, Déclaration sur la deuxième réunion du Comité d'urgence du Règlement sanitaire international, 2005.

P. Dashraath, J. Lin-jeslyn, W. , M. Xian-karen, L. et al., Coronavirus Disease 2019 (COVID-19) Pandemic and Pregnancy, Am J Obstet Gynecol, 2020.

S. A. Rasmussen, J. C. Smulian, J. A. Lednicky, T. S. Wen, and D. J. Jamieson, Coronavirus Disease 2019 (COVID-19) and Pregnancy: What obstetricians need to know, Am J Obstet Gynecol, 2020.

D. A. Schwartz and A. L. Graham, Potential Maternal and Infant Outcomes from (Wuhan) Coronavirus 2019-nCoV Infecting Pregnant Women: Lessons from SARS, MERS, and Other Human Coronavirus Infections, Viruses. 2020, vol.10, issue.2

A. M. Siston, S. A. Rasmussen, M. A. Honein, A. M. Fry, K. Seib et al., Pandemic 2009 influenza A(H1N1) virus illness among pregnant women in the United States, JAMA, vol.303, issue.15, pp.1517-1542, 2010.

C. Egloff, C. Vauloup-fellous, O. Picone, L. Mandelbrot, and P. Roques, Evidence and possible mechanisms of rare maternal-fetal transmission of SARS-CoV-2, J Clin Virol Off Publ Pan Am Soc Clin Virol, vol.128, p.104447, 2020.

H. Chen, J. Guo, C. Wang, F. Luo, X. Yu et al., Clinical characteristics and intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a retrospective review of medical records, Lancet Lond Engl, vol.395, pp.809-824, 10226.

D. A. Schwartz, 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.

S. Chen, B. Huang, D. J. Luo, X. Li, F. Yang et al.,

, Zhonghua Bing Li Xue Za Zhi, vol.49, issue.5, pp.418-441, 2020.

L. Dong, J. Tian, S. He, C. Zhu, J. Wang et al., Possible Vertical Transmission of SARS-CoV-2 From an Infected Mother to Her Newborn, JAMA, 2020.

H. Zeng, C. Xu, J. Fan, Y. Tang, Q. Deng et al., Antibodies in Infants Born to Mothers With COVID-19 Pneumonia, JAMA, 2020.

D. W. Kimberlin and S. Stagno, Can SARS-CoV-2 Infection Be Acquired In Utero?: More Definitive Evidence Is Needed, JAMA, 2020.

L. Zeng, S. Xia, W. Yuan, K. Yan, F. Xiao 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.

V. Peyronnet, J. Sibiude, P. Deruelle, C. Huissoud, X. Lescure et al.,

, Gynecol Obstet Fertil Senol, 2020.

S. Chen, E. Liao, and Y. Shao, Clinical analysis of pregnant women with 2019 novel coronavirus pneumonia, J Med Virol, 2020.

J. M. Sanders, M. L. Monogue, T. Z. Jodlowski, and J. B. Cutrell, Pharmacologic Treatments for Coronavirus Disease 2019 (COVID-19): A Review, JAMA, 2020.

, Covid-19 , Démarrage de l'essai clinique Discovery, 2020.

A. M. Henao-restrepo, V. Moorthy, and M. Preziosi, Public health emergency SOLIDARITY trial of treatments for COVID-19 infection in hospitalized patients

J. Sibiude, L. Mandelbrot, S. Blanche, L. Chenadec, J. Boullag-bonnet 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, vol.11, issue.4, p.1001635, 2014.

P. A. Tookey, C. Thorne, J. Van-wyk, and M. Norton, 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, vol.16, p.65, 2016.

K. Choy, A. Wong, P. Kaewpreedee, S. F. Sia, D. Chen et al., Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antiviral Res, vol.178, p.104786, 2020.

S. Jeon, M. Ko, J. Lee, I. Choi, S. Y. Byun et al., Identification of antiviral drug candidates against SARS-CoV-2 from FDA-approved drugs, Internet]. Microbiology, 2020.

F. Touret, M. Gilles, K. Barral, A. Nougairède, E. Decroly et al., In vitro screening of a FDA approved chemical library reveals potential inhibitors of SARS-CoV-2 replication [Internet]. Microbiology, 2020.

H. Retallack, D. Lullo, E. Arias, C. Knopp, K. A. Laurie et al., Zika virus cell tropism in the developing human brain and inhibition by azithromycin, Proc Natl Acad Sci, vol.113, issue.50, pp.14408-14421, 201613.

E. Bosseboeuf, A. M. Nhan, T. De-pina, J. J. Rolain, J. M. Raoult et al., Azithromycin Inhibits the Replication of Zika Virus, J Antivir Antiretrovir, p.2020, 2018.

P. B. Madrid, R. G. Panchal, T. K. Warren, A. C. Shurtleff, A. N. Endsley et al., Evaluation of Ebola Virus Inhibitors for Drug Repurposing, ACS Infect Dis, vol.1, issue.7, pp.317-343, 2015.

E. E. Essien and G. C. Afamefuna, Chloroquine and its metabolites in human cord blood, neonatal blood, and urine after maternal medication, Clin Chem, vol.28, issue.5, pp.1148-52, 1982.

I. Law, K. F. Ilett, L. P. Hackett, M. Page-sharp, F. Baiwog et al., Transfer of chloroquine and desethylchloroquine across the placenta and into milk in Melanesian mothers, Br J Clin Pharmacol, vol.65, issue.5, pp.674-683, 2008.

R. Mcgready, S. J. Lee, J. Wiladphaingern, E. A. Ashley, M. J. Rijken 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, vol.12, issue.5, pp.388-96, 2012.

A. Osadchy, T. Ratnapalan, and G. Koren, Ocular toxicity in children exposed in utero to antimalarial drugs: review of the literature, J Rheumatol, vol.38, issue.12, pp.2504-2512, 2011.

X. Yao, F. Ye, M. Zhang, C. Cui, B. Huang 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 Off Publ Infect Dis Soc Am, 2009.

J. Liu, R. Cao, M. Xu, X. Wang, H. Zhang et al., Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro, Cell Discov, vol.6, p.16, 2020.

W. Hong, Combating COVID-19 with Chloroquine, J Mol Cell Biol, 2020.

J. Chen, D. Liu, L. Liu, P. Liu, Q. Xu et al.,

Z. Chen, J. Hu, Z. Zhang, S. Jiang, S. Han et al., Efficacy of hydroxychloroquine in patients with COVID-19: results of a randomized clinical trial, Epidemiology, 2020.

W. Tang, Z. Cao, M. Han, Z. Wang, J. Chen et al., Hydroxychloroquine in patients mainly with mild to moderate COVID-19: an open-label, randomized, controlled trial, 2020.

J. M. Molina, C. Delaugerre, L. Goff, J. , M. -. Lima 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.

J. Geleris, Y. Sun, J. Platt, J. Zucker, M. Baldwin et al., Observational Study of Hydroxychloroquine in Hospitalized Patients with Covid-19, N Engl J Med, 2020.

M. Mahevas, V. Tran, M. Roumier, A. Chabrol, R. Paule 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.

,

J. Mallat, F. Hamed, M. Balkis, M. A. Mohamed, M. Mooty et al., Hydroxychloroquine is associated with slower viral clearance in clinical COVID-19 patients with mild to moderate disease: A retrospective study, 2009.

B. Yu, D. W. Wang, and C. Li, Hydroxychloroquine application is associated with a decreased mortality in critically ill patients with COVID-19, Emergency Medicine, 2020.

M. R. Mehra, S. S. Desai, F. Ruschitzka, and A. N. Patel, Hydroxychloroquine or chloroquine with or without a macrolide for treatment of COVID-19: a multinational registry analysis. The Lancet, 2020.

M. Million, J. Lagier, P. Gautret, P. Colson, P. Fournier 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.

, JORF n°0012 du 15 janvier 2020. Texte n°13. Arrêté du 13 janvier 2020 portant classement sur les listes des substances vénéneuses.rtf

A. Jorge, C. Ung, L. H. Young, R. B. Melles, and H. K. Choi, Hydroxychloroquine retinopathyimplications of research advances for rheumatology care, Nat Rev Rheumatol, vol.14, issue.12, pp.693-703, 2018.

K. L. Carter and D. V. Do, Hydroxychloroquine-induced Retinal Toxicity, J Rheumatol, vol.47, issue.4, p.632, 2020.

C. Chatre, F. Roubille, H. Vernhet, C. Jorgensen, and Y. Pers, Cardiac Complications Attributed to Chloroquine and Hydroxychloroquine: A Systematic Review of the Literature, Drug Saf, vol.41, issue.10, pp.919-950, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02336998

N. Costedoat-chalumeau, Z. Amoura, G. Aymard, T. Le, B. Wechsler et al., Evidence of transplacental passage of hydroxychloroquine in humans, Arthritis Rheum, vol.46, issue.4, pp.1123-1127, 2002.

N. Costedoat-chalumeau, Z. Amoura, P. Duhaut, D. Huong, D. Sebbough 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, vol.48, issue.11, pp.3207-3218, 2003.

F. Vroom, H. De-walle, M. Van-de-laar, J. Brouwers, . De-jong-van-den et al., Disease-modifying antirheumatic drugs in pregnancy: current status and implications for the future, Drug Saf, vol.29, issue.10, pp.845-63, 2006.

C. Abarientos, K. Sperber, D. L. Shapiro, W. S. Aronow, C. P. Chao et al., Hydroxychloroquine in systemic lupus erythematosus and rheumatoid arthritis and its safety in pregnancy, Expert Opin Drug Saf, vol.10, issue.5, pp.705-719, 2011.

R. Cimaz, A. Brucato, E. Meregalli, M. Muscará, and P. Sergi, 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, vol.50, pp.3057-3058, 2004.

T. Yao, J. Qian, W. Zhu, Y. Wang, and G. Wang, 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.

T. P. Sheahan, A. C. Sims, S. R. Leist, A. Schäfer, J. Won et al., Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV, Nat Commun, vol.11, issue.1, p.222, 2020.

B. Cao, Y. Wang, D. Wen, W. Liu, J. Wang et al., A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19, N Engl J Med, 2020.

A. Blanc, F. Bonnet, F. Brun-vezinet, D. Costagliola, F. Dabis et al., Prise en charge médicale des personnes vivants avec le VIH. Recommandations du groupe d'experts. Désir d'enfant et grossesse (mai 2018), p.52, 2018.

, European Pregnancy and Paediatric HIV Cohort Collaboration (EPPICC) Study Group. Nucleoside reverse transcriptase inhibitor backbones and pregnancy outcomes, AIDS Lond Engl, vol.33, issue.2, pp.295-304, 201901.

R. Tubiana, L. Mandelbrot, L. Chenadec, J. Delmas, S. Rouzioux 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 Off Publ Infect Dis Soc Am, vol.57, issue.6, pp.891-902, 2013.

F. Fauchet, J. Treluyer, S. M. Illamola, C. Pressiat, G. Lui 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, vol.59, issue.9, pp.5727-5762, 2015.

L. Gavard, S. Gil, G. Peytavin, P. Ceccaldi, C. Ferreira et al., Placental transfer of lopinavir/ritonavir in the ex vivo human cotyledon perfusion model, Am J Obstet Gynecol, vol.195, issue.1, pp.296-301, 2006.

J. Ivanovic, E. Nicastri, M. M. Anceschi, P. Ascenzi, F. Signore et al., Transplacental transfer of antiretroviral drugs and newborn birth weight in HIV-infected pregnant women, Curr HIV Res, vol.7, issue.6, pp.620-625, 2009.

C. Marzolini, C. Rudin, L. A. Decosterd, A. Telenti, A. Schreyer et al., Transplacental passage of protease inhibitors at delivery, AIDS Lond Engl, vol.16, issue.6, pp.889-93, 2002.

T. R. Cressey, B. M. Best, J. Achalapong, A. Stek, J. Wang et al., Reduced indinavir exposure during pregnancy, Br J Clin Pharmacol, vol.76, issue.3, pp.475-83, 2013.

P. Ceccaldi, L. Gavard, L. Mandelbrot, E. Rey, R. Farinotti 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, p.726593, 2009.

L. Mandelbrot, D. Duro, E. Belissa, and G. Peytavin, Placental transfer of darunavir in an ex vivo human cotyledon perfusion model, Antimicrob Agents Chemother, vol.58, issue.9, pp.5617-5637, 2014.

M. G. Fowler, M. Qin, S. A. Fiscus, J. S. Currier, P. M. Flynn et al., Benefits and Risks of Antiretroviral Therapy for Perinatal HIV Prevention, N Engl J Med, vol.375, issue.18, pp.1726-1763, 201603.

K. M. Powis, D. Kitch, A. Ogwu, M. D. Hughes, S. Lockman 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, vol.204, issue.4, pp.506-520, 2011.

A. Simon, J. Warszawski, D. Kariyawasam, L. Chenadec, J. Benhammou et al., Association of prenatal and postnatal exposure to lopinavir-ritonavir and adrenal dysfunction among uninfected infants of HIV-infected mothers, JAMA, vol.306, issue.1, pp.70-78, 2011.

E. P. Tchesnokov, J. Y. Feng, D. P. Porter, and M. Götte, Mechanism of Inhibition of Ebola Virus RNA-Dependent RNA Polymerase by Remdesivir, Viruses, vol.11, issue.4, 201904.

S. Mulangu, L. E. Dodd, R. T. Davey, T. Mbaya, O. Proschan et al., Controlled Trial of Ebola Virus Disease Therapeutics, N Engl J Med, vol.12, issue.24, pp.2293-303, 2019.

T. P. Sheahan, A. C. Sims, R. L. Graham, V. D. Menachery, L. E. Gralinski et al., Broadspectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses, Sci Transl Med, vol.9, issue.396, 201728.

A. J. Brown, J. J. Won, R. L. Graham, K. H. Dinnon, A. C. Sims et al., Broad spectrum antiviral remdesivir inhibits human endemic and zoonotic deltacoronaviruses with a highly divergent RNA dependent RNA polymerase, Antiviral Res, vol.169, p.104541, 2019.

E. De-wit, F. Feldmann, J. Cronin, R. Jordan, A. Okumura et al., Prophylactic and therapeutic remdesivir (GS-5734) treatment in the rhesus macaque model of MERS-CoV infection, Proc Natl Acad Sci, vol.117, issue.12, pp.6771-6777, 2020.

C. J. Gordon, E. P. Tchesnokov, J. Y. Feng, D. P. Porter, and M. Gotte, The antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus, J Biol Chem, 2020.

J. H. Beigel, K. M. Tomashek, L. E. Dodd, A. K. Mehta, B. S. Zingman et al., Remdesivir for the Treatment of Covid-19 -Preliminary Report, N Engl J Med, 2020.

Y. Wang, D. Zhang, G. Du, R. Du, J. Zhao et al., Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. The Lancet, vol.395, pp.1569-78, 2020.

J. Grein, N. Ohmagari, D. Shin, G. Diaz, E. Asperges et al., Compassionate Use of Remdesivir for Patients with Severe Covid-19, N Engl J Med, 2020.

. Ema/, Assessment report on Medicinal products under development for the treatment of Ebola, 2016.

, Avis relatif à la prise en charge des cas confirmés d'infection au virus SARS-CoV-2, p.33, 2020.

, Avis relatif à la prise en charge des cas confirmés d'infection au virus SARS-CoV-2, vol.33, 2020.

Y. Jin, H. Yang, J. W. Wu, W. Chen, S. Zhang et al., Epidemiology, Pathogenesis, and Control of COVID-19, Viruses, vol.12, issue.4, 202027.

D. Meyer, S. Bojkova, D. Cinati, J. Van-damme, E. Buyck et al., Lack of Antiviral Activity of Darunavir against SARS-CoV-2 [Internet]. Infectious Diseases (except HIV/AIDS), 2020.

H. Huang, J. Wang, Q. Li, J. Duan, Q. Yao 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 Off J Eur Assoc Perinat Med Fed Asia Ocean Perinat Soc Int Soc Perinat Obstet, vol.30, issue.11, pp.1288-92, 2017.

C. D. Chambers, D. Johnson, R. Xu, Y. Luo, K. L. Jones et al., Oseltamivir use in pregnancy: Risk of birth defects, preterm delivery, and small for gestational age infants, Birth Defects Res, vol.111, issue.19, pp.1487-93, 201915.

S. L. Bixler, T. M. Bocan, J. Wells, K. S. Wetzel, S. A. Van-tongeren et al., Efficacy of favipiravir (T-705) in nonhuman primates infected with Ebola virus or Marburg virus, Antiviral Res, vol.151, pp.97-104, 2018.

L. Delang, R. Abdelnabi, and J. Neyts, Favipiravir as a potential countermeasure against neglected and emerging RNA viruses, Antiviral Res, vol.153, pp.85-94, 2018.

Y. Furuta, T. Komeno, and T. Nakamura, Favipiravir (T-705), a broad spectrum inhibitor of viral RNA polymerase, Proc Jpn Acad Ser B Phys Biol Sci, vol.93, issue.7, pp.449-63, 2017.

L. Oestereich, A. Lüdtke, S. Wurr, T. Rieger, C. Muñoz-fontela et al., Successful treatment of advanced Ebola virus infection with T-705 (favipiravir) in a small animal model, Antiviral Res, vol.105, pp.17-21, 2014.

R. Kerber, E. Lorenz, S. Duraffour, D. Sissoko, M. Rudolf 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, vol.220, issue.2, pp.195-202, 201919.

M. Baz, J. Carbonneau, C. Rhéaume, M. Cavanagh, and G. Boivin, 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, vol.10, 201803.

Y. Wang, G. Fan, A. Salam, P. Horby, F. G. Hayden et al., Comparative effectiveness of combined favipiravir and oseltamivir therapy versus oseltamivir monotherapy in critically ill patients with influenza virus infection, J Infect Dis, 2019.

F. G. Hayden and N. Shindo, Influenza virus polymerase inhibitors in clinical development, Curr Opin Infect Dis, vol.32, issue.2, pp.176-86, 2019.

A. S. Omrani, M. M. Saad, K. Baig, A. Bahloul, M. Abdul-matin et al., Ribavirin and interferon alfa-2a for severe Middle East respiratory syndrome coronavirus infection: a retrospective cohort study, Lancet Infect Dis, vol.14, issue.11, pp.1090-1095, 2014.

Y. M. Arabi, S. Shalhoub, Y. Mandourah, F. Al-hameed, A. et al., Ribavirin and Interferon Therapy for Critically Ill Patients With Middle East Respiratory Syndrome: A Multicenter Observational Study, Clin Infect Dis Off Publ Infect Dis Soc Am, 2019.

S. S. Roberts, R. K. Miller, J. K. Jones, K. L. Lindsay, M. F. Greene et al., The Ribavirin Pregnancy Registry: Findings after 5 years of enrollment, Birt Defects Res A Clin Mol Teratol, vol.88, issue.7, pp.551-560, 2003.

J. Sibiude, J. Warszawski, R. Tubiana, C. Dollfus, F. A. Rouzioux et al., Premature delivery in HIV-infected women starting protease inhibitor therapy during pregnancy: role of the ritonavir boost?, vol.54, pp.1348-60, 2012.

N. Yamamoto, R. Yang, Y. Yoshinaka, S. Amari, T. Nakano et al., HIV protease inhibitor nelfinavir inhibits replication of SARS-associated coronavirus, Biochem Biophys Res Commun, vol.318, issue.3, pp.719-744, 2004.

R. A. Khamitov, S. I. Loginova, V. N. Shchukina, S. V. Borisevich, V. A. Maksimov et al.,

, Vopr Virusol, vol.53, issue.4, pp.9-13, 2008.

L. Deng, C. Li, Q. Zeng, X. Liu, X. Li et al., Arbidol combined with LPV/r versus LPV/r alone against Corona Virus Disease 2019: A retrospective cohort study, J Infect, 2020.

N. Lian, H. Xie, S. Lin, J. Huang, J. Zhao et al., Umifenovir treatment is not associated with improved outcomes in patients with coronavirus disease 2019: a retrospective study, Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis, 2020.

N. Yu, W. Li, Q. Kang, Z. Xiong, S. Wang 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.

R. Pérez, C. Palma, M. J. Núñez, and J. Cox, Pharmacokinetics of ivermectin after maternal or fetal intravenous administration in sheep, J Vet Pharmacol Ther, vol.31, issue.5, pp.406-420, 2008.

P. Nicolas, M. F. Maia, Q. Bassat, K. C. Kobylinski, W. Monteiro et al., Safety of oral ivermectin during pregnancy: a systematic review and meta-analysis. Lancet Glob Health, vol.8, pp.92-100, 2020.

, Highlights of prescribing information, FDA.pdf, AVONEX

P. K. Coyle, S. M. Sinclair, A. E. Scheuerle, J. M. Thorp, J. D. Albano et al., Final results from the Betaseron (interferon ?-1b) Pregnancy Registry: a prospective observational study of birth defects and pregnancy-related adverse events, BMJ Open, vol.4, issue.5, p.4536, 2014.

K. Hellwig, Y. Geissbuehler, M. Sabidó, C. Popescu, A. Adamo et al., Pregnancy outcomes in interferon-beta-exposed patients with multiple sclerosis: results from the European Interferon-beta Pregnancy Registry, J Neurol, 2020.

K. Hellwig, D. Caron, F. Wicklein, E. Bhatti, A. Adamo et al., Pregnancy outcomes from the global pharmacovigilance database on interferon beta-1b exposure, Ther Adv Neurol Disord, vol.13, p.1756286420910310, 2020.

A. Waysbort, M. Giroux, V. Mansat, M. Teixeira, J. C. Dumas et al., Experimental study of transplacental passage of alpha interferon by two assay techniques, Antimicrob Agents Chemother, vol.37, issue.6, pp.1232-1239, 1993.

J. C. Dumas, M. Giroux, M. G. Teixeira, J. Puel, A. Waysbort et al.,

, Therapie, vol.48, issue.1, pp.73-78, 1993.

J. C. Pons, P. Lebon, R. Frydman, and J. F. Delfraissy, Pharmacokinetics of interferon-alpha in pregnant women and fetoplacental passage, Fetal Diagn Ther, vol.10, issue.1, pp.7-10, 1995.

H. K. Wong and C. K. Lee, Pivotal Role of Convalescent Plasma in Managing Emerging Infectious Diseases. Vox Sang, 2020.

G. Marano, S. Vaglio, S. Pupella, G. Facco, L. Catalano et al., Convalescent plasma: new evidence for an old therapeutic tool? Blood Transfus Trasfus Sangue, vol.14, pp.152-159, 2016.

J. Van-griensven, T. Edwards, X. De-lamballerie, M. G. Semple, P. Gallian et al., Evaluation of Convalescent Plasma for Ebola Virus Disease in Guinea, N Engl J Med, vol.374, issue.1, pp.33-42, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01252702

Y. Cheng, R. Wong, Y. Soo, W. S. Wong, C. K. Lee et al., Use of convalescent plasma therapy in SARS patients in Hong Kong, Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol, vol.24, issue.1, pp.44-50, 2005.

B. Zhang, S. Liu, T. Tan, W. Huang, Y. Dong et al., Treatment with convalescent plasma for critically ill patients with SARS-CoV-2 infection, Chest, 2020.

N. E. Simister, Placental transport of immunoglobulin G. Vaccine, vol.21, pp.3365-3374, 2003.

R. Channappanavar and S. Perlman, Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology, Semin Immunopathol, vol.39, issue.5, pp.529-568, 2017.

Y. Tada, M. Sakai, Y. Nakao, A. Maruyama, N. Ono et al., Placental transfer of tocilizumab in a patient with rheumatoid arthritis, Rheumatol Oxf Engl, vol.58, issue.9, pp.1694-1699, 201901.

J. Saito, N. Yakuwa, C. Takai, K. Nakajima, K. Kaneko et al., Tocilizumab concentrations in maternal serum and breast milk during breastfeeding and a safety assessment in infants: a case study, Rheumatology, vol.57, issue.8, pp.1499-501, 2018.

K. Nakajima, O. Watanabe, M. Mochizuki, A. Nakasone, N. Ishizuka et al., Pregnancy outcomes after exposure to tocilizumab: A retrospective analysis of 61 patients in Japan, Mod Rheumatol, vol.26, issue.5, pp.667-71, 2016.

M. Hoeltzenbein, E. Beck, R. Rajwanshi, G. Skorpen, C. Berber et al., Tocilizumab use in pregnancy: Analysis of a global safety database including data from clinical trials and post-marketing data, Semin Arthritis Rheum, vol.46, issue.2, pp.238-283, 2016.

C. Götestam-skorpen, M. Hoeltzenbein, A. Tincani, R. Fischer-betz, E. Elefant et al., The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation, Ann Rheum Dis, vol.75, issue.5, pp.795-810, 2016.

T. Youngstein, P. Hoffmann, A. Gül, T. Lane, R. Williams et al., International multi-centre study of pregnancy outcomes with interleukin-1 inhibitors. Rheumatol Oxf Engl, vol.56, pp.2102-2110, 201701.

G. Comi, H. Hartung, R. Bakshi, I. M. Williams, and H. Wiendl, Benefit-Risk Profile of Sphingosine-1-Phosphate Receptor Modulators in Relapsing and Secondary Progressive Multiple Sclerosis, Drugs, vol.77, issue.16, pp.1755-68, 2017.

T. Saida, Y. Itoyama, S. Kikuchi, Q. Hao, T. Kurosawa 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, vol.17, issue.1, p.17, 2017.

Y. Geissbühler, J. Vile, G. Koren, M. Guennec, H. Butzkueven et al., Evaluation of pregnancy outcomes in patients with multiple sclerosis after fingolimod exposure, Ther Adv Neurol Disord, vol.11, p.1756286418804760, 2018.

Z. T. Al-salama and L. J. Scott, Baricitinib: A Review in Rheumatoid Arthritis, Drugs, vol.78, issue.7, pp.761-72, 2018.

G. Costanzo, D. Firinu, F. Losa, M. Deidda, M. P. Barca et al., Baricitinib exposure during pregnancy in rheumatoid arthritis, Ther Adv Musculoskelet Dis, vol.12, pp.1759720-19899296, 2020.

M. Gerosa, L. M. Argolini, C. Artusi, and C. B. Chighizola, The use of biologics and small molecules in pregnant patients with rheumatic diseases, Expert Rev Clin Pharmacol, vol.11, issue.10, pp.987-98, 2018.

U. Mahadevan, S. Kane, W. J. Sandborn, R. D. Cohen, K. Hanson et al., Intentional infliximab use during pregnancy for induction or maintenance of remission in Crohn's disease, Aliment Pharmacol Ther, vol.21, issue.6, pp.733-741, 2005.

E. A. Vasiliauskas, J. A. Church, N. Silverman, M. Barry, S. R. Targan et al., Case report: evidence for transplacental transfer of maternally administered infliximab to the newborn, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc, vol.4, issue.10, pp.1255-1263, 2006.

C. Steenholdt, M. Al-khalaf, M. A. Ainsworth, and J. Brynskov, Therapeutic infliximab drug level in a child born to a woman with ulcerative colitis treated until gestation week 31, J Crohns Colitis, vol.6, issue.3, pp.358-61, 2012.

M. Cumhur-cure, A. Kucuk, and E. Cure, Colchicine may not be effective in COVID-19 infection; it may even be harmful?, Clin Rheumatol, 2020.

O. Gendelman, H. Amital, N. L. Bragazzi, A. Watad, and G. Chodick, Continuous hydroxychloroquine or colchicine therapy does not prevent infection with SARS-CoV-2: Insights from a large healthcare database analysis, Autoimmun Rev, 2020.

S. G. Deftereos, G. Siasos, G. Giannopoulos, D. A. Vrachatis, C. Angelidis 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 Kardiologike Epitheorese, 2020.

Z. Amoura, J. M. Schermann, B. Wechsler, X. Zerah, and P. Goodeau, Transplacental passage of colchicine in familial Mediterranean fever, J Rheumatol, vol.21, issue.2, p.383, 1994.

P. L. Indraratna, S. Virk, D. Gurram, and R. O. Day, Use of colchicine in pregnancy: a systematic review and meta-analysis, Rheumatol Oxf Engl, vol.57, issue.2, pp.382-389, 2018.

O. Diav-citrin, S. Shechtman, V. Schwartz, M. Avgil-tsadok, V. Finkel-pekarsky et al., Pregnancy outcome after in utero exposure to colchicine, Am J Obstet Gynecol, vol.203, issue.2, pp.144-145, 2010.

M. C. Petersen, R. L. Nation, J. J. Ashley, and W. G. Mcbride, The placental transfer of betamethasone, Eur J Clin Pharmacol, vol.18, issue.3, pp.245-252, 1980.

R. M. Ward, Drug therapy of the fetus, J Clin Pharmacol, vol.33, issue.9, pp.780-789, 1993.

K. Yang, Placental 11 beta-hydroxysteroid dehydrogenase: barrier to maternal glucocorticoids, Rev Reprod, vol.2, issue.3, pp.129-161, 1997.

P. Crowley, Prophylactic corticosteroids for preterm birth, Cochrane Database Syst Rev, issue.2, p.65, 2000.

J. H. Chow, M. A. Mazzeffi, and M. T. Mccurdy, Angiotensin II for the Treatment of COVID-19-Related Vasodilatory Shock, Anesth Analg, 2020.

J. H. Kuhn, W. Li, H. Choe, and M. Farzan, Angiotensin-converting enzyme 2: a functional receptor for SARS coronavirus, Cell Mol Life Sci CMLS, vol.61, issue.21, pp.2738-2781, 2004.

M. Vaduganathan, O. Vardeny, T. Michel, J. Mcmurray, M. A. Pfeffer et al., Renin-Angiotensin-Aldosterone System Inhibitors in Patients with Covid-19, N Engl J Med, 2020.

S. Alwan, J. E. Polifka, and J. M. Friedman, Angiotensin II receptor antagonist treatment during pregnancy, Birt Defects Res A Clin Mol Teratol, vol.73, issue.2, pp.123-153, 2005.

S. Nadeem, S. Hashmat, M. J. Defreitas, K. D. Westreich, I. F. Shatat et al., Renin Angiotensin System Blocker Fetopathy: A Midwest Pediatric Nephrology Consortium Report, J Pediatr, vol.167, issue.4, pp.881-886, 2015.

S. J. Delforce, E. R. Lumbers, S. J. Ellery, P. Murthi, and K. G. Pringle, Dysregulation of the placental renin-angiotensin system in human fetal growth restriction, Reprod Camb Engl, vol.158, issue.3, pp.237-282, 2019.

L. Shang, Y. Wang, and Y. Lyu, Clinical effect of mifepristone on patients with ovarian cancer in pregnancy, Pak J Pharm Sci, vol.32, issue.1, pp.421-427, 2019.

T. Korenaga and K. S. Tewari, Gynecologic cancer in pregnancy, Gynecol Oncol, 2020.

S. Polizzi and V. B. Mahajan, Intravitreal Anti-VEGF Injections in Pregnancy: Case Series and Review of Literature, J Ocul Pharmacol Ther Off J Assoc Ocul Pharmacol Ther, vol.31, issue.10, pp.605-615, 2015.

R. M. Tarantola, J. C. Folk, H. C. Boldt, and V. B. Mahajan, Intravitreal bevacizumab during pregnancy. Retina Phila Pa, vol.30, pp.1405-1416, 2010.

W. Lenz, A short history of thalidomide embryopathy, Teratology, vol.38, issue.3, pp.203-218, 1988.

G. W. Mellin and M. Katzenstein, The saga of thalidomide. Neuropathy to embryopathy, with case reports of congenital anomalies, N Engl J Med, vol.267, pp.1184-1192, 1962.

I. Nobuhara, N. Harada, N. Haruta, Y. Higashiura, H. Watanabe 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, vol.57, issue.4, pp.588-93, 2018.

K. Zarchi, M. Behtash, N. , M. Gilani, and 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, vol.277, issue.1, pp.75-83, 2008.

H. Ngan, K. Tam, K. Lam, and K. Chan, Methotrexate, bleomycin, and Etoposide in the treatment of gestational trophoblastic neoplasia, Obstet Gynecol, vol.107, issue.5, pp.1012-1019, 2006.

Y. Song, Z. Wang, Z. Hao, L. Li, J. Lu et al., Requirement for etoposide in the treatment of pregnancy related hemophagocytic lymphohistiocytosis: a multicenter retrospective study, Orphanet J Rare Dis, vol.14, issue.1, p.50, 201918.

J. Sibiude, J. Warszawski, R. Tubiana, L. Chenadec, J. Meier et al., Liver Enzyme Elevation in Pregnant Women Receiving Antiretroviral Therapy in the ANRS-French Perinatal Cohort, J Acquir Immune Defic Syndr, vol.81, issue.1, pp.83-94, 1999.
URL : https://hal.archives-ouvertes.fr/inserm-02070249

D. Matsui, Ethics of studies of drugs in pregnancy, Paediatr Drugs, vol.17, issue.1, pp.31-36, 2015.

K. Heyrana, H. M. Byers, and P. Stratton, Increasing the Participation of Pregnant Women in Clinical Trials, JAMA, vol.320, issue.20, pp.2077-2085, 201827.

R. G. Beran, The ethics of excluding women who become pregnant while participating in clinical trials of anti-epileptic medications, Seizure, vol.15, issue.8, pp.563-70, 2006.

D. K. Kaye, The moral imperative to approve pregnant women's participation in randomized clinical trials for pregnancy and newborn complications, Philos Ethics Humanit Med PEHM, vol.14, issue.1, p.11, 201906.

P. Bourget, C. Roulot, and H. Fernandez, Models for Placental Transfer Studies of Drugs: Clin Pharmacokinet, vol.28, pp.161-80, 1995.

A. Dallmann, X. I. Liu, G. J. Burckart, and J. Van-den-anker, Drug Transporters Expressed in the Human Placenta and Models for Studying Maternal-Fetal Drug Transfer, J Clin Pharmacol, vol.59, issue.1, pp.70-81, 2019.

V. Faure-bardon, L. Mandelbrot, D. Duro, C. Dussaux, M. Le et al., Placental transfer of elvitegravir and cobicistat in an ex-vivo human cotyledon double perfusion model, AIDS Lond Engl, vol.32, issue.3, pp.321-326, 201828.

L. Gavard, D. Beghin, F. Forestier, Y. Cayre, G. Peytavin 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, vol.147, issue.2, pp.157-60, 2009.

D. Sousa-mendes, M. Hirt, D. Vinot, C. Valade, E. Lui et al., Prediction of human fetal pharmacokinetics using ex vivo human placenta perfusion studies and physiologically based models, Br J Clin Pharmacol, vol.81, issue.4, pp.646-57, 2016.

A. Bérard, J. Zhao, I. Shui, and S. Colilla, Leflunomide use during pregnancy and the risk of adverse pregnancy outcomes, Ann Rheum Dis, vol.77, issue.4, pp.500-509, 2018.

V. Ehrenstein, N. R. Kristensen, B. U. Monz, B. Clinch, A. Kenwright et al., Oseltamivir in pregnancy and birth outcomes, BMC Infect Dis, vol.18, issue.1, p.519, 2018.

S. M. Sinclair, J. K. Jones, R. K. Miller, M. F. Greene, P. Y. Kwo et al., The Ribavirin Pregnancy Registry: An Interim Analysis of Potential Teratogenicity at the Mid-Point of Enrollment, Drug Saf, vol.40, issue.12, pp.1205-1223, 2017.

F. M. Russo, S. Conings, K. Allegaert, T. Van-mieghem, J. Toelen et al., Sildenafil crosses the placenta at therapeutic levels in a dually perfused human cotyledon model, Am J Obstet Gynecol, vol.219, issue.6, pp.619-620, 2018.

L. Dunn, R. Greer, V. Flenady, and S. Kumar, Sildenafil in Pregnancy: A Systematic Review of Maternal Tolerance and Obstetric and Perinatal Outcomes, Fetal Diagn Ther, vol.41, issue.2, pp.81-89, 2017.

F. M. Russo, A. Benachi, T. Van-mieghem, D. Hoon, J. Van-calsteren 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, vol.19, issue.1, p.524, 2018.

S. Ferreira-rd-da, R. Negrini, W. M. Bernardo, R. Simões, and S. Piato, The effects of sildenafil in maternal and fetal outcomes in pregnancy: A systematic review and meta-analysis, PloS One, vol.14, issue.7, p.219732, 2019.