doi: 10.1136/annrheumdis-2017-212078. [DOI] [PubMed] [Google Scholar]
  • 52.Gavard L., Gil S., Peytavin G., et al. Placental transfer of lopinavir/ritonavir in the ex vivo human cotyledon perfusion model. Am J Obstet Gynecol. 2006;195:296–301. doi: 10.1016/j.ajog.2006.01.017. [DOI] [PubMed] [Google Scholar]
  • 53.Ivanovic J., Nicastri E., Anceschi M.M., et al. Transplacental transfer of antiretroviral drugs and newborn birth weight in HIV-infected pregnant women. Curr HIV Res. 2009;7:620–625. doi: 10.2174/157016209789973628. [DOI] [PubMed] [Google Scholar]
  • 54.Marzolini C., Rudin C., Decosterd L.A., et al. Transplacental passage of protease inhibitors at delivery. AIDS Lond Engl. 2002;16:889–893. doi: 10.1097/00002030-200204120-00008. [DOI] [PubMed] [Google Scholar]
  • 55.Huang H., Wang J., Li Q., et al. Transplacental transfer of oseltamivir phosphate and its metabolite oseltamivir carboxylate using the ex vivo human placenta perfusion model in Chinese Hans population. J Matern Fetal Neonatal Med. 2017;30:1288–1292. doi: 10.1080/14767058.2016.1211634. [DOI] [PubMed] [Google Scholar]
  • 56.Ehrenstein V., Kristensen N.R., Monz B.U., Clinch B., Kenwright A., Sørensen H.T. Oseltamivir in pregnancy and birth outcomes. BMC Infect Dis. 2018;18:519. doi: 10.1186/s12879-018-3423-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chambers C.D., Johnson D., Xu R., Luo Y., Jones K.L., OTIS Collaborative. Research Group Oseltamivir use in pregnancy: risk of birth defects, preterm delivery, and small for gestational age infants. Birth Defects Res. 2019;111:1487–1493. doi: 10.1002/bdr2.1566. [DOI] [PubMed] [Google Scholar]
  • 58.Sinclair S.M., Jones J.K., Miller R.K., Greene M.F., Kwo P.Y., Maddrey W.C. The Ribavirin Pregnancy Registry: an interim analysis of potential teratogenicity at the mid-point of enrollment. Drug Saf. 2017;40:1205–1218. doi: 10.1007/s40264-017-0566-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Russo F.M., Conings S., Allegaert K., et al. Sildenafil crosses the placenta at therapeutic levels in a dually perfused human cotyledon model. Am J Obstet Gynecol. 2018;219:619.e1–619.e10. doi: 10.1016/j.ajog.2018.08.041. [DOI] [PubMed] [Google Scholar]
  • 60.Dunn L., Greer R., Flenady V., Kumar S. Sildenafil in pregnancy: a systematic review of maternal tolerance and obstetric and perinatal outcomes. Fetal Diagn Ther. 2017;41:81–88. doi: 10.1159/000453062. [DOI] [PubMed] [Google Scholar]
  • 61.Russo F.M., Benachi A., Van Mieghem T., et al. Antenatal sildenafil administration to prevent pulmonary hypertension in congenital diaphragmatic hernia (SToP-PH): study protocol for a phase I/IIb placenta transfer and safety study. Trials. 2018;19:524. doi: 10.1186/s13063-018-2897-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ferreira R.D.D.S., Negrini R., Bernardo W.M., Simões R., Piato S. The effects of sildenafil in maternal and fetal outcomes in pregnancy: a systematic review and meta-analysis. PloS One. 2019;14 doi: 10.1371/journal.pone.0219732. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 63.Madrid P.B., Panchal R.G., Warren T.K., et al. Evaluation of Ebola virus inhibitors for drug repurposing. ACS Infect Dis. 2015;1:317–326. doi: 10.1021/acsinfecdis.5b00030. [DOI] [PubMed] [Google Scholar]
  • 64.Arabi Y.M., Deeb A.M., Al-Hameed F., et al. Macrolides in critically ill patients with Middle East Respiratory Syndrome. Int J Infect Dis. 2019;81:184–190. doi: 10.1016/j.ijid.2019.01.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wang D., Hu B., Hu C., et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323:1061–1069. doi: 10.1001/jama.2020.1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gautret P., Lagier J.C., Parola P., et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents. 2020 doi: 10.1016/j.ijantimicag.2020.105949. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kemp M.W., Miura Y., Payne M.S., et al. Maternal intravenous administration of azithromycin results in significant fetal uptake in a sheep model of second trimester pregnancy. Antimicrob Agents Chemother. 2014;58:6581–6591. doi: 10.1128/AAC.03721-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Acosta E.P., Grigsby P.L., Larson K.B., et al. Transplacental transfer of azithromycin and its use for eradicating intra-amniotic ureaplasma infection in a primate model. J Infect Dis. 2014;209:898–904. doi: 10.1093/infdis/jit578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ooi E.E., Chew J.S.W., Loh J.P., Chua R.C.S. In vitro inhibition of human influenza A virus replication by chloroquine. Virol J. 2006;3:39. doi: 10.1186/1743-422X-3-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Paton N.I., Lee L., Xu Y., et al. Chloroquine for influenza prevention: a randomised, double-blind, placebo controlled trial. Lancet Infect Dis. 2011;11:677–683. doi: 10.1016/S1473-3099(11)70065-2. [DOI] [PubMed] [Google Scholar]
  • 71.Savarino A., Di Trani L., Donatelli I., Cauda R., Cassone A. New insights into the antiviral effects of chloroquine. Lancet Infect Dis. 2006;6:67–69. doi: 10.1016/S1473-3099(06)70361-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Vincent M.J., Bergeron E., Benjannet S., et al. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J. 2005;2:69. doi: 10.1186/1743-422X-2-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Savarino A., Boelaert J.R., Cassone A., Majori G., Cauda R. Effects of chloroquine on viral infections: an old drug against today’s diseases? Lancet Infect Dis. 2003;3:722–727. doi: 10.1016/S1473-3099(03)00806-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Keyaerts E., Vijgen L., Maes P., Neyts J., Van Ranst M. In vitro inhibition of severe acute respiratory syndrome coronavirus by chloroquine. Biochem Biophys Res Commun. 2004;323:264–268. doi: 10.1016/j.bbrc.2004.08.085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wang M., Cao R., Zhang L., et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269–271. doi: 10.1038/s41422-020-0282-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Devaux C.A., Rolain J.M., Colson P., Raoult D. New insights on the antiviral effects of chloroquine against coronavirus: what to expect for COVID-19? Int J Antimicrob Agents. 2020;55:105938. doi: 10.1016/j.ijantimicag.2020.105938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Iglesias Cubero G., Rodriguez Reguero J.J., Rojo Ortega J.M. Restrictive cardiomyopathy caused by chloroquine. Br Heart J. 1993;69:451–452. doi: 10.1136/hrt.69.5.451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tönnesmann E., Stroehmann I., Kandolf R., et al. Cardiomyopathy caused by longterm treatment with chloroquine: a rare disease, or a rare diagnosis? J Rheumatol. 2012;39:1099–1103. doi: 10.3899/jrheum.110959. [DOI] [PubMed] [Google Scholar]
  • 79.Yogasundaram H., Hung W., Paterson I.D., Sergi C., Oudit G.Y. Chloroquine-induced cardiomyopathy: a reversible cause of heart failure. ESC Heart Fail. 2018;5:372–375. doi: 10.1002/ehf2.12276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Vereckei A., Fazakas A., Baló T., Fekete B., Molnár M.J., Karádi I. Chloroquine cardiotoxicity mimicking connective tissue disease heart involvement. Immunopharmacol Immunotoxicol. 2013;35:304–306. doi: 10.3109/08923973.2013.766801. [DOI] [PubMed] [Google Scholar]
  • 81.Salman S., Baiwog F., Page-Sharp M., et al. Optimal antimalarial dose regimens for chloroquine in pregnancy based on population pharmacokinetic modelling. Int J Antimicrob Agents. 2017;50:542–551. doi: 10.1016/j.ijantimicag.2017.05.011. [DOI] [PubMed] [Google Scholar]
  • 82.Karunajeewa H.A., Salman S., Mueller I., et al. Pharmacokinetics of chloroquine and monodesethylchloroquine in pregnancy. Antimicrob Agents Chemother. 2010;54:1186–1192. doi: 10.1128/AAC.01269-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Augustijns P., Jongsma H.W., Verbeke N. Transplacental distribution of chloroquine in sheep. Dev Pharmacol Ther. 1991;17:191–199. doi: 10.1159/000457522. [DOI] [PubMed] [Google Scholar]
  • 84.Essien E.E., Afamefuna G.C. Chloroquine and its metabolites in human cord blood, neonatal blood, and urine after maternal medication. Clin Chem. 1982;28:1148–1152. [PubMed] [Google Scholar]
  • 85.Blanc A., Bonnet F., Brun-Vezinet F., et al. Prise en charge médicale des personnes vivants avec le VIH. Recommandations du groupe d’experts. Désir d’enfant et grossesse. 2018;2018:52. [Google Scholar]
  • 86.European Pregnancy and Paediatric HIV Cohort Collaboration (EPPICC) Study Group Nucleoside reverse transcriptase inhibitor backbones and pregnancy outcomes. AIDS. 2019;33:295–304. doi: 10.1097/QAD.0000000000002039. [DOI] [PubMed] [Google Scholar]
  • 87.McGready R., Lee S.J., Wiladphaingern J., et al. Adverse effects of falciparum and vivax malaria and the safety of antimalarial treatment in early pregnancy: a population-based study. Lancet Infect Dis. 2012;12:388–396. doi: 10.1016/S1473-3099(11)70339-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Yao X., Ye F., Zhang M., et al. In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Clin Infect Dis. 2020 doi: 10.1093/cid/ciaa237. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Liu J., Cao R., Xu M., et al. Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro. Cell Discov. 2020;6:16. doi: 10.1038/s41421-020-0156-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hong W. Combating COVID-19 with chloroquine. J Mol Cell Biol. 2020;12:249–250. doi: 10.1093/jmcb/mjaa015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Geleris J., Sun Y., Platt J., et al. Observational study of hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med. 2020;382:2411–2418. doi: 10.1056/NEJMoa2012410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Chen J., Liu D., Liu L., et al. [A pilot study of hydroxychloroquine in treatment of patients with moderate COVID-19] Zhejiang Da Xue Xue Bao Yi Xue Ban. 2020;49:215–219. doi: 10.3785/j.issn.1008-9292.2020.03.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chen Z., Hu J., Zhang Z., et al. Efficacy of hydroxychloroquine in patients with COVID-19: results of a randomized clinical trial. medRxiv. Preprint posted online April 10, 2020. http://medrxiv.org/lookup/doi/10.1101/2020.03.22.20040758 Available at:
  • 94.Tang W., Cao Z., Han M., et al. Hydroxychloroquine in patients with mainly mild to moderate coronavirus disease 2019: an open-label, randomised controlled trial. BMJ. 2020;369:m1849. doi: 10.1136/bmj.m1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Molina J.M., Delaugerre C., Le Goff J., et al. No evidence of rapid antiviral clearance or clinical benefit with the combination of hydroxychloroquine and azithromycin in patients with severe COVID-19 infection. Med Mal Infect. 2020;50:384. doi: 10.1016/j.medmal.2020.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mahevas M., Tran V.-T., Roumier M., et al. No evidence of clinical efficacy of hydroxychloroquine in patients hospitalized for COVID-19 infection with oxygen requirement: results of a study using routinely collected data to emulate a target trial. 2020. medRxiv. Preprint posted online April 14, 2020. http://medrxiv.org/lookup/doi/10.1101/2020.04.10.20060699 Available at:
  • 97.Mallat J., Hamed F., Balkis M., et al. Hydroxychloroquine is associated with slower viral clearance in clinical COVID-19 patients with mild to moderate disease: a retrospective study. 2020. medRxiv. Preprint posted online May 2, 2020. http://medrxiv.org/lookup/doi/10.1101/2020.04.27.20082180 Available at: [DOI] [PMC free article] [PubMed]
  • 98.Yu B., Wang D.W., Li C. Hydroxychloroquine application is associated with a decreased mortality in critically ill patients with COVID-19. 2020. medRxiv. Preprint posted online May 1, 2020. http://medrxiv.org/lookup/doi/10.1101/2020.04.27.20073379 Available at:
  • 99.Mehra M.R., Desai S.S., Ruschitzka F., Patel A.N. Retraction-Hydroxychloroquine or chloroquine with or without a macrolide for treatment of COVID-19: a multinational registry analysis [retraction of: Lancet 2020 May 22] Lancet. 2020;395:1820. doi: 10.1016/S0140-6736(20)31324-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Million M., Lagier J.C., Gautret P., et al. Early treatment of COVID-19 patients with hydroxychloroquine and azithromycin: a retrospective analysis of 1061 cases in Marseille, France. Travel Med Infect Dis. 2020 doi: 10.1016/j.tmaid.2020.101738. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Légifrance [Order of January 13, 2020 , classification on the lists of poisonous substances] https://www.legifrance.gouv.fr/affichTexte.do?cidTexte=JORFTEXT000041400024&categorieLien=id Available at: Accessed May 1, 2020.
  • 102.Jorge A., Ung C., Young L.H., Melles R.B., Choi H.K. Hydroxychloroquine retinopathy - implications of research advances for rheumatology care. Nat Rev Rheumatol. 2018;14:693–703. doi: 10.1038/s41584-018-0111-8. [DOI] [PubMed] [Google Scholar]
  • 103.Carter K.L., Do D.V. Hydroxychloroquine-induced retinal toxicity. J Rheumatol. 2020;47:632. doi: 10.3899/jrheum.190538. [DOI] [PubMed] [Google Scholar]
  • 104.Chatre C., Roubille F., Vernhet H., Jorgensen C., Pers Y.M. Cardiac complications attributed to chloroquine and hydroxychloroquine: a systematic review of the literature. Drug Saf. 2018;41:919–931. doi: 10.1007/s40264-018-0689-4. [DOI] [PubMed] [Google Scholar]
  • 105.Vroom F., de Walle H.E.K., van de Laar M.A.J.F., Brouwers J.R.B.J., de Jong-van den Berg L.T.W. Disease-modifying antirheumatic drugs in pregnancy: current status and implications for the future. Drug Saf. 2006;29:845–863. doi: 10.2165/00002018-200629100-00003. [DOI] [PubMed] [Google Scholar]
  • 106.Abarientos C., Sperber K., Shapiro D.L., Aronow W.S., Chao C.P., Ash J.Y. Hydroxychloroquine in systemic lupus erythematosus and rheumatoid arthritis and its safety in pregnancy. Expert Opin Drug Saf. 2011;10:705–714. doi: 10.1517/14740338.2011.566555. [DOI] [PubMed] [Google Scholar]
  • 107.Yao T.T., Qian J.D., Zhu W.Y., Wang Y., Wang G.Q. A systematic review of lopinavir therapy for SARS coronavirus and MERS coronavirus-a possible reference for coronavirus disease-19 treatment option. J Med Virol. 2020;92:556–563. doi: 10.1002/jmv.25729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Sheahan T.P., Sims A.C., Leist S.R., et al. Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat Commun. 2020;11:222. doi: 10.1038/s41467-019-13940-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Cao B., Wang Y., Wen D., et al. A trial of lopinavir-ritonavir in adults hospitalized with severe Covid-19. N Engl J Med. 2020;382:1787–1799. doi: 10.1056/NEJMoa2001282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Tubiana R., Mandelbrot L., Le Chenadec J., et al. Lopinavir/ritonavir monotherapy as a nucleoside analogue-sparing strategy to prevent HIV-1 mother-to-child transmission: the ANRS 135 PRIMEVA phase 2/3 randomized trial. Clin Infect Dis. 2013;57:891–902. doi: 10.1093/cid/cit390. [DOI] [PubMed] [Google Scholar]
  • 111.Fauchet F., Treluyer J.M., Illamola S.M., et al. Population approach to analyze the pharmacokinetics of free and total lopinavir in HIV-infected pregnant women and consequences for dose adjustment. Antimicrob Agents Chemother. 2015;59:5727–5735. doi: 10.1128/AAC.00863-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cressey T.R., Best B.M., Achalapong J., et al. Reduced indinavir exposure during pregnancy. Br J Clin Pharmacol. 2013;76:475–483. doi: 10.1111/bcp.12078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ceccaldi P.F., Gavard L., Mandelbrot L., et al. Functional role of p-glycoprotein and binding protein effect on the placental transfer of lopinavir/ritonavir in the ex vivo human perfusion model. Obstet Gynecol Int. 2009;2009:726593. doi: 10.1155/2009/726593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mandelbrot L., Duro D., Belissa E., Peytavin G. Placental transfer of darunavir in an ex vivo human cotyledon perfusion model. Antimicrob Agents Chemother. 2014;58:5617–5620. doi: 10.1128/AAC.03184-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Fowler M.G., Qin M., Fiscus S.A., et al. Benefits and risks of antiretroviral therapy for perinatal HIV prevention. N Engl J Med. 2016;375:1726–1737. doi: 10.1056/NEJMoa1511691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Powis K.M., Kitch D., Ogwu A., et al. Increased risk of preterm delivery among HIV-infected women randomized to protease versus nucleoside reverse transcriptase inhibitor-based HAART during pregnancy. J Infect Dis. 2011;204:506–514. doi: 10.1093/infdis/jir307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Simon A., Warszawski J., Kariyawasam D., et al. Association of prenatal and postnatal exposure to lopinavir-ritonavir and adrenal dysfunction among uninfected infants of HIV-infected mothers. JAMA. 2011;306:70–78. doi: 10.1001/jama.2011.915. [DOI] [PubMed] [Google Scholar]
  • 118.Tchesnokov E.P., Feng J.Y., Porter D.P., Götte M. Mechanism of inhibition of Ebola virus RNA-dependent RNA polymerase by remdesivir. Viruses. 2019;11:326. doi: 10.3390/v11040326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Mulangu S., Dodd L.E., Davey R.T., et al. A randomized, controlled trial of Ebola virus disease therapeutics. N Engl J Med. 2019;381:2293–2303. doi: 10.1056/NEJMoa1910993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Sheahan T.P., Sims A.C., Graham R.L., et al. Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci Transl Med. 2017;9 doi: 10.1126/scitranslmed.aal3653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Brown A.J., Won J.J., Graham R.L., et al. Broad spectrum antiviral remdesivir inhibits human endemic and zoonotic deltacoronaviruses with a highly divergent RNA dependent RNA polymerase. Antiviral Res. 2019;169:104541. doi: 10.1016/j.antiviral.2019.104541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.de Wit E., Feldmann F., Cronin J., et al. Prophylactic and therapeutic remdesivir (GS-5734) treatment in the rhesus macaque model of MERS-CoV infection. Proc Natl Acad Sci USA. 2020;117:6771–6776. doi: 10.1073/pnas.1922083117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Gordon C.J., Tchesnokov E.P., Feng J.Y., Porter D.P., Götte M. The antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus. J Biol Chem. 2020;295:4773–4779. doi: 10.1074/jbc.AC120.013056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Beigel J.H., Tomashek K.M., Dodd L.E., et al. Remdesivir for the treatment of Covid-19 - preliminary report. N Engl J Med. 2020 doi: 10.1056/NEJMc2022236. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 125.Wang Y., Zhang D., Du G., et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395:1569–1578. doi: 10.1016/S0140-6736(20)31022-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Grein J., Ohmagari N., Shin D., et al. Compassionate use of remdesivir for patients with severe Covid-19. N Engl J Med. 2020;382:2327–2336. doi: 10.1056/NEJMoa2007016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Energy Market Authority of Singapore/204393/2016, Committee for Medicinal Products for Human Use (CHMP) Assessment report on Medicinal products under development for the treatment of Ebola. 2016 [Google Scholar]
  • 128.Haut Conseil de la Santé Publique Avis relatif à la prise en charge des cas confirmés d’infection au virus SARS-CoV-2. https://www.vie-publique.fr/sites/default/files/rapport/pdf/273958.pdf Available at: Accessed May 1, 2020.
  • 129.Jin Y., Yang H., Ji W., et al. Virology, epidemiology, pathogenesis, and control of COVID-19. Viruses. 2020;12:372. doi: 10.3390/v12040372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.De Meyer S., Bojkova D., Cinatl J., et al. Lack of antiviral activity of darunavir against SARS-CoV-2. Int J Infect Dis. 2020;97:7–10. doi: 10.1016/j.ijid.2020.05.085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Bixler S.L., Bocan T.M., Wells J., et al. Efficacy of favipiravir (T-705) in nonhuman primates infected with Ebola virus or Marburg virus. Antiviral Res. 2018;151:97–104. doi: 10.1016/j.antiviral.2017.12.021. [DOI] [PubMed] [Google Scholar]
  • 132.Delang L., Abdelnabi R., Neyts J. Favipiravir as a potential countermeasure against neglected and emerging RNA viruses. Antiviral Res. 2018;153:85–94. doi: 10.1016/j.antiviral.2018.03.003. [DOI] [PubMed] [Google Scholar]
  • 133.Furuta Y., Komeno T., Nakamura T. Favipiravir (T-705), a broad spectrum inhibitor of viral RNA polymerase. Proc Jpn Acad Ser B Phys Biol Sci. 2017;93:449–463. doi: 10.2183/pjab.93.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Oestereich L., Lüdtke A., Wurr S., Rieger T., Muñoz-Fontela C., Günther S. Successful treatment of advanced Ebola virus infection with T-705 (favipiravir) in a small animal model. Antiviral Res. 2014;105:17–21. doi: 10.1016/j.antiviral.2014.02.014. [DOI] [PubMed] [Google Scholar]
  • 135.Kerber R., Lorenz E., Duraffour S., et al. Laboratory findings, compassionate use of favipiravir, and outcome in patients With Ebola virus disease, Guinea, 2015-a retrospective observational study. J Infect Dis. 2019;220:195–202. doi: 10.1093/infdis/jiz078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Baz M., Carbonneau J., Rhéaume C., Cavanagh M.-H., Boivin G. Combination therapy with oseltamivir and favipiravir delays mortality but does not prevent oseltamivir resistance in immunodeficient mice infected with pandemic A(H1N1) influenza virus. Viruses. 2018;10:610. doi: 10.3390/v10110610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wang Y., Fan G., Salam A., et al. Comparative effectiveness of combined favipiravir and oseltamivir therapy versus oseltamivir monotherapy in critically ill patients with influenza virus infection. J Infect Dis. 2020;221:1688–1698. doi: 10.1093/infdis/jiz656. [DOI] [PubMed] [Google Scholar]
  • 138.Hayden F.G., Shindo N. Influenza virus polymerase inhibitors in clinical development. Curr Opin Infect Dis. 2019;32:176–186. doi: 10.1097/QCO.0000000000000532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Omrani A.S., Saad M.M., Baig K., et al. Ribavirin and interferon alfa-2a for severe Middle East respiratory syndrome coronavirus infection: a retrospective cohort study. Lancet Infect Dis. 2014;14:1090–1095. doi: 10.1016/S1473-3099(14)70920-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Arabi Y.M., Shalhoub S., Mandourah Y., et al. Ribavirin and interferon therapy for critically ill patients With Middle East respiratory syndrome: a multicenter observational study. Clin Infect Dis. 2020;70:1837–1844. doi: 10.1093/cid/ciz544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Roberts S.S., Miller R.K., Jones J.K., et al. The Ribavirin Pregnancy Registry: Findings after 5 years of enrollment, 2003–2009. Birth Defects Res A Clin Mol Teratol. 2010;88:551–559. doi: 10.1002/bdra.20682. [DOI] [PubMed] [Google Scholar]
  • 142.Sibiude J., Warszawski J., Tubiana R., et al. Premature delivery in HIV-infected women starting protease inhibitor therapy during pregnancy: role of the ritonavir boost? Clin Infect Dis. 2012;54:1348–1360. doi: 10.1093/cid/cis198. [DOI] [PubMed] [Google Scholar]
  • 143.Yamamoto N., Yang R., Yoshinaka Y., et al. HIV protease inhibitor nelfinavir inhibits replication of SARS-associated coronavirus. Biochem Biophys Res Commun. 2004;318:719–725. doi: 10.1016/j.bbrc.2004.04.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Khamitov R.A., Loginova SIa, Shchukina V.N., Borisevich S.V., Maksimov V.A., Shuster A.M. Antiviral activity of arbidol and its derivatives against the pathogen of severe acute respiratory syndrome in the cell cultures[in Russian] Vopr Virusol. 2008;53:9–13. [PubMed] [Google Scholar]
  • 145.Deng L., Li C., Zeng Q., et al. Arbidol combined with LPV/r versus LPV/r alone against Corona Virus Disease 2019: a retrospective cohort study. J Infect. 2020;81:e1–e5. doi: 10.1016/j.jinf.2020.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Lian N., Xie H., Lin S., Huang J., Zhao J., Lin Q. Umifenovir treatment is not associated with improved outcomes in patients with coronavirus disease 2019: a retrospective study. Clin Microbiol Infect. 2020;26:917–921. doi: 10.1016/j.cmi.2020.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Yu N., Li W., Kang Q., et al. Clinical features and obstetric and neonatal outcomes of pregnant patients with COVID-19 in Wuhan, China: a retrospective, single-centre, descriptive study. Lancet Infect Dis. 2020;20:559–564. doi: 10.1016/S1473-3099(20)30176-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Pérez R., Palma C., Núñez M.J., Cox J. Pharmacokinetics of ivermectin after maternal or fetal intravenous administration in sheep. J Vet Pharmacol Ther. 2008;31:406–414. doi: 10.1111/j.1365-2885.2008.00971.x. [DOI] [PubMed] [Google Scholar]
  • 149.Avonex Highlights of prescribing information. 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/103780s5204lbl.pdf Available at: Accessed April 20, 2020.
  • 150.Hellwig K., Geissbuehler Y., Sabidó M., et al. Pregnancy outcomes in interferon-beta-exposed patients with multiple sclerosis: results from the European interferon-beta Pregnancy Registry. J Neurol. 2020;267:1715–1723. doi: 10.1007/s00415-020-09762-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Wong H.K., Lee C.K. Pivotal role of convalescent plasma in managing emerging infectious diseases. Vox Sang. 2020 doi: 10.1111/vox.12927. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Marano G., Vaglio S., Pupella S., et al. Convalescent plasma: new evidence for an old therapeutic tool? Blood Transfus. 2016;14:152–157. doi: 10.2450/2015.0131-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.van Griensven J., Edwards T., de Lamballerie X., et al. Evaluation of convalescent plasma for Ebola virus disease in guinea. N Engl J Med. 2016;374:33–42. doi: 10.1056/NEJMoa1511812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Cheng Y., Wong R., Soo Y.O., et al. Use of convalescent plasma therapy in SARS patients in Hong Kong. Eur J Clin Microbiol Infect Dis. 2005;24:44–46. doi: 10.1007/s10096-004-1271-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Zhang B., Liu S., Tan T., et al. Treatment with convalescent plasma for critically ill patients with Severe Acute Respiratory Syndrome Coronavirus 2 infection. Chest. 2020 doi: 10.1016/j.chest.2020.03.039. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Simister N.E. Placental transport of immunoglobulin G. Vaccine. 2003;21:3365–3369. doi: 10.1016/s0264-410x(03)00334-7. [DOI] [PubMed] [Google Scholar]
  • 157.Channappanavar R., Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 2017;39:529–539. doi: 10.1007/s00281-017-0629-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Saito J., Yakuwa N., Takai C., et al. Tocilizumab concentrations in maternal serum and breast milk during breastfeeding and a safety assessment in infants: a case study. Rheumatology (Oxford) 2018;57:1499–1501. doi: 10.1093/rheumatology/key091. [DOI] [PubMed] [Google Scholar]
  • 159.Götestam Skorpen C., Hoeltzenbein M., Tincani A., et al. The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis. 2016;75:795–810. doi: 10.1136/annrheumdis-2015-208840. [DOI] [PubMed] [Google Scholar]
  • 160.Comi G., Hartung H.P., Bakshi R., Williams I.M., Wiendl H. Benefit-risk profile of sphingosine-1-phosphate receptor modulators in relapsing and secondary progressive multiple sclerosis. Drugs. 2017;77:1755–1768. doi: 10.1007/s40265-017-0814-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Saida T., Itoyama Y., Kikuchi S., et al. Long-term efficacy and safety of fingolimod in Japanese patients with relapsing multiple sclerosis: 3-year results of the phase 2 extension study. BMC Neurol. 2017;17:17. doi: 10.1186/s12883-017-0794-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Geissbühler Y., Vile J., Koren G., et al. Evaluation of pregnancy outcomes in patients with multiple sclerosis after fingolimod exposure. Ther Adv Neurol Disord. 2018;11 doi: 10.1177/1756286418804760. 1756286418804760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Al-Salama Z.T., Scott L.J. Baricitinib: a review in rheumatoid arthritis. Drugs. 2018;78:761–772. doi: 10.1007/s40265-018-0908-4. [DOI] [PubMed] [Google Scholar]
  • 164.Costanzo G., Firinu D., Losa F., Deidda M., Barca M.P., Del Giacco S. Baricitinib exposure during pregnancy in rheumatoid arthritis. Ther Adv Musculoskelet Dis. 2020;12 doi: 10.1177/1759720X19899296. 1759720X19899296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Gerosa M., Argolini L.M., Artusi C., Chighizola C.B. The use of biologics and small molecules in pregnant patients with rheumatic diseases. Expert Rev Clin Pharmacol. 2018;11:987–998. doi: 10.1080/17512433.2018.1525293. [DOI] [PubMed] [Google Scholar]
  • 166.Mahadevan U., Kane S., Sandborn W.J., et al. Intentional infliximab use during pregnancy for induction or maintenance of remission in Crohn’s disease. Aliment Pharmacol Ther. 2005;21:733–738. doi: 10.1111/j.1365-2036.2005.02405.x. [DOI] [PubMed] [Google Scholar]
  • 167.Vasiliauskas E.A., Church J.A., Silverman N., Barry M., Targan S.R., Dubinsky M.C. Case report: evidence for transplacental transfer of maternally administered infliximab to the newborn. Clin Gastroenterol Hepatol. 2006;4:1255–1258. doi: 10.1016/j.cgh.2006.07.018. [DOI] [PubMed] [Google Scholar]
  • 168.Steenholdt C., Al-Khalaf M., Ainsworth M.A., Brynskov J. Therapeutic infliximab drug level in a child born to a woman with ulcerative colitis treated until gestation week 31. J Crohns Colitis. 2012;6:358–361. doi: 10.1016/j.crohns.2011.10.002. [DOI] [PubMed] [Google Scholar]
  • 169.Cumhur Cure M., Kucuk A., Cure E. Colchicine may not be effective in COVID-19 infection; it may even be harmful? Clin Rheumatol. 2020;39:2101–2102. doi: 10.1007/s10067-020-05144-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Gendelman O., Amital H., Bragazzi N.L., Watad A., Chodick G. Continuous hydroxychloroquine or colchicine therapy does not prevent infection with SARS-CoV-2: insights from a large healthcare database analysis. Autoimmun Rev. 2020;19:102566. doi: 10.1016/j.autrev.2020.102566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Deftereos S.G., Siasos G., Giannopoulos G., et al. The Greek study in the effects of colchicine in COvid-19 complications prevention (GRECCO-19 study): rationale and study design. Hell J Cardiol HJC Hell Kardiol Epitheor. 2020 doi: 10.1016/j.hjc.2020.03.002. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Indraratna P.L., Virk S., Gurram D., Day R.O. Use of colchicine in pregnancy: a systematic review and meta-analysis. Rheumatology (Oxford) 2018;57:382–387. doi: 10.1093/rheumatology/kex353. [DOI] [PubMed] [Google Scholar]
  • 173.Petersen M.C., Nation R.L., Ashley J.J., McBride W.G. The placental transfer of betamethasone. Eur J Clin Pharmacol. 1980;18:245–247. doi: 10.1007/BF00563006. [DOI] [PubMed] [Google Scholar]
  • 174.Ward R.M. Drug therapy of the fetus. J Clin Pharmacol. 1993;33:780–789. doi: 10.1002/j.1552-4604.1993.tb01952.x. [DOI] [PubMed] [Google Scholar]
  • 175.Yang K. Placental 11 beta-hydroxysteroid dehydrogenase: barrier to maternal glucocorticoids. Rev Reprod. 1997;2:129–132. doi: 10.1530/ror.0.0020129. [DOI] [PubMed] [Google Scholar]
  • 176.Crowley P. Prophylactic corticosteroids for preterm birth. Cochrane Database Syst Rev. 2000;2:CD000065. doi: 10.1002/14651858.CD000065. [DOI] [PubMed] [Google Scholar]
  • 177.Chow J.H., Mazzeffi M.A., McCurdy M.T. Angiotensin II for the treatment of COVID-19-Related vasodilatory shock. Anesth Analg. 2020;131:102–105. doi: 10.1213/ANE.0000000000004825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Kuhn J.H., Li W., Choe H., Farzan M. Angiotensin-converting enzyme 2: a functional receptor for SARS coronavirus. Cell Mol Life Sci. 2004;61:2738–2743. doi: 10.1007/s00018-004-4242-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Vaduganathan M., Vardeny O., Michel T., McMurray J.J.V., Pfeffer M.A., Solomon S.D. Renin-angiotensin-aldosterone system inhibitors in patients with Covid-19. N Engl J Med. 2020;382:1653–1659. doi: 10.1056/NEJMsr2005760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Alwan S., Polifka J.E., Friedman J.M. Angiotensin II receptor antagonist treatment during pregnancy. Birth Defects Res A Clin Mol Teratol. 2005;73:123–130. doi: 10.1002/bdra.20102. [DOI] [PubMed] [Google Scholar]
  • 181.Nadeem S., Hashmat S., Defreitas M.J., et al. Renin angiotensin system blocker fetopathy: a Midwest Pediatric Nephrology Consortium Report. J Pediatr. 2015;167:881–885. doi: 10.1016/j.jpeds.2015.05.045. [DOI] [PubMed] [Google Scholar]
  • 182.Delforce S.J., Lumbers E.R., Ellery S.J., Murthi P., Pringle K.G. Dysregulation of the placental renin-angiotensin system in human fetal growth restriction. Reproduction. 2019;158:237–245. doi: 10.1530/REP-18-0633. [DOI] [PubMed] [Google Scholar]
  • 183.Matsui D. Ethics of studies of drugs in pregnancy. Pediatr Drugs. 2015;17:31–35. doi: 10.1007/s40272-014-0104-2. [DOI] [PubMed] [Google Scholar]
  • 184.Heyrana K., Byers H.M., Stratton P. Increasing the participation of pregnant women in clinical trials. JAMA. 2018;320:2077–2078. doi: 10.1001/jama.2018.17716. [DOI] [PubMed] [Google Scholar]
  • 185.Beran R.G. The ethics of excluding women who become pregnant while participating in clinical trials of anti-epileptic medications. Seizure. 2006;15:563–570. doi: 10.1016/j.seizure.2006.08.008. [DOI] [PubMed] [Google Scholar]
  • 186.Kaye D.K. The moral imperative to approve pregnant women’s participation in randomized clinical trials for pregnancy and newborn complications. Philos Eth Humanit Med. 2019;14:11. doi: 10.1186/s13010-019-0081-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Bourget P., Roulot C., Fernandez H. Models for placental transfer studies of drugs. Clin Pharmacokinet. 1995;28:161–180. doi: 10.2165/00003088-199528020-00006. [DOI] [PubMed] [Google Scholar]
  • 188.Dallmann A., Liu X.I., Burckart G.J., van den Anker J. Drug transporters expressed in the human placenta and models for studying maternal-fetal drug transfer. J Clin Pharmacol. 2019;59(Suppl 1):S70–S81. doi: 10.1002/jcph.1491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Shang L., Wang Y., Lyu Y. Clinical effect of mifepristone on patients with ovarian cancer in pregnancy. Pak J Pharm Sci. 2019 32(1(Special)):421–426. [PubMed] [Google Scholar]
  • 190.Korenaga T-RK, Tewari K.S. Gynecologic cancer in pregnancy. Gynecol Oncol. 2020;157:799–809. doi: 10.1016/j.ygyno.2020.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Polizzi S., Mahajan V.B. Intravitreal anti-VEGF injections in pregnancy: case series and review of literature. J Ocul Pharmacol Ther. 2015;31:605–610. doi: 10.1089/jop.2015.0056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Tarantola R.M., Folk J.C., Boldt H.C., Mahajan V.B. Intravitreal bevacizumab during pregnancy. Retina. 2010;30:1405–1411. doi: 10.1097/IAE.0b013e3181f57d58. [DOI] [PubMed] [Google Scholar]
  • 193.Lenz W. A short history of thalidomide embryopathy. Teratology. 1988;38:203–215. doi: 10.1002/tera.1420380303. [DOI] [PubMed] [Google Scholar]
  • 194.Mellin G.W., Katzenstein M. The saga of thalidomide. Neuropathy to embryopathy, with case reports of congenital anomalies. N Engl J Med. 1962;267:1184–1193. doi: 10.1056/NEJM196212062672305. [DOI] [PubMed] [Google Scholar]
  • 195.Nobuhara I., Harada N., Haruta N., et al. Multiple metastatic gestational trophoblastic disease after a twin pregnancy with complete hydatidiform mole and coexisting fetus, following assisted reproductive technology: case report and literature review. Taiwan J Obstet Gynecol. 2018;57:588–593. doi: 10.1016/j.tjog.2018.06.020. [DOI] [PubMed] [Google Scholar]
  • 196.Karimi Zarchi M., Behtash N., Modares Gilani M. Good pregnancy outcome after prenatal exposure to bleomycin, etoposide and cisplatin for ovarian immature teratoma: a case report and literature review. Arch Gynecol Obstet. 2008;277:75–78. doi: 10.1007/s00404-007-0416-3. [DOI] [PubMed] [Google Scholar]
  • 197.Ngan H.Y.S., Tam K.F., Lam K.W., Chan K.K.L. Methotrexate, bleomycin, and etoposide in the treatment of gestational trophoblastic neoplasia. Obstet Gynecol. 2006;107:1012–1017. doi: 10.1097/01.AOG.0000207577.67765.8e. [DOI] [PubMed] [Google Scholar]
  • 198.Song Y., Wang Z., Hao Z., et al. Requirement for etoposide in the treatment of pregnancy related hemophagocytic lymphohistiocytosis: a multicenter retrospective study. Orphanet J Rare Dis. 2019;14:50. doi: 10.1186/s13023-019-1033-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Sibiude J., Warszawski J., Tubiana R., et al. Liver enzyme elevation in pregnant women receiving antiretroviral therapy in the ANRS-French perinatal cohort. J Acquir Immune Defic Syndr. 2019 May 1;81:83–94. doi: 10.1097/QAI.0000000000001963. [DOI] [PubMed] [Google Scholar]
  • 200.Faure-Bardon V., Mandelbrot L., Duro D., Dussaux C., Le M., Peytavin G. Placental transfer of elvitegravir and cobicistat in an ex-vivo human cotyledon double perfusion model. AIDS Lond Engl. 2018;32:321–325. doi: 10.1097/QAD.0000000000001681. [DOI] [PubMed] [Google Scholar]
  • 201.Gavard L., Beghin D., Forestier F., et al. Contribution and limit of the model of perfused cotyledon to the study of placental transfer of drugs. Example of a protease inhibitor of HIV: nelfinavir. Eur J Obstet Gynecol Reprod Biol. 2009;147:157–160. doi: 10.1016/j.ejogrb.2009.08.007. [DOI] [PubMed] [Google Scholar]
  • 202.De Sousa Mendes M., Hirt D., Vinot C., et al. Prediction of human fetal pharmacokinetics using ex vivo human placenta perfusion studies and physiologically based models. Br J Clin Pharmacol. 2016;81:646–657. doi: 10.1111/bcp.12815. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Obstetrics & Gynecology Mfm are provided here courtesy of Elsevier

RESOURCES