[Skip to Navigation]
Sign In
Figure.  Simplified Representation of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Viral Lifecycle and Potential Drug Targets
Simplified Representation of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Viral Lifecycle and Potential Drug Targets

Schematic represents virus-induced host immune system response and viral processing within target cells. Proposed targets of select repurposed and investigational products are noted. ACE2, angiotensin-converting enzyme 2; S protein, spike protein; and TMPRSS2, type 2 transmembrane serine protease.

Table 1.  Summary of Pharmacology for Select Proposed COVID-19 Treatments
Summary of Pharmacology for Select Proposed COVID-19 Treatments
Table 2.  Summary of Treatment and Clinical Outcomes From Early COVID-19 Clinical Series
Summary of Treatment and Clinical Outcomes From Early COVID-19 Clinical Series
1.
Zhu  N, Zhang  D, Wang  W,  et al; China Novel Coronavirus Investigating and Research Team.  A novel coronavirus from patients with pneumonia in China, 2019.   N Engl J Med. 2020;382(8):727-733. doi:10.1056/NEJMoa2001017 PubMedGoogle ScholarCrossref
2.
Chinese Clinical Trials. http://www/chictr.org/enindex.aspx. Accessed March 31, 2020.
3.
Hoffmann  M, Kleine-Weber  H, Schroeder  S,  et al.  SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor.   Cell. Published online March 4, 2020. doi:10.1016/j.cell.2020.02.052 PubMedGoogle Scholar
4.
Chen  Y, Liu  Q, Guo  D.  Emerging coronaviruses: genome structure, replication, and pathogenesis.   J Med Virol. 2020;92(4):418-423. doi:10.1002/jmv.25681 PubMedGoogle ScholarCrossref
5.
Fehr  AR, Perlman  S.  Coronaviruses: an overview of their replication and pathogenesis.   Methods Mol Biol. 2015;1282:1-23. doi:10.1007/978-1-4939-2438-7_1 PubMedGoogle ScholarCrossref
6.
Fung  TS, Liu  DX.  Coronavirus infection, ER stress, apoptosis and innate immunity.   Front Microbiol. 2014;5:296. doi:10.3389/fmicb.2014.00296 PubMedGoogle ScholarCrossref
7.
Savarino  A, Boelaert  JR, Cassone  A, Majori  G, Cauda  R.  Effects of chloroquine on viral infections: an old drug against today’s diseases?   Lancet Infect Dis. 2003;3(11):722-727. doi:10.1016/S1473-3099(03)00806-5 PubMedGoogle ScholarCrossref
8.
Al-Bari  MAA.  Targeting endosomal acidification by chloroquine analogs as a promising strategy for the treatment of emerging viral diseases.   Pharmacol Res Perspect. 2017;5(1):e00293. doi:10.1002/prp2.293 PubMedGoogle Scholar
9.
Zhou  D, Dai  SM, Tong  Q.  COVID-19: a recommendation to examine the effect of hydroxychloroquine in preventing infection and progression.  [published online March 20, 2020].  J Antimicrob Chemother. 2020;dkaa114. doi:10.1093/jac/dkaa114 PubMedGoogle Scholar
10.
Devaux  CA, Rolain  JM, Colson  P, Raoult  D.  New insights on the antiviral effects of chloroquine against coronavirus: what to expect for COVID-19?   Int J Antimicrob Agents. Published online March 11, 2020. doi:10.1016/j.ijantimicag.2020.105938 PubMedGoogle Scholar
11.
Colson  P, Rolain  JM, Lagier  JC, Brouqui  P, Raoult  D.  Chloroquine and hydroxychloroquine as available weapons to fight COVID-19.   Int J Antimicrob Agents. Published online March 4, 2020. doi:10.1016/j.ijantimicag.2020.105932 PubMedGoogle Scholar
12.
National Health Commission and State Administration of Traditional Chinese Medicine. Diagnosis and treatment protocol for novel coronavirus pneumonia. Accessed March 18, 2020. https://www.chinalawtranslate.com/wp-content/uploads/2020/03/Who-translation.pdf
13.
Chloroquine [database online]. Hudson, OH: Lexicomp Inc; 2016. Accessed March 17, 2020. http://online.lexi.com
14.
Aralen (chloroquine phosphate) [package insert]. Bridgewater, NJ: Sanofi-Aventis; 2008. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/006002s045lbl.pdf
15.
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. Published online March 9, 2020. doi:10.1093/cid/ciaa237 PubMedGoogle Scholar
16.
Gautret  P, Lagier  JC, 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. Published online March 20, 2020. doi:10.1016/j.ijantimicag.2020.105949 PubMedGoogle Scholar
17.
Chen  J, Liu  D, Liu  L,  et al.  A pilot study of hydroxychloroquine in treatment of patients with common coronavirus disease-19 (COVID-19).   J Zhejiang Univ (Med Sci). Published online March 6, 2020. doi:10.3785/j.issn.1008-9292.2020.03.03Google Scholar
18.
Hydroxychloroquine [database online]. Hudson, OH: Lexicomp Inc; 2016. Accessed March 17, 2020. http://online.lexi.com
19.
Plaquenil (Hydroxychloroquine sulfate) [package insert]. St Michael, Barbados: Concordia Pharmaceuticals Inc; 2018. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/009768Orig1s051lbl.pdf
20.
Lim  HS, Im  JS, Cho  JY,  et al.  Pharmacokinetics of hydroxychloroquine and its clinical implications in chemoprophylaxis against malaria caused by Plasmodium vivax.   Antimicrob Agents Chemother. 2009;53(4):1468-1475. doi:10.1128/AAC.00339-08 PubMedGoogle ScholarCrossref
21.
Chu  CM, Cheng  VC, Hung  IF,  et al; HKU/UCH SARS Study Group.  Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings.   Thorax. 2004;59(3):252-256. doi:10.1136/thorax.2003.012658 PubMedGoogle ScholarCrossref
22.
de Wilde  AH, Jochmans  D, Posthuma  CC,  et al.  Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture.   Antimicrob Agents Chemother. 2014;58(8):4875-4884. doi:10.1128/AAC.03011-14 PubMedGoogle ScholarCrossref
23.
Cao  B, Wang  Y, Wen  D,  et al.  A trial of lopinavir-ritonavir in adults hospitalized with severe COVID-19.   N Engl J Med. Published online March 18, 2020. doi:10.1056/NEJMoa2001282 PubMedGoogle Scholar
24.
Lopinavir/ritonavir [database online]. Hudson (OH): Lexicomp Inc; 2016. Accessed March 17, 2020. http://online.lexi.com
25.
Kaletra (Lopinavir and ritonavir) [package insert]. North Chicago, IL: Abbvie; 2019. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021226s048lbl.pdf
26.
Department of Health and Human Services Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Accessed March 17, 2020. http://www.aidsinfo.nih.gov/ContentFiles/ AdultandAdolescentGL.pdf
27.
Kadam  RU, Wilson  IA.  Structural basis of influenza virus fusion inhibition by the antiviral drug Arbidol.   Proc Natl Acad Sci U S A. 2017;114(2):206-214. doi:10.1073/pnas.1617020114 PubMedGoogle ScholarCrossref
28.
Khamitov  RA, Loginova  SIa, Shchukina  VN, Borisevich  SV, Maksimov  VA, Shuster  AM.  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(4):9-13.PubMedGoogle Scholar
29.
Wang  Z, Yang  B, Li  Q, Wen  L, Zhang  R.  Clinical Features of 69 cases with coronavirus disease 2019 in Wuhan, China.   Clin Infect Dis. Published online March 16, 2020. doi:10.1093/cid/ciaa272 PubMedGoogle Scholar
30.
Siegel  D, Hui  HC, Doerffler  E,  et al.  Discovery and synthesis of a phosphoramidate prodrug of a pyrrolo[2,1-f][triazin-4-amino] adenine C-nucleoside (GS-5734) for the treatment of Ebola and emerging viruses.   J Med Chem. 2017;60(5):1648-1661. doi:10.1021/acs.jmedchem.6b01594 PubMedGoogle ScholarCrossref
31.
Al-Tawfiq  JA, Al-Homoud  AH, Memish  ZA.  Remdesivir as a possible therapeutic option for the COVID-19.   Travel Med Infect Dis. Published online March 5, 2020. doi:10.1016/j.tmaid.2020.101615 PubMedGoogle Scholar
32.
Sheahan  TP, Sims  AC, Leist  SR,  et al.  Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV.   Nat Commun. 2020;11(1):222. doi:10.1038/s41467-019-13940-6 PubMedGoogle ScholarCrossref
33.
Hayden  FG, Shindo  N.  Influenza virus polymerase inhibitors in clinical development.   Curr Opin Infect Dis. 2019;32(2):176-186. doi:10.1097/QCO.0000000000000532 PubMedGoogle ScholarCrossref
34.
Avigan (favipiravir) [package insert]. Tokyo, Japan: Taisho Toyama Pharmaceutical Co Ltd; 2017, 4th version. Accessed March 25, 2020.
35.
Xu X, Han M, Li T, et al. Effective treatment of severe COVID-19 patients with tocilizumab. chinaXiv. Preprint posted March 5, 2020. doi:10.12074/202003.00026
36.
Actemra (tocilizumab) [package insert]. South San Francisco, CA: Genentech, Inc; 2019. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/125276s127,125472s040lbl.pdf
37.
Stockman  LJ, Bellamy  R, Garner  P.  SARS: systematic review of treatment effects.   PLoS Med. 2006;3(9):e343. doi:10.1371/journal.pmed.0030343 PubMedGoogle Scholar
38.
Morra  ME, Van Thanh  L, Kamel  MG,  et al.  Clinical outcomes of current medical approaches for Middle East respiratory syndrome: a systematic review and meta-analysis.   Rev Med Virol. 2018;28(3):e1977. doi:10.1002/rmv.1977 PubMedGoogle Scholar
39.
Gao  J, Tian  Z, Yang  X.  Breakthrough: chloroquine phosphate has shown apparent efficacy in treatment of COVID-19 associated pneumonia in clinical studies.   Biosci Trends. 2020;14(1):72-73. doi:10.5582/bst.2020.01047 PubMedGoogle ScholarCrossref
40.
ClinicalTrials.gov. Accessed March 18, 2020. https://clinicaltrials.gov/
41.
Kalil  AC.  Treating COVID-19—off-label drug use, compassionate use, and randomized clinical trials during pandemics.   JAMA. Published March 24, 2020. doi:10.1001/jama.2020.4742 PubMedGoogle Scholar
42.
Interview with David Juurlink.  Coronavirus (COVID-19) update: chloroquine/hydroxychloroquine and azithromycin.   JAMA. March 24, 2020. Accessed April 3, 2020. https://edhub.ama-assn.org/jn-learning/audio-player/18337225Google Scholar
43.
Osadchy  A, Ratnapalan  T, Koren  G.  Ocular toxicity in children exposed in utero to antimalarial drugs: review of the literature.   J Rheumatol. 2011;38(12):2504-2508. doi:10.3899/jrheum.110686 PubMedGoogle ScholarCrossref
44.
Dong  L, Hu  S, Gao  J.  Discovering drugs to treat coronavirus disease 2019 (COVID-19).   Drug Discov Ther. 2020;14(1):58-60. doi:10.5582/ddt.2020.01012 PubMedGoogle ScholarCrossref
45.
Yao  TT, Qian  JD, Zhu  WY, Wang  Y, Wang  GQ.  A systematic review of lopinavir therapy for SARS coronavirus and MERS coronavirus-A possible reference for coronavirus disease-19 treatment option.  [published online February 27, 2020].  J Med Virol. 2020. doi:10.1002/jmv.25729 PubMedGoogle Scholar
46.
Chan  KS, Lai  ST, Chu  CM,  et al.  Treatment of severe acute respiratory syndrome with lopinavir/ritonavir: a multicentre retrospective matched cohort study.   Hong Kong Med J. 2003;9(6):399-406.PubMedGoogle Scholar
47.
Wu  C, Chen  X, Cai  Y,  et al.  Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China.   JAMA Intern Med. Published online March 13, 2020. PubMedGoogle Scholar
48.
Foolad  F, Aitken  SL, Shigle  TL,  et al.  Oral versus aerosolized ribavirin for the treatment of respiratory syncytial virus infections in hematopoietic cell transplant recipients.   Clin Infect Dis. 2019;68(10):1641-1649. doi:10.1093/cid/ciy760 PubMedGoogle ScholarCrossref
49.
Arabi  YM, 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. Published online June 25, 2019. doi:10.1093/cid/ciz544 PubMedGoogle Scholar
50.
Altınbas  S, Holmes  JA, Altınbas  A.  Hepatitis C virus infection in pregnancy: an update.   Gastroenterol Nurs. 2020;43(1):12-21. doi:10.1097/SGA.0000000000000404 PubMedGoogle ScholarCrossref
51.
Wang  D, Hu  B, Hu  C,  et al.  Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China.   JAMA. Published online February 7, 2020. doi:10.1001/jama.2020.1585 PubMedGoogle Scholar
52.
Totura  AL, Bavari  S.  Broad-spectrum coronavirus antiviral drug discovery.   Expert Opin Drug Discov. 2019;14(4):397-412. doi:10.1080/17460441.2019.1581171 PubMedGoogle ScholarCrossref
53.
Li  G, De Clercq  E.  Therapeutic options for the 2019 novel coronavirus (2019-nCoV).   Nat Rev Drug Discov. 2020;19(3):149-150. doi:10.1038/d41573-020-00016-0 PubMedGoogle ScholarCrossref
54.
Coleman  CM, Sisk  JM, Mingo  RM, Nelson  EA, White  JM, Frieman  MB.  Abelson kinase inhibitors are potent inhibitors of severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome coronavirus fusion.   J Virol. 2016;90(19):8924-8933. doi:10.1128/JVI.01429-16 PubMedGoogle ScholarCrossref
55.
Dyall  J, Gross  R, Kindrachuk  J,  et al.  Middle East respiratory syndrome and severe acute respiratory syndrome: current therapeutic options and potential targets for novel therapies.   Drugs. 2017;77(18):1935-1966. doi:10.1007/s40265-017-0830-1 PubMedGoogle ScholarCrossref
56.
Pfefferle  S, Schöpf  J, Kögl  M,  et al.  The SARS-coronavirus-host interactome: identification of cyclophilins as target for pan-coronavirus inhibitors.   PLoS Pathog. 2011;7(10):e1002331. doi:10.1371/journal.ppat.1002331 PubMedGoogle Scholar
57.
de Wilde  AH, Zevenhoven-Dobbe  JC, van der Meer  Y,  et al.  Cyclosporin A inhibits the replication of diverse coronaviruses.   J Gen Virol. 2011;92(pt 11):2542-2548. doi:10.1099/vir.0.034983-0 PubMedGoogle ScholarCrossref
58.
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(3):269-271. doi:10.1038/s41422-020-0282-0 PubMedGoogle ScholarCrossref
59.
Rossignol  JF.  Nitazoxanide, a new drug candidate for the treatment of Middle East respiratory syndrome coronavirus.   J Infect Public Health. 2016;9(3):227-230. doi:10.1016/j.jiph.2016.04.001 PubMedGoogle ScholarCrossref
60.
Gurwitz  D.  Angiotensin receptor blockers as tentative SARS-CoV-2 therapeutics.   Drug Dev Res. Published online March 4, 2020. doi:10.1002/ddr.21656 PubMedGoogle Scholar
61.
American Heart Association. Patients taking angiotensin converting enzyme inhibitors (ACE-i) or angiotensin receptor blocker (ARB) medications should continue therapy as prescribed [news release]. Published March 17, 2020. Accessed March 18, 2020. https://newsroom.heart.org/news/patients-taking-ace-i-and-arbs-who-contract-covid-19-should-continue-treatment-unless-otherwise-advised-by-their-physician
62.
European Society for Cardiology. Position statement of the ESC Council on Hypertension on ACE-Inhibitors and Angiotensin Receptor Blockers. Published March 13, 2020. Accessed March 18, 2020. https://www.escardio.org/Councils/Council-on-Hypertension-(CHT)/News/position-statement-of-the-esc-council-on-hypertension-on-ace-inhibitors-and-ang
63.
World Health Organization. WHO R&D blueprint: ad-hoc expert consultation on clinical trials for Ebola therapeutics. Published October 2018. Accessed March 20, 2020. https://www.who.int/ebola/drc-2018/summaries-of-evidence-experimental-therapeutics.pdf
64.
Jacobs  M, Rodger  A, Bell  DJ,  et al.  Late Ebola virus relapse causing meningoencephalitis: a case report.   Lancet. 2016;388(10043):498-503. doi:10.1016/S0140-6736(16)30386-5 PubMedGoogle ScholarCrossref
65.
Holshue  ML, DeBolt  C, Lindquist  S,  et al; Washington State 2019-nCoV Case Investigation Team.  First case of 2019 novel coronavirus in the United States.   N Engl J Med. 2020;382(10):929-936. doi:10.1056/NEJMoa2001191 PubMedGoogle ScholarCrossref
66.
Kujawski  SA, Wong  K, Collins  JP,  et al. First 12 patients with coronavirus disease 2019 (COVID-19) in the United States. medRxiv. Preprint posted March 9, 2020. doi:10.1101/2020.03.09.20032896
67.
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(7):449-463. doi:10.2183/pjab.93.027 PubMedGoogle ScholarCrossref
68.
Mentré  F, Taburet  AM, Guedj  J,  et al.  Dose regimen of favipiravir for Ebola virus disease.   Lancet Infect Dis. 2015;15(2):150-151. doi:10.1016/S1473-3099(14)71047-3 PubMedGoogle ScholarCrossref
69.
Sissoko  D, Laouenan  C, Folkesson  E,  et al; JIKI Study Group.  Experimental treatment with favipiravir for Ebola virus disease (the JIKI Trial): a historically controlled, single-arm proof-of-concept trial in Guinea  [published correction appears in PLoS Med. 2016;13(4):e1002009].  PLoS Med. 2016;13(3):e1001967. doi:10.1371/journal.pmed.1001967 PubMedGoogle Scholar
70.
Shiraki  K, Daikoku  T.  Favipiravir, an anti-influenza drug against life-threatening RNA virus infections.  [published online February 22, 2020].  Pharmacol Ther. 2020;107512. doi:10.1016/j.pharmthera.2020.107512 PubMedGoogle Scholar
71.
Chinello  P, Petrosillo  N, Pittalis  S, Biava  G, Ippolito  G, Nicastri  E; INMI Ebola Team.  QTc interval prolongation during favipiravir therapy in an Ebolavirus-infected patient.   PLoS Negl Trop Dis. 2017;11(12):e0006034. doi:10.1371/journal.pntd.0006034 PubMedGoogle Scholar
72.
Kumagai  Y, Murakawa  Y, Hasunuma  T,  et al.  Lack of effect of favipiravir, a novel antiviral agent, on QT interval in healthy Japanese adults.   Int J Clin Pharmacol Ther. 2015;53(10):866-874. doi:10.5414/CP202388 PubMedGoogle ScholarCrossref
73.
Chen  C, Huang  J, Cheng  Z,  et al. Favipiravir versus Arbidol for COVID-19: a randomized clinical trial. medRxiv. Preprint posted March 27, 2020. doi:10.1101/2020.03.17.20037432
74.
Liu  C, Zhou  Q, Li  Y,  et al.  Research and development of therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases.   ACS Cent Sci. 2020;6(3):315-331. doi:10.1021/acscentsci.0c00272 PubMedGoogle ScholarCrossref
75.
Gordon DE, Jang GM, Bouhaddou M, et al. A SARS-CoV-2-human protein-protein interaction map reveals drug targets and potential drug-repurposing. bioRxiv. Preprint posted March 22, 2020. doi:10.1101/2020.03.22.002386
76.
Russell  CD, Millar  JE, Baillie  JK.  Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury.   Lancet. 2020;395(10223):473-475. doi:10.1016/S0140-6736(20)30317-2 PubMedGoogle ScholarCrossref
77.
Arabi  YM, Mandourah  Y, Al-Hameed  F,  et al; Saudi Critical Care Trial Group.  Corticosteroid therapy for critically ill patients with Middle East respiratory syndrome.   Am J Respir Crit Care Med. 2018;197(6):757-767. doi:10.1164/rccm.201706-1172OC PubMedGoogle ScholarCrossref
78.
Ni  YN, Chen  G, Sun  J, Liang  BM, Liang  ZA.  The effect of corticosteroids on mortality of patients with influenza pneumonia: a systematic review and meta-analysis.   Crit Care. 2019;23(1):99. doi:10.1186/s13054-019-2395-8 PubMedGoogle ScholarCrossref
79.
Mehta  P, McAuley  DF, Brown  M, Sanchez  E, Tattersall  RS, Manson  JJ; HLH Across Speciality Collaboration, UK.  COVID-19: consider cytokine storm syndromes and immunosuppression.   Lancet. 2020;395(10229):1033-1034. doi:10.1016/S0140-6736(20)30628-0 PubMedGoogle ScholarCrossref
80.
Zhou  F, Yu  T, Du  R,  et al.  Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study.   Lancet. 2020;395(10229):1054-1062. doi:10.1016/S0140-6736(20)30566-3 PubMedGoogle ScholarCrossref
81.
Sanofi. Sanofi and Regeneron begin global Kevzara (sarilumab) clinical trial program in patients with severe COVID-19 [news release]. Published March 16, 2020. Accessed March 18, 2020. http://www.news.sanofi.us/2020-03-16-Sanofi-and-Regeneron-begin-global-Kevzara-R-sarilumab-clinical-trial-program-in-patients-with-severe-COVID-19
82.
Chen  L, Xiong  J, Bao  L, Shi  Y.  Convalescent plasma as a potential therapy for COVID-19.   Lancet Infect Dis. 2020;20(4):398-400. doi:10.1016/S1473-3099(20)30141-9 PubMedGoogle ScholarCrossref
83.
Soo  YO, Cheng  Y, Wong  R,  et al.  Retrospective comparison of convalescent plasma with continuing high-dose methylprednisolone treatment in SARS patients.   Clin Microbiol Infect. 2004;10(7):676-678. doi:10.1111/j.1469-0691.2004.00956.x PubMedGoogle ScholarCrossref
84.
Arabi  Y, Balkhy  H, Hajeer  AH,  et al.  Feasibility, safety, clinical, and laboratory effects of convalescent plasma therapy for patients with Middle East respiratory syndrome coronavirus infection: a study protocol.   Springerplus. 2015;4:709. doi:10.1186/s40064-015-1490-9 PubMedGoogle ScholarCrossref
85.
Hung  IF, To  KK, Lee  CK,  et al.  Convalescent plasma treatment reduced mortality in patients with severe pandemic influenza A (H1N1) 2009 virus infection.   Clin Infect Dis. 2011;52(4):447-456. doi:10.1093/cid/ciq106 PubMedGoogle ScholarCrossref
86.
Mair-Jenkins  J, Saavedra-Campos  M, Baillie  JK,  et al; Convalescent Plasma Study Group.  The effectiveness of convalescent plasma and hyperimmune immunoglobulin for the treatment of severe acute respiratory infections of viral etiology: a systematic review and exploratory meta-analysis.   J Infect Dis. 2015;211(1):80-90. doi:10.1093/infdis/jiu396 PubMedGoogle ScholarCrossref
87.
Shen  C, Wang  Z, Zhao  F,  et al.  Treatment of 5 critically ill patients with COVID-19 with convalescent plasma.   JAMA. 2020. Published online March 27, 2020. doi:10.1001/jama.2020.4783PubMedGoogle Scholar
88.
Cao  W, Liu  X, Bai  T,  et al.  High-dose intravenous immunoglobulin as a therapeutic option for deteriorating patients with coronavirus disease 2019.   Open Forum Infect Dis. Published online March 21, 2020. doi:10.1093/ofid/ofaa102 Google Scholar
89.
US Food and Drug Administration. Investigational COVID-19 Convalescent plasma: emergency INDs. Updated April 3, 2020. Accessed March 26, 2020. https://www.fda.gov/vaccines-blood-biologics/investigational-new-drug-ind-or-device-exemption-ide-process-cber/investigational-covid-19-convalescent-plasma-emergency-inds
90.
Wang C, Li W, Drabek D, et al. A human monoclonal antibody blocking SARS-CoV-2 infection. bioRxiv. Preprint posted March 11, 2020. doi:10.1101/2020.03.11.987958.2020
91.
Huang  C, Wang  Y, Li  X,  et al.  Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China.   Lancet. 2020;395(10223):497-506. doi:10.1016/S0140-6736(20)30183-5 PubMedGoogle ScholarCrossref
92.
Chen  N, Zhou  M, Dong  X,  et al.  Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study.   Lancet. 2020;395(10223):507-513. doi:10.1016/S0140-6736(20)30211-7 PubMedGoogle ScholarCrossref
93.
Yang  X, Yu  Y, Xu  J,  et al.  Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study.   Lancet Respir Med. Published online February 24, 2020. doi:10.1016/S2213-2600(20)30079-5 PubMedGoogle Scholar
94.
Young  BE, Ong  SWX, Kalimuddin  S,  et al; Singapore 2019 Novel Coronavirus Outbreak Research Team.  Epidemiologic features and clinical course of patients infected with SARS-CoV-2 in Singapore.   JAMA. Published online March 3, 2020. doi:10.1001/jama.2020.3204 PubMedGoogle Scholar
95.
Guan  WJ, Ni  ZY, Hu  Y,  et al; China Medical Treatment Expert Group for Covid-19.  Clinical Characteristics of Coronavirus Disease 2019 in China.   N Engl J Med. Published online February 28, 2020. doi:10.1056/NEJMoa2002032 PubMedGoogle Scholar
96.
Centers for Disease Control and Prevention. Coronavirus disease 2019 (COVID-19) clinical care. Updated March 30, 2020. Accessed March 18, 2020. https://www.cdc.gov/coronavirus/2019-ncov/hcp/clinical-guidance-management-patients.html
97.
World Health Organization. Clinical management of severe acute respiratory infection when COVID-19 is suspected. Updated March 13, 2020. Accessed March 18, 2020. https://www.who.int/publications-detail/clinical-management-of-severe-acute-respiratory-infection-when-novel-coronavirus-(ncov)-infection-is-suspected
98.
Kupferschmidt  K, Cohen  J. WHO launches global megatrial of the four most promising coronavirus treatments. Science. Published March 22, 2020. Accessed March 23, 2020. https://www.sciencemag.org/news/2020/03/who-launches-global-megatrial-four-most-promising-coronavirus-treatments#
19 Comments for this article
EXPAND ALL
Naproxen
Richard Brown, MA (Cantab) MSc (Edin) | Farm Veterinarian Scotland
I am surprised there is no mention of naproxen. The French may be about to start a trial with naproxen as a component of therapy (NCT04325633). Even if an RCT is not performed for Naproxen as a component, an epidemiological study should be performed to follow those who take naproxen long term (and who have been advised to continue to use it, ie some rheumatoid arthritis sufferers) and see if through this pandemic they have ( paradoxically) been less at risk.
CONFLICT OF INTEREST: None Reported
COVID-19 Angiotensin Paradigm can also be addressed
Andrew Ashworth, MbChB | Bonhard Medical, Scotland
This review of some pharmacological interventions (1) is a helpful summary but it restricts itself, perhaps based on the methodology, to interventions directed toward SARS-COV-2 virus itself and so does not include potential interventions to mitigate the clinical effects of COVID-19.

SARS-COV-2 targets ACE2 and, while reduced expression of ACE2 has not been shown, the assumption of such a reduction is consistent with clinical findings. ACE2 converts inactivates Angiotensin II to Angiotensin 1-7.(3). Without ACE2, Angiotensin II causes contraction of smooth muscle via the phosphodiesterase 5 (PDE5) pathway (4) with vasoconstriction and cough.

This ‘COVID-19 Angiotensin Paradigm”
is consistent with clinical effects mediated by vasoconstriction:
• Reduced gas exchange and therefore reduced oxygen supply to the systemic circulation
• Reduced blood-borne immune response to the viral particles
• Increased pressure in the Pulmonary artery with shunting of deoxygenated pulmonary arterial blood to the systemic circulation.
- Bronchiolar smooth muscle-mediated cough

Current pharmacological interventions appear to be focussed on antivirals. Current therapy relies on increasing alveolar oxygen concentration. If ACE2 fails to protect distal pulmonary vessels from Angiotensin II, then mitigating its effects has the immediate potential significantly to alter the progress of the disease. PDE-5 inhibitors are widely available and appear to have significant promise in addressing the increased exposure of pulmonary smooth muscle to Angiotensin II in COVID-19. There is an anecdotal report of a PDE5 inhibitor being used effectively in a similar historical case (5).

PDE5 inhibitors in COVID-19 offer a means of treatment in poorer countries where ventilatory support is less available. A clinical trial is required.

REFERENCES

1. Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic Treatments for Coronavirus Disease 2019 (COVID-19): A Review. JAMA. Published online April 13, 2020. doi:10.1001/jama.2020.6019(2)
2. Hoffmann et al., 2020, Cell 181, 1–10 April 16, 2020 a 2020 Elsevier Inc. https://doi.org/10.1016/j.cell.2020.02.052,
3. Tikellis, C.,Thomas, M.C. Angiotensin-Converting Enzyme 2 (ACE2) Is a Key Modulator of the Renin Angiotensin System in Health and Disease International Journal of Peptides Volume 2012, Article ID 256294, doi:10.1155/2012/256294
4. Dongsoo Kima, Toru Aizawab, Heng Weic, et al. Angiotensin II increases phosphodiesterase 5A expression in vascular smooth muscle cells: A mechanism by which angiotensin II antagonizes cGMP signaling J Mol Cell Cardiol. 2005 January ; 38(1): 175–184. doi:10.1016/j.yjmcc.2004.10.013
5. Ashworth AJ. Enhanced recovery from respiratory infection following treatment with a PDE-5 inhibitor: a single case study Prim Care Respir J 2012; 21(1): 17-18 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6547895/
CONFLICT OF INTEREST: None Reported
READ MORE
Pulmonary Delivery of Possible Therapeutic Agents for COVID-19
Hasham Shafi, PhD Pharmaceutics | CSIR-Central drug research institute, University of Kashmir
The review is well articulated and all therapeutic agents in trials or being used are discused. I would have expected researchers around the globe to give prime consideration to the route of drug delivery, It's well established that pulmonary delivery as a dry powder inhalation of some of the drug candidates can target these drugs directly to the site of infection and can reduce the drug dose especially in drugs with toxicity issues.
CONFLICT OF INTEREST: None Reported
Darunavir Has No Activity Against SARS-CoV-2
Marcelo Radisic, MD | d.Institute, Instituto de Trasplante y Alta Complejidad / Sanatorio Finochietto. Buenos Aires, Argentina.
Although darunavir is an effective inhibitor of the HIV-dimeric aspartyl protease, it has no demonstrated activity against SARS-CoV-2 protease, which is a cysteine protease. Darunavir has low affinity with the catalytic center of the SARS-CoV-2 protease active site.

Janssen, the manufacturer of darunavir, has reported that results from a single center, open label, randomised controlled trial conducted at Shanghai Public Health Clinical Center (SPHCC) testing darunavir and cobicistat (DRV/c) in treating 30 COVID-19 patients showed that DRV/c was not effective (1). In addition, the in-vitro antiviral activity of darunavir against SARS-CoV-2 was assessed and darunavir showed no activity
against SARS-CoV-2 at clinically relevant concentrations (EC50>100 μM).

These data do not support the use of darunavir for the treatment of COVID-19

REFERENCES

1. https://www.janssen.com/ireland/lack-evidence-support-use-darunavir-based-treatments-sars-cov-2, accessed April 4th, 2020
CONFLICT OF INTEREST: None Reported
READ MORE
Is Remdesivir the Answer for COVID-19?
Sarosh Ahmed Khan, MBBS; MD; FACP; FRCP Edin | Naseem Medical Center, Baghe Mehtab, Srinagar, Kashmir 190019
The anti-viral drug remdesivir, a nucleotide analogue prodrug that inhibits viral RNA polymerases, had until now shown only in vitro activity against SARS-CoV-2. It had also been tried in non-clinical models in ebola and other coronaviruses (SARS-CoV and MERSCoV) (1,2). It was investigated in Ebola virus infection & found to have a favorable clinical safety profile, as reported on the basis of experience in about 500 persons (volunteers and patients) (3,4).

But a recent small industry-conducted (Gilead Sciences) study appeared to show efficacy in seriously ill COVID-19 patients (5) . About 2/3rd of patients given the drug on compassionate-use
basis showed signs of clinical improvement. Patients had confirmed COVID-19 with an O2-sat of =< 94% while they were breathing ambient air or were receiving O2 support. Patients received a 10-day course of remdesivir, consisting of 200 mg administered IV on day 1, followed by 100 mg daily for the remaining 9 days. Of the 61 patients who received at least one dose of Remdesivir, data from 8 was not analyzed. At baseline, 57% were receiving mechanical ventilation(MV) and 8% were receiving ECMO. During a median follow-up of 18 days, 68% patients had an improvement in O2-support class, including 57% receiving MV who were extubated. A total of 25 patients were discharged, & 7 died; mortality was 18% among patients receiving invasive ventilation and 5% (1 of 19) among those not receiving invasive ventilation.

The downsides:
1. Small size of the cohort
2. Viral load data to confirm the antiviral effects of remdesivir or any association between baseline viral load & viral suppression were not collected
3. The duration of remdesivir therapy was not uniform
4. Shorter duration of therapy (e.g., 5 vs 10 days) was not studied 
5. 60% reported adverse events including raised liver enzymes, diarrhea, rash, renal impairment, & hypotension. AEs were more common in patients receiving invasive ventilation. Of 23% who had serious AEs, common ones were multi-organ dysfunction syndrome, septic shock, acute kidney injury, & hypotension. 4 patients had to stop treatment because of these
6. The study did not have a control arm so we don't know the contribution of other factors like type of supportive care (concomitant medications or variations in ventilatory practices) & differences in institutional treatment protocols & thresholds for hospitalization

Pharma companies seeing the desperate need of a drug have come up with a new name for “off-label use”: compassionate-use

We may not be able to draw definitive conclusions, but if these caveats are taken care of, it seems that remdesivir may have clinical benefit in patients with severe Covid-19

References

1. de Wit E et al. Prophylactic & therapeutic Remdesivir (GS-5734) treatment in the rhesus macaque model of MERS-CoV infection. Proc Natl Acad Sci U S A 2020; 117: 6771-6
2. Sheahan TP et al. Broad-spectrum antiviral GS-5734 inhibits both epidemic & zoonotic coronaviruses. Sci Tran Med 2017; 9(396): eaal3653
3. Mulangu S et al. A randomized, controlled trial of Ebola virus disease therapeutics. NEJM 2019; 381: 2293-303
4. EMA. Summary on compassionate use: Remdesivir Gilead. April 3 2020
5. Grein J et al. Compassionate Use of Remdesivir for Patients with Severe Covid-19. NEJM April 10 2020 DOI: 10.1056/NEJMoa2007016
CONFLICT OF INTEREST: None Reported
READ MORE
Novel CoViD Impairment of hypoxic pulmonary vasoconstriction hypothesis
Darren Emerick, MBBS | University of Queensland
Working on the hypothesis that novel CoViD unusually causes impairment of hypoxic pulmonary vasoconstriction, COVID-19 would be a hyperaemic pneumonia, resulting in major ventilation-perfusion mismatch.

Therapies that may modulate hypoxic pulmonary vasoconstriction include:

Buffered L-lactic acid infusion [+R shift ODC]
Angiotensin II infusion
Beta 2 agonist infusion
TASK-1 channel blockers eg doxapram, almitrine, 2-phenyl-3-(piperazinomethyl)imidazo[1,2-a]pyridine derivatives [Bayer phase 1 trials]
Desferrioxamine infusion
Hyperthermia [+R shift ODC]
Methanandamide/Anandamide infusion
Bupivacaine infusion

[ET-1 infusion seems unlikely to represent a therapeutic strategy for enhancing HPV during acute (<4 h) hypoxia]
CONFLICT OF INTEREST: None Reported
What Does "Most Promising" Mean?
Marlowe Fox, JD, MS | None
This meta-analysis seems to be an important splash of cold water on potential treatments. In particular, the HCQ/CQ trials that resulted in statistically significant p-values but failed to control/adjust for several confounding variables (1,2). However, the description of remdesivir as the "most promising treatment” seems to have even less empirical support.

It also begs the question of what constitutes the “most promising treatment.” Does it mean:

1. Best in-vitro effects (best mechanistic explanation/identification of mediator)
2. Highest probability of similar in-vivo effects
3. Robust empirical support (sample size; sufficiently articulated methods/evaluation procedures; controlling for comorbidities, symptom
onset, prescriptions i.e. RAAS inhibitors; statistical significance)
4. Probability of non-toxic, low side effect dosing
5. Availability, cost-effectiveness, other pragmatic concerns
6. Potential for prophylactic use as well
7. Reducing the contagious phase of the infected

And if this was the case, why did the article not articulate as much in its methods? This would obviate any concern of an ad hoc conclusion or any other potential bias.

Remdesivir’s “promise” seems to be based on its “potent” in vitro effects against SARS-CoV-2 as well as two studies in which a total of four patients received remdesivir (3,4). In one study, a 35 y/o healthy male, after progressively worsening symptoms, received treatment on day 7 of hospital admission (day 11 of symptom onset). Within 24 hours, the patient’s oxygen saturation went from 90% to 96%, and he was taken off supplemental oxygen (3). In the other study 12 patients were examined, of which 3 received the treatment. The study was plagued by confounding variables. Not to mention, all 12 patients recovered (4). It should be noted that these studies are only footnoted in the article.

Whatever the HCQ trials were lacking, they offered at least some scientific rigor (1,2). The article criticizes the French study (2) for “a small sample size…the removal of 6 patients in the hydroxychloroquine group...” Similarly, it critiques the Wuhan study: “At day 7, virologic clearance was similar, with 86.7% vs 93.3% clearance for the hydroxychloroquine plus standard of care group and standard care group, respectively (P > .05).” However, the Wuhan study’s most noteworthy findings may have been the average reduction of about 1 day in fever and coughing, both of which were supported by p-values of 0.0008 and .0016 respectively (1). Assuming these results are accurate, cutting a day off symptoms could substantially decrease the infection rate as well as increase the availability of hospital resources. There are certainly concerns with the Wuhan study—p-hacking or HARKING could very well have been involved—but the same problems exist with the remdesivir reports.

With no clinical trials that sufficiently control for confounding variables, the most promising treatment would likely be the one with the most empirically supported mechanism (mediator). I would be interested in a meta-analysis that surveyed all the potential causal mechanisms, whether they be anti-viral or inhibition/blocking somewhere along the RAAS-pathway (5).

References

1. https://www.medrxiv.org/content/10.1101/2020.03.22.20040758v3
2. https://www.medrxiv.org/content/10.1101/2020.03.16.20037135v1
3. https://www.nejm.org/doi/10.1056/NEJMoa2001191
4. https://www.medrxiv.org/content/10.1101/2020.03.09.20032896v1
5. https://jamanetwork.com/journals/jama/fullarticle/2763803
CONFLICT OF INTEREST: None Reported
READ MORE
Androgen Pathway Targets and Incomplete TMPRSS2 Inhibitors list
Carlos Wambier, MD, PhD | Warren Alpert Medical School, Brown University
This article mentions Transmembrane Protease Serine 2 (TMPRSS2) but fails to address the pivotal role of androgen receptor (RA) activation for transcription of the TMPRSS2 gene. The androgen pathway is key to individual vulnerability, since androgen-promoted proteins are increasingly expressed after puberty (1).

Clinical signs of androgen expression such as androgenic alopecia (2) could be strictly linked to vulnerability.

Thus, targeting any step of the androgen pathway with the following agents may theoretically increase host resistance:

LH (GnRH) analogues: Degarelix, Goserelin, Leuprolide, Leuprorelin, Nafarelin 
Testosterone (steroidogenesis) inhibitors: Ketoconazole, Fluconazole, Itraconazole 
5-alpha reductase inhibitors: Dutasteride, Finasteride 
RA inhibitors:
Spironolactone, Bicalutamide, Darolutamide, Enzalutamide, Flutamide, Nilutamide: 

All of which are used for androgenic suppression.

Finally, the first TMPRSS2 inhibitor described was bromhexine, a common cough medication (3), to add to camostat, a new drug.

The list above indexes the main medications that might be used to target the androgen pathway of viral entry in cells (through TMPRSS2 priming of both viral spike and ACE2) (1).

Studies for male prophylaxis with medications that have a favorable side-effect profile (5-alpha reductase inhibitors), and in life-threatening circumstances, chemical castration, as done in metastatic prostate cancer, could also be tested in clinical trials.

Some patients present with increased risk of thrombosis from androgen blockade. A phytochemical compound, quercetin-3-β-O-D-glucoside (isoquercetin), with antiviral activity against Zika and Ebola virus(4,5), inhibits the androgen receptor (6) and targets extracellular protein disulfide isomerase (PDI), improving markers of coagulation in advanced cancer patients (7). PDI is a thiol isomerase secreted by vascular cells, that is required for thrombus formation.

REFERENCES:
1. Wambier CG, Goren A. SARS-COV-2 infection is likely to be androgen mediated. J Am Acad Dermatol. April 2020. doi:10.1016/j.jaad.2020.04.032
2. Goren A, McCoy J, Wambier CG, et al. What does androgenetic alopecia have to do with COVID-19? An insight into a potential new therapy. Dermatol Ther. April 2020:e13365. doi:10.1111/dth.13365
3. Lucas JM, Heinlein C, Kim T, et al. The Androgen-Regulated Protease TMPRSS2 Activates a Proteolytic Cascade Involving Components of the Tumor Microenvironment and Promotes Prostate Cancer Metastasis. Cancer Discov. 2014;4(11):1310-1325. doi:10.1158/2159-8290.CD-13-1010
4. Qiu X, Kroeker A, He S, et al. Prophylactic efficacy of quercetin 3-β-O-D-glucoside against Ebola virus infection. Antimicrob Agents Chemother. 2016;60(9):5182-5188. doi:10.1128/AAC.00307-16
5. Wong G, He S, Siragam V, et al. Antiviral activity of quercetin-3-β-O-D-glucoside against Zika virus infection. Virol Sin. 2017;32(6):545-547. doi:10.1007/s12250-017-4057-9
6. Xing N. Quercetin inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells. Carcinogenesis. 2001. doi:10.1093/carcin/22.3.409
7. Zwicker JI, Schlechter BL, Stopa JD, et al. Targeting protein disulfide isomerase with the flavonoid isoquercetin to improve hypercoagulability in advanced cancer. JCI insight. 2019;4(4):1-12. doi:10.1172/jci.insight.125851
CONFLICT OF INTEREST: None Reported
READ MORE
Reconsider Corticosteroids
Lei Zhang, M.D. | Tianjin Cancer Institution and Hospital
People have a negative attitude towards use of corticosteroids for COVID-19 (1) for several reasons. First is that the original protocol in SARS used large doses (2) with potential for serious complications, including femoral head necrosis, secondary infections, etc. (3) with outcomes attributed to SARS rather than the treatment. Secondly, no potential effective antiviral drugs such as remdesivir were developed at the time when SARS broke out, (4) so large doses undoubtedly caused an increase in viral replication and delay in viral clearance (5).

Postmortem analysis of COVID-19 patients has confirmed the lung tissue injury caused by cytokine storms
and the formation of acute respiratory distress syndrome (ARDS) (6), which is a problem that antiviral drugs cannot solve. Twenty years ago, methylprednisolone therapy was proven effective for improving lung injury and reducing mortality in ARDS (7). Recently, in treatment of COVID-19, it has also been reported that the use of steroids, especially low-dose therapy, can effectively reverse the condition of severe patients and reduce death. (8) (9) If we could change the traditional usage pattern, adopt early low-dose corticosteroids therapy, and use them with effective antiviral drugs, the mortality rate of severe COVID-19 patients might be reduced .

REFERENCE
1, Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic Treatments for Coronavirus Disease 2019 (COVID-19): A Review [published online ahead of print, 2020 Apr 13]. JAMA. 2020;10.1001/jama.2020.6019. doi:10.1001/jama.2020.6019
2, Sung JJ, Wu A, Joynt GM, et al. Severe acute respiratory syndrome: report of treatment and outcome after a major outbreak. Thorax.2004;59(5):414–420.
3, Hong N, Du XK. Avascular necrosis of bone in severe acute respiratory syndrome. Clin Radiol. 2004;59(7):602–608. doi:10.1016/j.crad.2003.12.008
4, Grein J, Ohmagari N, Shin D, et al. Compassionate Use of Remdesivir for Patients with Severe Covid-19 [published online ahead of print, 2020 Apr 10]. N Engl J Med. 2020;10.1056/NEJMoa2007016.doi:10.1056/NEJMoa2007016
5, Lee N, Allen Chan KC, Hui DS, Ng EK, Wu A, et al. (2004) Effects of early corticosteroid treatment on plasma SARS-associated coronavirus RNA concentrations in adult patients. J Clin Virol 31: 304–309.
6, Xu Z, Shi L, Wang Y, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med 2020 Feb 18 [Epub ahead of print].
7, Meduri GU, Headley AS, Golden E, et al. Effect of prolonged methylprednisolone therapy in unresolving acute respiratory distress syndrome: a randomized controlled trial. JAMA. 1998;280(2):159–165. doi:10.1001/jama.280.2.159
8, Zheng C, Wang J, Guo H, et al. Risk-adapted Treatment Strategy For COVID-19 Patients [published online ahead of print, 2020 Mar 27]. Int J Infect Dis. 2020;S1201-9712(20)30179-X. doi:10.1016/j.ijid.2020.03.047
9, Wu C, Chen X, Cai Y, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. Published online March 13,
CONFLICT OF INTEREST: None Reported
READ MORE
The Social Life of SARS-Cov-2 With Therapeutic Implications
Arturo Tozzi, Pediatrician | University of North Texas
Viruses may spread not just as single particles, but also as collective aggregates (Segredo-Otero and Sanjuán, 2019; Andreu-Moreno and Sanjuán, 2020). These assemblies stand for “viral communities” with enhanced infectious capacity and improved spread compared with “free” viral particles (Cuevas et al., 2020). It is well known that coronavirus particles are able to stick together through virion-virion binding and to form aggregates. In particular, their particles tightly adhere with their projections sticking into each other, forming a mosaic patch that leads virions to squeeze and lose their spherical shape (Lin et al., 2004; Groneberg et al., 2005). It has been shown that, when SARS-Cov-2 grows in supernatants of infected cells, virions tend to aggregate in small globular assemblies that progressively give rise to larger net-like aggregates (Peter Doherty Institute for Infection and Immunity, https://www.youtube.com/watch?v=qTt3P5V8M1A&feature=youtu.be).

The ability to build particles assemblies and achieve collective dynamical behavior may provide invaluable advantages to SARS-Cov-2. The squeezing in their spherical shape allows particles to achieve a best package, increasing their number in a given amount of host fluids and maximizing viral load. SARS-CoV-2 positive patients with few/no symptoms and modest levels of detectable viral RNA in the oropharynx have been described (Zou et al., 2020). This finding, together with the observation that SARS-CoV-2 displays a well-known decay rate both in aerosols and various surfaces (van Doremalen et al., 2020), suggests the possibility that reduced viral loads could be correlated with decreased viral ability to build particles clustering. The globular-like arrangement of multiple SARS-CoV-2 virions may provide another advantage against host immunity and environmental offenses: even if immune systems or environmental factors engage the external core of the viral assembly, an inner viral sanctuary might be spared from further damages. It is noteworthy that, while VSV multi-virion complexes occur unfrequently in standard cell cultures, they are abundant in other fluids such as saliva (Cuevas et al., 2020). Further, it might be hypothesized that the lower symptomatic response in children to COVID-19 (Huang et al., 2020; Bi et al., 2020) could be correlated with local factors endowed in the pediatric respiratory airways that are able to scatter the viral assemblies responsible for symptoms severity. In sum, clustered SARS-Cov-2 dissemination stands for a potential target leading to novel antiviral strategies able to mechanically disrupt virionic assemblies.

Arturo Tozzi
Center for Nonlinear Science, Department of Physics, University of North Texas, Denton, Texas, USA
tozziarturo@libero.it
Arturo.Tozzi@unt.edu

James F. Peters
Department of Electrical and Computer Engineering, University of Manitoba
Department of Mathematics, Adıyaman University, 02040 Adıyaman, Turkey
james.peters3@umanitoba.ca

Isabella Annesi-Maesano
French NIH (INSERM), EPAR Department, IPLESP, INSERM
Sorbonne University, Paris, France.
isabella.annesi-maesano@inserm.fr

Gennaro D'Amato
Division of Respiratory and Allergic Diseases, Department of Chest Diseases, High Specialty A. Cardarelli Hospital, Napoli, Italy
Medical School of Specialization in Respiratory Diseases, University on Naples Federico II.
gdamatomail@gmail.com
CONFLICT OF INTEREST: None Reported
READ MORE
Clarification on Angiotensin Receptor Blockers' Proposed Role in SARS-CoV-2
James Cutrell, MD | UT Southwestern Medical Center, Dallas, Texas
We would like to clarify a statement in our current review on page E7 which reads, “In contrast, angiotensin receptor blockers could theoretically provide clinical benefit via blockade of ACE2 receptors.”

One proposed mechanism for angiotensin receptor blockers' (ARBs') amelioration of SARS-CoV-2 lung injury stems from ARB inhibition of the angiotensin receptor 1 (AT1R), not direct inhibition of the ACE2 receptor. This blockade in theory dampens angiotensin II mediated AT1R activation and downstream signaling that underlies SARS-CoV-2 mediated lung injury. An additional purported ARB mechanism of lung protection is ACE2 upregulation and subsequent increased conversion of angiotensin II
to angiotensin 1-7, a known vasodilator. These clinical benefits of ARBs have not been established but are being studied in ongoing clinical trials.

This clarification does not affect the clinical recommendations in our review which are concordant with major clinical societies and practice guidelines recommending continued therapy with ACE inhibitors or ARBs in patients already on these agents.

1. Gurwitz D. Angiotensin receptor blockers as tentative SARS-CoV-2 therapeutics. Drug Dev Res. Published online March, 4, 2020. doi:10.1002/ddr.21656

2. Patel AB, Verma A. COVID-19 and Angiotensin-Converting Enzyme Inhibitors and Angiotensin Receptor Blockers: What is the Evidence? JAMA. Published online March 24, 2020. doi:10.1001/jama.2020.4812
CONFLICT OF INTEREST: Dr. Cutrell received non-financial support from Regeneron and Gilead outside the submitted work.
READ MORE
Absence of Effective Treatment
Eduardo Quinteros, MD. Board IM Cardio | Internal Medicine and Critical care, Clinica Mayo Bell Ville, Argentina
After reading this review I thought the title might better be "Absence of Probable Treatments for COVID19." People are desperate for an oseltamivir for SARS-CoV-2, and medical journals become repositories of letters and reviews with 70-patient studies that are too limited to provide a solution. Some comments are at the level of what you hear in a supermarket line, spreading more confusion. We'd do well to remember the first principle of medicine: first, do no harm.
CONFLICT OF INTEREST: None Reported
Is There Evidence for Treatment with Hydroxychloroquine & Azithromycin?
John Baer, M.D. |
Regarding the hydroxychloroquine-azithromycin study cited (ref 16), I have concerns. I recommend having a look at the methods & the detailed patient data present at the end of the draft of the paper (Supplementary Table 1: https://www.medrxiv.org/content/10.1101/2020.03.16.20037135v1.full.pdf). This table is not included in the final publication. Method of testing was cell culture cytotoxicity, then PCR of the supernatant. Lots of room for false positive and negatives, e.g. some patients in both the control and treatment groups oscillated between positive and negative; is "POS" a patient with cytotoxicity alone, without PCR confirmation? There is lots of missing and heterogeneous data in the control group. The methods and detailed patient data raise significant questions about whether there was a treatment effect.
CONFLICT OF INTEREST: None Reported
READ MORE
Statins and COVID-19
Timo Strandberg, MD, PhD | Universities of Helsinki and Oulu, and Helsinki University Hospital
Sanders et al present various pharmacologic possibilities to prevent and treat COVID-19 (1). They do not mention statin treatment, which is a potential way to improve host resistance without directly attacking the virus (2).

Statins have favorable effects on endothelium dysfunction and may also prevent thromboembolic complications (3). Although statins have anti-inflammatory and immunomodulatory properties and their use has been associated with less complications during infections (2), I don’t believe that statins would act as direct antimicrobial agents in COVID-19. But a considerable portion of patients have cardiovascular diseases or diabetes, and statins are evidence-based treatment to reduce
morbidity and mortality in those individuals (4). Statin treatment is also associated with mortality benefit among older, frail patients (5), a considerable group of COVID-19 victims.

That low cholesterol is associated with worse prognosis in many acute diseases and in frailty is no argument to avoid statin treatment, because the mechanisms of ‘endogenous’ (due to acute conditions, frailty) and ‘exogenous’ (caused by statins) cholesterol reduction are different (6).

Statins are well-known, generally safe, cheap, and effective drugs. However, adherence is frequently not optimal due to, for example, fake information on the internet and social media. I think appropriate use of statins among patients with cardiovascular risk should be actively promoted during the COVID-19 pandemia.

REFERENCES

1. Sanders JM, Monogue ML, Jodlowski TZ, et al. Pharmacologic treatments for coronavirus disease 2019 (COVID-19)A Review. JAMA. Published online April 13, 2020. doi:10.1001/jama.2020.6019
2. Fedson DS. Treating the host response to emerging virus disease: lessons learned from sepsis, pneumonia, influenza and Ebola. Ann Transl Med 2016;4:421
3. Kunutsor SK, Seidu S, Khunti K. Statins and primary prevention of venous thromboembolism: a systematic review and meta-analysis. Lancet Haematol. 2017 Feb;4(2):e83-e93. doi: 10.1016/S2352-3026(16)30184-3.
4. Cholesterol Treatment Trialists' Collaboration. Efficacy and safety of statin therapy in older people: a meta-analysis of individual participant data from 28 randomised controlled trials. Lancet. 2019;393(10170):407-415
5. Strandberg TE. Deprescribing statins-Is it ethical? J Am Geriatr Soc. 2016;64(9):1926-7.
6. Gnanenthiran SR, Ng ACC, Cumming R, et al. Low total cholesterol is associated with increased major adverse cardiovascular events in men aged ≥70 years not taking statins. Heart. 2019 Oct 13. pii: heartjnl-2019-315449. doi: 10.1136/heartjnl-2019-315449
CONFLICT OF INTEREST: Collaborations (research, consultative, educational) with companies (including Amgen, Merck, Orion, Sanofi, Servier) and other entities interested in cholesterol-lowering. I take a statin daily.
READ MORE
Vitamin C
Harri Hemila, MD,PhD | University of Helsinki
The literature summarizing a possible role for vitamin C in COVID-19 is briefly summarized at https://pubpeer.com/publications/61151FA38F4AC67A54273EDC4C1C6E
CONFLICT OF INTEREST: None Reported
Disappointing Review
Todd Clark, MD | ER Physician in Private Hospital System with Academic Affiliations
Disappointing review mostly in its overemphasis of certain weak therapies (remdesivir for one), and complete omission of other promising therapies (vitamin C (Dr. Marik’s critical care protocol) and heparin (DIC prevention), to name a few).

Remdesivir study patients were cherry-picked and would have gotten better without the drug.

Only 15 comments after over 700,000 views of this article is alarming as well. Hoping people aren’t just taking what is written here as gospel.
CONFLICT OF INTEREST: None Reported
Biased Review
Dinesh Ranjan, MD, FACS | PRAN Philanthropic Clinic
Sanders and colleagues have published a detailed review of pharmacologic treatment options for Covid-19 in JAMA (1). Two drugs, hydroxychloroquine (HCQ) and remdesivir, have garnered most attention by medical journals and public media lately. While the French study touting HCQ with azithromycin had several shortcomings (2), it was hailed by President Trump regardless. The academic medicine, medical journals and main-stream media have condemned HCQ. In contrast however, remdesivir seems to have caught the fancy of the same group who seem to be willing to ignore the shortcomings of remdesivir data. This double standard is evident in this review.

The
authors state that they reviewed English language”articles catalogued in PubMed. However, they cite a Chinese language paper not catalogued in PubMed, showing no benefit with HCQ (3). They appear to ignore other English language papers supporting HCQ. Finally, the authors conclude that they “do not support adoption” of HCQ/Azithromycin “without additional studies”.

In contrast, when discussing remdesivir; the authors recommend that “inclusion of this agent for treatment of Covid-19 may be considered”. This recommendation is based upon “anticipated results from RCTs” and “successful case reports” in Covid-19 patients. Recommendations are based on anticipated results? And the successful case reports they state includes a study of 3 (out of 7 hospitalized) patients, without any difference in outcome. The authors, while making a case for its antiviral properties, state that remdesivir was used in clinical trials in Ebola – but they fail to mention that their cited reference did not include humans (4). They mention other single case reports of remdesivir use in Ebola. Unfortunately, they neglect to mention that the definitive study on Ebola therapeutics: a randomized trial of four therapeutic options, had not supported Remdesivir (5). Surely, a search in PubMed had brought up this NEJM paper? Why was this ignored while the authors were using single case reports to support remdesivir?

That remdesivir has become the favorite in journals and media is obvious (6). And it may yet be the best option for our patients once we have results from trials. We just wish that the reviews and recommendations published in respected journals will use an even-handed approach and not be openly cherry-picking information to support possible preexisting biases.

REFERENCE

1. Sanders JM, Monogue ML, Jodlowski TZ et al. JAMA. 2020 Apr 13. doi: 10.1001/jama.2020.6019.
2. Gautret P, Lagier JC, Parola P et al. . Int J Antimicrob Agents. 2020 Mar 20:105949. doi: 10.1016/j.ijantimicag.2020
3. Chen J,Liu D,Liu L, etal. J Zhejiang Univ (MedSci). Published on line March 6, 2020.doi:10.3785/j
4. https://www.who. int/ebola/drc-2018/summaries-of-evidenceexperimental-therapeutics.pdf
5. Mulangu S, Dodd LE, Davey RT. Randomized, Controlled Trial of Ebola Virus Disease Therapeutics N Engl J Med. 2019 Dec 12;381(24):2293-2303
6. 7. Grein J, Ohmagari N, Shin D et al. Compassionate Use of Remdesivir for Patients with Severe Covid-19. N Engl J Med. 2020 Apr 10. doi: 10.1056/NEJMoa2007016
CONFLICT OF INTEREST: None Reported
READ MORE
Be Humble, Alert, and Creative!
Bert Govig, MD, MPH | McGill University
Thanks to Dr Sanders and his team for this clear paper and particularly for repetition of the fundamental point: there is no known treatment for COVID-19 ... RCTs are needed.

This sober fact is humbling. In our search for treatments we should remember:

* Graphical illustration helps us think about these drugs, but should not fool us into thinking we understand the disease. It is quite likely that the drugs we will use to treat COVID-19 will work through unknown or unanticipated mechanisms. ACE inhibitors, Beta Blockers, statins, PDE-5 inhibitors, SGLT-2 inhibitors, and even hydroxy chloroquine
are used today for purposes that were accidentally discovered after they were in clinical use.
* Most of the candidate drugs will either have no effect or will cause harm. That is the nature of pharmacologic research. However we have hundreds of thousands of patients that will die from COVID and outcomes (particularly hard ones like death) drive power. We have the power, and the professional and moral mandate to rapidly eliminate drugs that don’t work until solutions are found. This requires focus, discipline, coordination, and leadership.
* We should not ignore nonpharmacologic treatment. Prone ventilation seemed foolish until it worked. Similarly, we have discovered in clinical medicine that sleep, diet, stress, smoking, body weight, and a host of other behaviours dramatically influence health and disease. As we look for pharmacologic treatments for COVID-19 patients, we should cast our scientific gaze broadly. The goal is to save patients by all means possible.
CONFLICT OF INTEREST: None Reported
READ MORE
Sivelestat for ARDS in COVID-19
Hiroyuki Okura, MD. | Department of Cardiology, Gifu University Graduate School of Medicine, Gifu, Japan
This review provides comprehensive, useful and timely information to the doctors who are currently fighting against the devastating pandemic situation all over the world. While I was reading this review article I noticed that one drug commonly used for the treatment of acute respiratory distress syndrome (ARDS) in Japan is missing. The drug is sivelestat, a selective neutrophil elastase inhibitor, which is commercially available in Japan, but not in China, USA or European countries. Based on favorable results of a phase III trial (1), this drug was approved for the treatment of acute lung injury caused by systemic inflammatory response syndrome in Japan. On the other hand, because an international randomized trial failed to demonstrate efficacy of sivelestat in patients with moderate to severe acute respiratory distress syndrome (ARDS) (2), it has not been on the global market. Therefore, it is not surprising that there are no published reports regarding the use of sivelestat during treatment of ARDS caused by COVID-19 (as of May 8, 2020). A recent retrospective analysis using a Japanese nationwide administrative database (Diagnostic Procedure Combination; DPC) in 2012 demonstrated that the early (within 7 days) use of sivelestat may improve outcome in patients with acute lung injury/ ARDS (3). Although I do not have full access to the Japanese nationwide status of the drugs used for COVID-19, I found a case report (written in Japanese) describing 2 clinical cases who were successfully recovered after intensive treatments including use of sivelestat (4). As of May, 9, Japan is one of the countries with the lowest mortality rate due to COVID-19 (case-fatality rate of 3.6 % and deaths/100k population of 0.44) (5). Although exact mechanisms for the differences in mortality due to COVID-19 is unclear, the inter-country differences in the specific drugs used for ARDS such as sevelestat may, in-part, explain the differences in mortality.

References
1. Tamakuma S, Shibaya T, Hirasawa H, Ogawa M, Nakashima M. A Phase Ⅲ Clinical Study of a Neutrophil Elastase lnhibitor;ONO-50460・Na in SIRS Patients. Rinsyoiyaku. 1998;14(2):289-318.
2. Zeiher BG, Matsuoka S, Kawabata K, Repine JE. Neutrophil elastase and acute lung injury: prospects for sivelestat and other neutrophil elastase inhibitors as therapeutics. Crit Care Med. 2002;30(5 Suppl):S281-287.
3. Kido T, Muramatsu K, Yatera K, et al. Efficacy of early sivelestat administration on acute lung injury and acute respiratory distress syndrome. Respirology. 2017;22(4):708-713.
4. The Japanese Association for Infectious Diseases. case report (http://www.kansensho.or.jp/uploads/files/topics/2019ncov/covid19_casereport_200312_4.pdf). Accessed on May 8, 2020.
5. The Johns Hopkins Hospital. COVID-19 data analysis center. Mortality Analysis. (https://coronavirus.jhu.edu/data/mortality) Accessed on May 8, 2020.
CONFLICT OF INTEREST: Research grant from ONO PHARMACEUTICAL CO., LTD.
READ MORE
Review
April 13, 2020

Pharmacologic Treatments for Coronavirus Disease 2019 (COVID-19): A Review

Author Affiliations
  • 1Department of Pharmacy, University of Texas Southwestern Medical Center, Dallas
  • 2Division of Infectious Diseases and Geographic Medicine, Department of Medicine, University of Texas Southwestern Medical Center, Dallas
  • 3Pharmacy Service, VA North Texas Health Care System, Dallas
JAMA. 2020;323(18):1824-1836. doi:10.1001/jama.2020.6019
Abstract

Importance  The pandemic of coronavirus disease 2019 (COVID-19) caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents an unprecedented challenge to identify effective drugs for prevention and treatment. Given the rapid pace of scientific discovery and clinical data generated by the large number of people rapidly infected by SARS-CoV-2, clinicians need accurate evidence regarding effective medical treatments for this infection.

Observations  No proven effective therapies for this virus currently exist. The rapidly expanding knowledge regarding SARS-CoV-2 virology provides a significant number of potential drug targets. The most promising therapy is remdesivir. Remdesivir has potent in vitro activity against SARS-CoV-2, but it is not US Food and Drug Administration approved and currently is being tested in ongoing randomized trials. Oseltamivir has not been shown to have efficacy, and corticosteroids are currently not recommended. Current clinical evidence does not support stopping angiotensin-converting enzyme inhibitors or angiotensin receptor blockers in patients with COVID-19.

Conclusions and Relevance  The COVID-19 pandemic represents the greatest global public health crisis of this generation and, potentially, since the pandemic influenza outbreak of 1918. The speed and volume of clinical trials launched to investigate potential therapies for COVID-19 highlight both the need and capability to produce high-quality evidence even in the middle of a pandemic. No therapies have been shown effective to date.

Introduction

The global pandemic of novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) began in Wuhan, China, in December 2019, and has since spread worldwide.1 As of April 5, 2020, there have been more than 1.2 million reported cases and 69 000 deaths in more than 200 countries. This novel Betacoronavirus is similar to severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV); based on its genetic proximity, it likely originated from bat-derived coronaviruses with spread via an unknown intermediate mammal host to humans.1 The viral genome of SARS-CoV-2 was rapidly sequenced to enable diagnostic testing, epidemiologic tracking, and development of preventive and therapeutic strategies.

Currently, there is no evidence from randomized clinical trials (RCTs) that any potential therapy improves outcomes in patients with either suspected or confirmed COVID-19. There are no clinical trial data supporting any prophylactic therapy. More than 300 active clinical treatment trials are underway. This narrative review summarizes current evidence regarding major proposed treatments, repurposed or experimental, for COVID-19 and provides a summary of current clinical experience and treatment guidance for this novel epidemic coronavirus.

Methods

A literature review was performed using PubMed to identify relevant English-language articles published through March 25, 2020. Search terms included coronavirus, severe acute respiratory syndrome coronavirus 2, 2019-nCoV, SARS-CoV-2, SARS-CoV, MERS-CoV, and COVID-19 in combination with treatment and pharmacology. The search resulted in 1315 total articles. Due to the lack of RCTs, the authors also included case reports, case series, and review articles. The authors independently reviewed the titles and abstracts for inclusion. Additional relevant articles were identified from the review of citations referenced. Active clinical trials were identified using the disease search term coronavirus infection on ClinicalTrials.gov and the index of studies of novel coronavirus pneumonia in the Chinese Clinical Trial Registry.2

SARS-CoV-2: Virology and Drug Targets

Quiz Ref IDSARS-CoV-2, a single-stranded RNA-enveloped virus, targets cells through the viral structural spike (S) protein that binds to the angiotensin-converting enzyme 2 (ACE2) receptor. Following receptor binding, the virus particle uses host cell receptors and endosomes to enter cells. A host type 2 transmembrane serine protease, TMPRSS2, facilitates cell entry via the S protein.3 Once inside the cell, viral polyproteins are synthesized that encode for the replicase-transcriptase complex. The virus then synthesizes RNA via its RNA-dependent RNA polymerase. Structural proteins are synthesized leading to completion of assembly and release of viral particles.4-6 These viral lifecycle steps provide potential targets for drug therapy (Figure). Promising drug targets include nonstructural proteins (eg, 3-chymotrypsin-like protease, papain-like protease, RNA-dependent RNA polymerase), which share homology with other novel coronaviruses (nCoVs). Additional drug targets include viral entry and immune regulation pathways.7,8 Table 1 summarizes the mechanism of action and major pharmacologic parameters of select proposed treatments or adjunctive therapies for COVID-19.

Ongoing Clinical Trials

The search terms COVID OR coronavirus OR SARS-COV-2 on ClinicalTrials.gov resulted in 351 active trials, with 291 trials specific to COVID-19 as of April 2, 2020. Of these 291 trials, approximately 109 trials (including those not yet recruiting, recruiting, active, or completed) included pharmacological therapy for the treatment of COVID-19 in adult patients. Of these 109 trials, 82 are interventional studies, with 29 placebo-controlled trials. Per description of the studies, there are 11 phase 4, 36 phase 3, 36 phase 2, and 4 phase 1 trials. Twenty-two trials were not categorized by phase or not applicable.

Review of Selected Repurposed Drugs

Quiz Ref IDAgents previously used to treat SARS and MERS are potential candidates to treat COVID-19. Various agents with apparent in vitro activity against SARS-CoV and MERS-CoV were used during the SARS and MERS outbreaks, with inconsistent efficacy. Meta-analyses of SARS and MERS treatment studies found no clear benefit of any specific regimen.37,38 Below, the in vitro activity and published clinical experiences of some of the most promising repurposed drugs for COVID-19 are reviewed.

Chloroquine and Hydroxychloroquine

Chloroquine and hydroxychloroquine have a long-standing history in the prevention and treatment of malaria and the treatment of chronic inflammatory diseases including systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA).7 Chloroquine and hydroxychloroquine appear to block viral entry into cells by inhibiting glycosylation of host receptors, proteolytic processing, and endosomal acidification. These agents also have immunomodulatory effects through attenuation of cytokine production and inhibition of autophagy and lysosomal activity in host cells.9,10 Chloroquine inhibits SARS-CoV-2 in vitro with a half-maximal effective concentration (EC50) in the low micromolar range. Hydroxychloroquine has in vitro activity with a lower EC50 for SARS-CoV-2 compared with chloroquine after 24 hours of growth (hydroxychloroquine: EC50 = 6.14 μM and chloroquine: EC50 = 23.90 μM).15

No high-quality evidence exists for the efficacy of chloroquine/hydroxychloroquine treatment of SARS or MERS. A news briefing from China reported chloroquine was successfully used to treat a series of more than 100 COVID-19 cases resulting in improved radiologic findings, enhanced viral clearance, and reduced disease progression.39 However, the clinical trial design and outcomes data have not yet been presented or published for peer review, preventing validation of these claims. A recent open-label nonrandomized French study of 36 patients (20 in the hydroxychloroquine group and 16 in the control group) reported improved virologic clearance with hydroxychloroquine, 200 mg, by mouth every 8 hours compared with control patients receiving standard supportive care. Virologic clearance at day 6, measured by nasopharyngeal swabs, was 70% (14/20) vs 12.5% (2/16) for the hydroxychloroquine and control groups, respectively (P = .001). The authors also reported that addition of azithromycin to hydroxychloroquine in 6 patients resulted in numerically superior viral clearance (6/6, 100%) compared with hydroxychloroquine monotherapy (8/14, 57%).16

Despite these promising results, this study had several major limitations: a small sample size (only 20 in the intervention arm and only 6 receiving hydroxychloroquine and azithromycin); the removal of 6 patients in the hydroxychloroquine group from analysis due to early cessation of treatment resulting from critical illness or intolerance of the medications; variable baseline viral loads between hydroxychloroquine monotherapy and combination therapy groups; and no clinical or safety outcomes reported. These limitations coupled with concerns of additive cardiotoxicity with combination therapy do not support adoption of this regimen without additional studies. Another prospective study of 30 patients in China randomized patients to hydroxychloroquine, 400 mg, daily for 5 days plus standard of care (supportive care, interferon, and other antivirals) or standard care alone in a 1:1 fashion; there was no difference in virologic outcomes. At day 7, virologic clearance was similar, with 86.7% vs 93.3% clearance for the hydroxychloroquine plus standard of care group and standard care group, respectively (P > .05).17 Currently, there are several RCTs of both chloroquine and hydroxychloroquine examining their role in COVID-19 treatment. Studies of chloroquine prophylaxis in health care workers (NCT04303507) and hydroxychloroquine for postexposure prophylaxis after high-risk exposures (NCT04308668) are planned or enrolling.40

Dosing of chloroquine to treat COVID-19 has consisted of 500 mg orally once or twice daily.11,12 However, a paucity of data exists regarding the optimal dose to ensure the safety and efficacy of chloroquine. Hydroxychloroquine dosing recommendations for SLE generally are 400 mg orally daily.18 However, a physiologically based pharmacokinetic modeling study recommended that the optimal dosing regimen for hydroxychloroquine in COVID-19 treatment is a loading dose of 400 mg twice daily for 1 day followed by 200 mg twice daily.15 In contrast, alternative recommendations are made for 600 mg total daily dose based on safety and clinical experience for Whipple disease.11 Further studies are needed to delineate the optimal dose for COVID-19.

Chloroquine and hydroxychloroquine are relatively well tolerated as demonstrated by extensive experience in patients with SLE and malaria. However, both agents can cause rare and serious adverse effects (<10%), including QTc prolongation, hypoglycemia, neuropsychiatric effects, and retinopathy.41,42 Baseline electrocardiography to evaluate for prolonged QTc is advisable prior to and following initiation of these medications because of the potential for arrhythmias, especially in critically ill patients and those taking concomitant QT-interval prolonging medications such as azithromycin and fluoroquinolones.13 No significant adverse effects have been reported for chloroquine at the doses and durations proposed for COVID-19.39 Use of chloroquine and hydroxychloroquine in pregnancy is generally considered safe.13,18 A review of 12 studies including 588 patients receiving chloroquine or hydroxychloroquine during pregnancy found no overt infant ocular toxicity.43

Lopinavir/Ritonavir and Other Antiretrovirals

Lopinavir/ritonavir, a US Food and Drug Administration (FDA)–approved oral combination agent for treating HIV, demonstrated in vitro activity against other novel coronaviruses via inhibition of 3-chymotrypsin-like protease.21,22 No published SARS-CoV-2 in vitro data exist for lopinavir/ritonavir.44 A systematic review of lopinavir/ritonavir for the treatment of SARS and MERS found limited available studies, with most of these investigating SARS. Clinical studies in SARS were associated with reduced mortality and intubation rates, but their retrospective, observational nature prevents definitive conclusions. The timing of administration during the early peak viral replication phase (initial 7-10 days) appears to be important because delayed therapy initiation with lopinavir/ritonavir had no effect on clinical outcomes.45,46

Early reports of lopinavir/ritonavir for the treatment of COVID-19 are mostly case reports and small retrospective, nonrandomized cohort studies, making it difficult to ascertain the direct treatment effect of lopinavir/ritonavir.45,46 More recently, Cao and colleagues23 reported the results of an open-label RCT comparing the efficacy of lopinavir/ritonavir vs standard care in 199 patients with COVID-19. Importantly, the median time from symptom onset to randomization was 13 days (interquartile range [IQR], 11-16), with no between-group difference. The primary outcome of time to clinical improvement defined by a 2-point improvement on a 7-category ordinal scale or hospital discharge was similar in both groups (16 days [IQR, 13-17] vs 16 days [IQR, 15-17]; hazard ratio [HR], 1.31 [95% CI, 0.95-1.85]; P = .09). Additionally, no significant differences in viral clearance or 28-day mortality rates (19.2% vs 25.0%; absolute difference, −5.8% [95% CI, −17.3% to 5.7%]) were observed. Although delayed treatment initiation may partially explain the ineffectiveness of lopinavir/ritonavir for treating COVID-19, a subgroup analysis did not find shorter time to clinical improvement for patients who received therapy within 12 days (HR, 1.25 [95% CI, 0.77-2.05]).23 Although additional RCTs of lopinavir/ritonavir are ongoing, the current data suggest a limited role for lopinavir/ritonavir in COVID-19 treatment.

The most commonly used and studied lopinavir/ritonavir dosing regimen for COVID-19 treatment is 400 mg/100 mg twice daily for up to 14 days.12,23 Given the significant drug-drug interactions and potential adverse drug reactions (summarized in Table 1), careful review of concomitant medications and monitoring are required if this drug is used. Adverse effects of lopinavir/ritonavir include gastrointestinal distress such as nausea and diarrhea (up to 28%) and hepatotoxicity (2%-10%).24 In patients with COVID-19, these adverse effects may be exacerbated by combination therapy or viral infection because approximately 20% to 30% of patients have elevated transaminases at presentation with COVID-19.47 A recent RCT showed approximately 50% of lopinavir/ritonavir patients experienced an adverse effect and 14% of patients discontinued therapy due to gastrointestinal adverse effects.23 Drug-induced transaminitis is of particular concern because it may exacerbate liver injury resulting from COVID-19. Importantly, alanine transaminase elevations are an exclusion criterion in several COVID-19 investigational trials, meaning that lopinavir/ritonavir-induced hepatotoxicity could limit patients’ ability to access these other drugs.40

Other antiretrovirals, including protease inhibitors and integrase strand transfer inhibitors, were identified by enzyme activity screening as having SARS-CoV-2 activity.44 In vitro cell models demonstrated activity of darunavir against SARS-CoV-2. There is no human clinical data in COVID-19 with these drugs, but an RCT of darunavir/cobicistat in China is underway.40

Ribavirin

Ribavirin, a guanine analogue, inhibits viral RNA-dependent RNA polymerase. Its activity against other nCoVs makes it a candidate for COVID-19 treatment. However, its in vitro activity against SARS-CoV was limited and required high concentrations to inhibit viral replication, necessitating high-dose (eg, 1.2 g to 2.4 g orally every 8 hours) and combination therapy. Patients received either intravenous or enteral administration in previous studies.37 No evidence exists for inhaled ribavirin for nCoV treatment, and data with respiratory syncytial virus suggest inhaled administration offers no benefit over enteral or intravenous administration.48

A systematic review of the clinical experience with ribavirin for the treatment of SARS revealed inconclusive results in 26 of the 30 studies reviewed, with 4 studies demonstrating possible harm due to adverse effects including hematologic and liver toxicity.37 In the treatment of MERS, ribavirin, generally in combination with interferons, demonstrated no discernible effect on clinical outcomes or viral clearance.38,49 A paucity of clinical data with ribavirin for SARS-CoV-2 means its therapeutic role must be extrapolated from other nCoV data.

Ribavirin causes severe dose-dependent hematologic toxicity. The high doses used in the SARS trials resulted in hemolytic anemia in more than 60% of patients.37 Similar safety concerns were seen in the largest MERS observational trial, with approximately 40% of patients taking ribavirin plus interferon requiring blood transfusions.49 Seventy-five percent of patients taking ribavirin for SARS experienced transaminase elevations.37 Ribavirin is also a known teratogen and contraindicated in pregnancy.50

The inconclusive efficacy data with ribavirin for other nCoVs and its substantial toxicity suggest that it has limited value for treatment of COVID-19. If used, combination therapy likely provides the best chance for clinical efficacy.

Other Antivirals

Oseltamivir, a neuraminidase inhibitor approved for the treatment of influenza, has no documented in vitro activity against SARS-CoV-2. The COVID-19 outbreak in China initially occurred during peak influenza season so a large proportion of patients received empirical oseltamivir therapy until the discovery of SARS-CoV-2 as the cause of COVID-19.51 Several of the current clinical trials include oseltamivir in the comparison group but not as a proposed therapeutic intervention.40 This agent has no role in the management of COVID-19 once influenza has been excluded.

Umifenovir (also known as Arbidol) is a more promising repurposed antiviral agent with a unique mechanism of action targeting the S protein/ACE2 interaction and inhibiting membrane fusion of the viral envelope.27 The agent is currently approved in Russia and China for the treatment and prophylaxis of influenza and is of increasing interest for treating COVID-19 based on in vitro data suggesting activity against SARS.28 The current dose of 200 mg orally every 8 hours for influenza is being studied for COVID-19 treatment (NCT04260594). Limited clinical experience with umifenovir for COVID-19 has been described in China. A nonrandomized study of 67 patients with COVID-19 showed that treatment with umifenovir for a median duration of 9 days was associated with lower mortality rates (0% [0/36] vs 16% [5/31]) and higher discharge rates compared with patients who did not receive the agent.29 This observational data cannot establish the efficacy of umifenovir for COVID-19, but ongoing RCTs in China are further evaluating this agent.

Miscellaneous Agents

Interferon-α and -β have been studied for nCoVs, with interferon-β demonstrating activity against MERS.37,38 Most published studies reported results of therapy combined with ribavirin and/or lopinavir/ritonavir. Similar to other agents, delayed treatment may limit effectiveness of these agents. Given conflicting in vitro and animal data and the absence of clinical trials, the use of interferons to treat SARS-CoV-2 cannot currently be recommended.52 Current Chinese guidelines list interferons as an alternative for combination therapy.12 Several other immunomodulatory agents traditionally used for noninfectious indications demonstrate in vitro activity or possess mechanisms purported to inhibit SARS-CoV-2, including, but not limited to, baricitinib, imatinib, dasatinib, and cyclosporine.53-57 However, no animal or human data exist to recommend their use for COVID-19, and it remains to be seen whether they confer protection for patients already taking them for other indications.

Nitazoxanide, traditionally an antihelminthic agent, has broad antiviral activity and a relatively favorable safety profile. Nitazoxanide has demonstrated in vitro antiviral activity against MERS and SARS-CoV-2.58,59 Pending further evidence, the antiviral activity, immunomodulatory effects, and safety profile of nitazoxanide warrant its further study as a treatment option for SARS-CoV-2.

Camostat mesylate, an approved agent in Japan for the treatment of pancreatitis, prevents nCoV cell entry in vitro through inhibition of the host serine protease, TMPRSS2.3 This novel mechanism provides an additional drug target for future research.

SARS-CoV-2 uses the ACE2 receptor for entry into the host cell.3 This discovery has stimulated discussions about whether ACE inhibitors and/or angiotensin receptor blockers may potentially treat COVID-19 or, conversely, worsen disease.60 These drugs upregulate ACE2 receptors, which could theoretically lead to worse outcomes if viral entry is enhanced. In contrast, angiotensin receptor blockers could theoretically provide clinical benefit via blockade of ACE2 receptors. Conflicting in vitro data exist to determine if these agents have a detrimental or protective effect in patients with COVID-19. Pending further research, clinical societies and practice guidelines are recommending continuing therapy for patients already taking 1 of these agents.61,62

Review of Select Investigational Drugs
Remdesivir

Quiz Ref IDRemdesivir, formally known as GS-5734, is a monophosphate prodrug that undergoes metabolism to an active C-adenosine nucleoside triphosphate analogue. The agent was discovered amidst a screening process for antimicrobials with activity against RNA viruses, such as Coronaviridae and Flaviviridae. Research and development of the agent showed promise during the height of the Ebola virus outbreak due to its low EC50 and host polymerase selectivity against the Ebola virus.30 Currently, remdesivir is a promising potential therapy for COVID-19 due to its broad-spectrum, potent in vitro activity against several nCoVs, including SARS-CoV-2 with EC50 and EC90 values of 0.77 μM and 1.76 μM, respectively.31,58 In murine lung infection models with MERS-CoV, remdesivir prevented lung hemorrhage and reduced viral lung titers more than comparator agents.32

The safety and pharmacokinetics of remdesivir were evaluated in single- and multiple-dose phase 1 clinical trials.63 Intravenous infusions between 3 mg and 225 mg were well-tolerated without any evidence of liver or kidney toxicity. Remdesivir demonstrated linear pharmacokinetics within this dose range and an intracellular half-life of greater than 35 hours. Following multiple-dose administrations, reversible aspartate aminotransferase and alanine transaminase elevations occurred. The current dose under investigation is a single 200-mg loading dose, followed by 100-mg daily infusion. No hepatic or kidney adjustments are recommended at this time, but initiation is not recommended in patients with an estimated glomerular filtration rate less than 30 mL/min.

The first clinical use of remdesivir was for the treatment of Ebola64; however, successful case reports describing the use of remdesivir for COVID-19 have been reported.65,66 Clinical trials are ongoing to evaluate the safety and antiviral activity of remdesivir in patients with mild to moderate or severe COVID-19 (NCT04292899, NCT04292730, NCT04257656, NCT04252664, NCT04280705). Of particular importance, the National Institutes of Health is sponsoring an adaptive, randomized, double-blind, placebo-controlled trial that will shed light on the effectiveness of remdesivir compared with supportive care (NCT04280705).40 As the results from RCTs are anticipated, inclusion of this agent for treatment of COVID-19 may be considered. Notably, remdesivir is not currently FDA-approved and must be obtained via compassionate use (only for children <18 years and pregnant women), expanded access, or enrollment in a clinical trial.

Favipiravir

Favipiravir, previously known as T-705, is a prodrug of a purine nucleotide, favipiravir ribofuranosyl-5′-triphosphate. The active agent inhibits the RNA polymerase, halting viral replication. Most of favipiravir’s preclinical data are derived from its influenza and Ebola activity; however, the agent also demonstrated broad activity against other RNA viruses.67 In vitro, the EC50 of favipiravir against SARS-CoV-2 was 61.88 μM/L in Vero E6 cells.58

Various dosing regimens have been proposed based on the type of infectious indication. Dosing variations are likely due to the lower favipiravir EC50 values described against influenza compared with Ebola and SARS-CoV-2.68,69 Doses at the higher end of the dosing range should be considered for the treatment of COVID-19.69 A loading dose is recommended (2400 mg to 3000 mg every 12 hours × 2 doses) followed by a maintenance dose (1200 mg to 1800 mg every 12 hours). The half-life is approximately 5 hours.70 The agent has a mild adverse effect profile and is overall well-tolerated, although the adverse event profile for higher-dose regimens is limited.44,69,71,72 Favipiravir is currently available in Japan for the treatment of influenza, but not available in the United States for clinical use.

Limited clinical experience has been reported supporting the use of favipiravir for COVID-19. In a prospective, randomized, multicenter study, favipiravir (n = 120) was compared with Arbidol (n = 120) for the treatment of moderate and severe COVID-19 infections. Differences in clinical recovery at day 7 were observed in patients with moderate infections (71.4% favipiravir and 55.9% Arbidol, P = .019). No significant differences were observed in the severe or severe and moderate (combined) arms.73 These data support further investigation with RCTs of the efficacy of favipiravir for the treatment of COVID-19.

This review of proposed drugs is by necessity selective. A recent comprehensive review conducted by a division of the American Chemical Society analyzed scientific data related to therapeutic agents and vaccines in human coronaviruses since 2003, using both published literature and patents worldwide.74 This analysis reported more than 130 patents and more than 3000 potential small molecule drug candidates with potential activity against human coronaviruses. The same analysis identified more than 500 patents for biologic agents with activity against coronaviruses including therapeutic antibodies, cytokines, RNA therapies, and vaccines. Another preprint analysis of SARS-CoV-2–human protein-protein interaction maps identified 332 high-confidence protein-protein interactions, yielding 66 candidate druggable human proteins or host factors targeted by either existing FDA-approved or investigational drugs.75 This large amount of potential agents will hopefully yield more candidate therapeutics in the race to find effective treatments or preventive strategies against COVID-19.

Adjunctive Therapies

Quiz Ref IDAt present in the absence of proven therapy for SARS-CoV-2, the cornerstone of care for patients with COVID-19 remains supportive care, ranging from symptomatic outpatient management to full intensive care support. However, 3 adjunctive therapies that warrant special mention are corticosteroids, anticytokine or immunomodulatory agents, and immunoglobulin therapy.

Corticosteroids

The rationale for the use of corticosteroids is to decrease the host inflammatory responses in the lungs, which may lead to acute lung injury and acute respiratory distress syndrome (ARDS). However, this benefit may be outweighed by adverse effects, including delayed viral clearance and increased risk of secondary infection. Although direct evidence for corticosteroids in COVID-19 is limited, reviews of outcomes in other viral pneumonias are instructive.76 Observational studies in patients with SARS and MERS reported no associations of corticosteroids with improved survival, but demonstrated an association with delayed viral clearance from the respiratory tract and blood and high rates of complications including hyperglycemia, psychosis, and avascular necrosis.37,77 Additionally, a 2019 meta-analysis of 10 observational studies with 6548 patients with influenza pneumonia found that corticosteroids were associated with an increased risk of mortality (risk ratio [RR], 1.75 [95% CI, 1.3-2.4]; P < .001) and a 2-fold higher risk of secondary infections (RR, 1.98 [95% CI, 1.0-3.8]; P = .04).78 While the efficacy of corticosteroids in ARDS and septic shock more generally remains debated, Russell and colleagues76 argued that those most likely to benefit from corticosteroids are those with bacterial rather than viral infections. A recent retrospective study of 201 patients with COVID-19 in China found that, for those who developed ARDS, treatment with methylprednisolone was associated with a decreased risk of death (23/50 [46%] with steroids vs 21/34 [62%] without; HR, 0.38 [95% CI, 0.20-0.72]).47 However, the authors noted that bias and residual confounding between those who did or did not receive steroids may exist in this observational study. Therefore, the potential harms and lack of proven benefit for corticosteroids cautions against their routine use in patients with COVID-19 outside an RCT unless a concomitant compelling indication, such as chronic obstructive pulmonary disease exacerbation or refractory shock exists.

Anticytokine or Immunomodulatory Agents

Monoclonal antibodies directed against key inflammatory cytokines or other aspects of the innate immune response represent another potential class of adjunctive therapies for COVID-19. The rationale for their use is that the underlying pathophysiology of significant organ damage in the lungs and other organs is caused by an amplified immune response and cytokine release, or “cytokine storm.”79 IL-6 appears to be a key driver of this dysregulated inflammation based on early case series from China.80 Thus, monoclonal antibodies against IL-6 could theoretically dampen this process and improve clinical outcomes. Tocilizumab, a monoclonal antibody IL-6 receptor antagonist, is FDA approved to treat RA and cytokine release syndrome following chimeric antigen receptor T-cell therapy. Given this experience, tocilizumab has been used in small series of severe COVID-19 cases with early reports of success. A report of 21 patients with COVID-19 showed receipt of tocilizumab, 400 mg, was associated with clinical improvement in 91% of patients as measured by improved respiratory function, rapid defervescence, and successful discharge, with most patients only receiving 1 dose.35 The lack of a comparator group limits the interpretation of the drug-specific effect and warrants caution until more rigorous data are available. Several RCTs of tocilizumab, alone or in combination, in patients with COVID-19 with severe pneumonia are underway in China (NCT04310228, ChiCTR200002976), and it is included in the current Chinese national treatment guidelines.12

Sarilumab, another IL-6 receptor antagonist approved for RA, is being studied in a multicenter, double-blind, phase 2/3 trial for hospitalized patients with severe COVID-19 (NCT04315298).81 Other monoclonal antibody or immunomodulatory agents in clinical trials in China or available for expanded access in the US include bevacizumab (anti–vascular endothelial growth factor medication; NCT04275414), fingolimod (immunomodulator approved for multiple sclerosis; NCT04280588), and eculizumab (antibody inhibiting terminal complement; NCT04288713).40

Immunoglobulin Therapy

Another potential adjunctive therapy for COVID-19 is the use of convalescent plasma or hyperimmune immunoglobulins.82 The rationale for this treatment is that antibodies from recovered patients may help with both free virus and infected cell immune clearance. Anecdotal reports or protocols for convalescent plasma have been reported as salvage therapy in SARS and MERS.83,84 A 2009 prospective observational study in 93 critically ill patients with H1N1 influenza A, 20 of whom received convalescent plasma, demonstrated that receipt of convalescent plasma vs nonreceipt was associated with a reduction in mortality (20% vs 54.8%; P = .01).85 As part of a 2015 systematic review, Mair-Jenkins and colleagues86 conducted a post hoc meta-analysis of 8 observational studies including 714 patients with either SARS or severe influenza. Administration of convalescent plasma and hyperimmune immunoglobulin was associated with reduction in mortality (odds ratio, 0.25 [95% CI, 0.14-0.45]; I2 = 0%) with relatively few harms, although study quality was generally low and at risk of bias.86 In theory, the benefits of this therapy would accrue primarily within the first 7 to 10 days of infection, when viremia is at its peak and the primary immune response has not yet occurred. Although current commercial immunoglobulin preparations likely lack protective antibodies to SARS-CoV-2, this modality warrants further safety and efficacy trials as the pool of patients who have recovered from COVID-19 increases globally. Indeed, the first reported uncontrolled case series of 5 critically ill patients with COVID-19 treated with convalescent plasma in China was recently published.87 Additionally, a case series of 3 patients with COVID-19 in Wuhan, China, treated with intravenous immunoglobulin at a dose of 0.3 to 0.5 g/kg/d for 5 days was recently published.88 On March 24, 2020, the FDA released guidance for requesting an emergency investigational new drug application and screening donors for COVID-19 convalescent plasma.89 There are also early preprint reports describing preclinical development of a human monoclonal antibody against a common epitope to block SARS-COV-2 (and SARS-CoV) infection.90

The most effective long-term strategy for prevention of future outbreaks of this virus would be the development of a vaccine providing protective immunity. However, a minimum of 12 to 18 months would be required before widespread vaccine deployment. A comprehensive review of vaccine research for SARS-CoV-2 is beyond the scope of this review.

Current Clinical Treatment Experience and Recommendations

The published clinical treatment experience, outside the few clinical trials mentioned, mostly consists of descriptive reports and case series from China and other countries affected early in this pandemic. Therefore, outcomes including case-fatality rates must be interpreted with caution given the presence of confounding and selection bias as well as the shifting demographics, testing, and treatment approaches. Table 2 summarizes the clinical severity, complications, treatments, and clinical outcomes from early reported COVID-19 case series.

Quiz Ref IDThe current Centers for Disease Control and Prevention guidance for clinical care of patients with COVID-19 (as of March 7, 2020) highlights that no specific treatment for COVID-19 is available, and emphasizes that management should include “prompt implementation of recommended infection prevention and control measures and supportive management of complications.”96 The guidance from the Centers for Disease Control and Prevention specifically mentions that corticosteroids should be avoided unless indicated for other reasons. Investigational therapeutics, specifically remdesivir, are mentioned as options through either compassionate use or ongoing clinical trials.

Similarly, the current World Health Organization (WHO) clinical management guidance document (as of March 13, 2020) states “there is no current evidence to recommend any specific anti-COVID-19 treatment for patients with confirmed COVID-19.”97 The guidance emphasizes the role of supportive care based on severity of illness, ranging from symptomatic treatment for mild disease to evidence-based ventilatory management for ARDS and early recognition and treatment of bacterial infections and sepsis in critically ill patients. They recommend to “not routinely give systemic corticosteroids for treatment of viral pneumonia outside clinical trials” and state “investigational anti-COVID-19 therapeutics should be used only in approved, randomized, controlled trials.” In this regard, the WHO recently announced plans to launch a global “megatrial” called SOLIDARITY with a pragmatic trial design that will randomize confirmed cases into either standard care or 1 of 4 active treatment arms (remdesivir, chloroquine or hydroxychloroquine, lopinavir/ritonavir, or lopinavir/ritonavir plus interferon-β) based on local drug availability.98

Box 1 provides links to major US and international guidance documents for clinical treatment and other useful resources for drug-drug interactions and guidance in special populations. Box 2 answers frequently asked questions for clinicians about clinical management of patients with COVID-19.

Box Section Ref ID
Box 1.

Clinical Treatment Guidance and Other Useful Resources

International and Select National or Institutional Clinical Management Guidance
Clinical Trials Registries/Resources
Drug-Drug Interaction Websites
Guidance for Special Populations
Box Section Ref ID
Box 2.

COVID-19 Clinical Management: Frequently Asked Questions

1. Have any medical therapies been definitively shown to improve outcomes in a patient with COVID-19?

At this time there are no medical therapies that have been definitively shown to improve outcomes in patients with COVID-19. A number of drugs have demonstrated in vitro activity against the SARS-CoV-2 virus or potential clinical benefits in observational or small, nonrandomized studies. Adequately powered randomized clinical trials are currently enrolling and needed to establish the efficacy of these proposed therapies.

2. Should hydroxychloroquine and/or azithromycin be prescribed for patients with severe symptoms from COVID-19?

The reported clinical benefits of the combination of hydroxychloroquine and azithromycin for patients with COVID-19 come either from media reports or nonrandomized trials with small numbers of participants (<100 patients). The documented benefit of hydroxychloroquine with or without azithromycin is very limited, especially in severe disease. While these medications, individually or in combination, may prove efficacious, these benefits need to be established with randomized clinical trials prior to widespread adoption of these treatments.

3. Should I stop ARBs/ACE inhibitors in my older patients and those at high risk for severe illness from COVID-19?

Major institutions and societies, including the Centers for Disease Control and Prevention, the American Heart Association, the Heart Failure Society of America, and the American College of Cardiology recommend continuation of ACE inhibitors or ARB medications for all patients already prescribed those medications for another indication. There is currently no human evidence establishing a link between the use of these medications with an increased risk of COVID-19 acquisition or illness severity.

4. What is the role of immunomodulatory drugs such as IL-6 receptor antagonists or corticosteroids in the management of patients with COVID-19?

Given the important role the immune response plays in the complications of COVID-19, active clinical trials are evaluating immunomodulatory drugs (such as IL-6 receptor antagonists) in this disease. In patients with “cytokine storm,” characterized by marked elevation in inflammatory markers, use of IL-6 receptor antagonists can be considered, preferably in the context of a clinical trial, although these medications can increase risk of secondary infections. The role of corticosteroids remains controversial, and current guidelines from the World Health Organization do not recommend their use unless another concomitant indication exists such as chronic obstructive pulmonary disease exacerbation or pressor-refractory shock. However, their utility in patients with severe COVID-19 with acute respiratory distress syndrome should be further investigated in clinical trials.

5. Which medications have been repurposed to treat COVID-19?

Numerous agents demonstrate in vitro activity against novel coronaviruses, including SARS-CoV-2. Small molecule database screens identified thousands of potential agents. Of these, several repurposed agents used to treat a variety of other disease states (eg, HIV and autoimmune diseases) have been proposed as possible treatment options for COVID-19. Lopinavir/ritonavir and chloroquine or hydroxychloroquine are the medications with the most clinical evidence, either positive or negative, in the treatment of COVID-19. To date, available clinical trials have not demonstrated that any of these drugs are clearly effective.

6. Are there investigational drugs available to treat COVID-19?

Remdesivir is available to COVID-19–infected patients through enrollment in a clinical trial or application for emergency access. In the United States, there are 3 ongoing clinical trials differentiated by severity of disease (eg, moderate vs severe infection) and study design (eg, placebo-controlled). Emergency access is available through an expanded access program. Sites without access to a clinical trial may obtain the drug this way. Also, individual compassionate use for pregnant women and children younger than 18 years of age with confirmed COVID-19 and severe manifestations of the disease may obtain the drug in this manner. Favipiravir is not currently available in the United States.

7. How do I decide if a patient with COVID-19 needs a specific treatment or should receive only supportive care?

The priority should be to enroll a patient in a clinical trial if they qualify. If this is not possible, patients who are stable as an outpatient or have no evidence of oxygen requirement or pneumonia by imaging can generally be managed with supportive care alone. Patients who have evidence of hypoxia or pneumonia, especially those with risk factors for disease progression such as age older than 65 years, cardiac or pulmonary comorbidities, and immunosuppression, can be considered for specific COVID-19 therapy after discussing the risks and benefits with the patient and in accordance with local hospital treatment guidance. 

8. What are the limitations of repurposing medications to treat COVID-19?

The use of repurposed medications relies on the assumption that the benefits (in vitro/clinical evidence) outweigh associated risks (adverse drug reactions). One limitation to using repurposed agents is the propensity of these agents to cause acute toxicity. This acute toxicity may outweigh the undefined benefit of a specific antiviral agent. Augmented toxicity with combination therapy, such as heart or liver toxicity, creates potential additional risk and need for close risk vs benefit analysis. Overall, the paucity of evidence demonstrating a clear benefit may not justify the risk of the repurposed agent(s). This is of upmost concern in patients at high risk for toxicity and in situations where adverse events may preclude entry into investigational trials.

ACE indicates angiotensin-converting enzyme; ARB, angiotensin receptor blocker; COVID-19, coronavirus disease 2019; and SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

Limitations

This review has several limitations to note. First, the tremendous volume and fast pace of published literature on the treatment of COVID-19 means that research findings and recommendations are constantly evolving as new evidence arises. Second, the published treatment data to date derive exclusively from observational data or small clinical trials (none with more than 250 patients), introducing higher risks of bias or imprecision regarding the magnitude of treatment effect size. Third, our review focused only on adult patients and the data may not be applicable to pediatric populations. Fourth, the articles were limited to English-language publications or translations so relevant international data could be lacking.

Conclusions

The COVID-19 pandemic represents the greatest global public health crisis of this generation and, potentially, since the pandemic influenza outbreak of 1918. The speed and volume of clinical trials launched to investigate potential therapies for COVID-19 highlight both the need and capability to produce high-quality evidence even in the middle of a pandemic. No therapies have been shown effective to date.

Section Editors: Edward Livingston, MD, Deputy Editor, and Mary McGrae McDermott, MD, Deputy Editor.
Submissions: We encourage authors to submit papers for consideration as a Review. Please contact Edward Livingston, MD, at Edward.livingston@jamanetwork.org or Mary McGrae McDermott, MD, at mdm608@northwestern.edu.
Back to top
Article Information

Corresponding Author: James B. Cutrell, MD, Division of Infectious Diseases and Geographic Medicine, Department of Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9113 (james.cutrell@utsouthwestern.edu).

Accepted for Publication: April 3, 2020.

Published Online: April 13, 2020. doi:10.1001/jama.2020.6019

Author Contributions: Dr Cutrell had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Concept and design: All authors.

Acquisition, analysis, or interpretation of data: Monogue, Jodlowski, Cutrell.

Drafting of the manuscript: All authors.

Critical revision of the manuscript for important intellectual content: Monogue, Jodlowski, Cutrell.

Administrative, technical, or material support: Cutrell.

Supervision: Cutrell.

Conflict of Interest Disclosures: Dr Cutrell reported receiving nonfinancial support from Regeneron and Gilead outside the submitted work. No other disclosures were reported.

Additional Contributions: We acknowledge our infectious disease physician and pharmacy colleagues at UT Southwestern and its respective hospital sites, Clements University Hospital, Parkland Hospital, and the VA North Texas Health Care System for their thoughtful discussions regarding COVID-19 clinical management.

References
1.
Zhu  N, Zhang  D, Wang  W,  et al; China Novel Coronavirus Investigating and Research Team.  A novel coronavirus from patients with pneumonia in China, 2019.   N Engl J Med. 2020;382(8):727-733. doi:10.1056/NEJMoa2001017 PubMedGoogle ScholarCrossref
2.
Chinese Clinical Trials. http://www/chictr.org/enindex.aspx. Accessed March 31, 2020.
3.
Hoffmann  M, Kleine-Weber  H, Schroeder  S,  et al.  SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor.   Cell. Published online March 4, 2020. doi:10.1016/j.cell.2020.02.052 PubMedGoogle Scholar
4.
Chen  Y, Liu  Q, Guo  D.  Emerging coronaviruses: genome structure, replication, and pathogenesis.   J Med Virol. 2020;92(4):418-423. doi:10.1002/jmv.25681 PubMedGoogle ScholarCrossref
5.
Fehr  AR, Perlman  S.  Coronaviruses: an overview of their replication and pathogenesis.   Methods Mol Biol. 2015;1282:1-23. doi:10.1007/978-1-4939-2438-7_1 PubMedGoogle ScholarCrossref
6.
Fung  TS, Liu  DX.  Coronavirus infection, ER stress, apoptosis and innate immunity.   Front Microbiol. 2014;5:296. doi:10.3389/fmicb.2014.00296 PubMedGoogle ScholarCrossref
7.
Savarino  A, Boelaert  JR, Cassone  A, Majori  G, Cauda  R.  Effects of chloroquine on viral infections: an old drug against today’s diseases?   Lancet Infect Dis. 2003;3(11):722-727. doi:10.1016/S1473-3099(03)00806-5 PubMedGoogle ScholarCrossref
8.
Al-Bari  MAA.  Targeting endosomal acidification by chloroquine analogs as a promising strategy for the treatment of emerging viral diseases.   Pharmacol Res Perspect. 2017;5(1):e00293. doi:10.1002/prp2.293 PubMedGoogle Scholar
9.
Zhou  D, Dai  SM, Tong  Q.  COVID-19: a recommendation to examine the effect of hydroxychloroquine in preventing infection and progression.  [published online March 20, 2020].  J Antimicrob Chemother. 2020;dkaa114. doi:10.1093/jac/dkaa114 PubMedGoogle Scholar
10.
Devaux  CA, Rolain  JM, Colson  P, Raoult  D.  New insights on the antiviral effects of chloroquine against coronavirus: what to expect for COVID-19?   Int J Antimicrob Agents. Published online March 11, 2020. doi:10.1016/j.ijantimicag.2020.105938 PubMedGoogle Scholar
11.
Colson  P, Rolain  JM, Lagier  JC, Brouqui  P, Raoult  D.  Chloroquine and hydroxychloroquine as available weapons to fight COVID-19.   Int J Antimicrob Agents. Published online March 4, 2020. doi:10.1016/j.ijantimicag.2020.105932 PubMedGoogle Scholar
12.
National Health Commission and State Administration of Traditional Chinese Medicine. Diagnosis and treatment protocol for novel coronavirus pneumonia. Accessed March 18, 2020. https://www.chinalawtranslate.com/wp-content/uploads/2020/03/Who-translation.pdf
13.
Chloroquine [database online]. Hudson, OH: Lexicomp Inc; 2016. Accessed March 17, 2020. http://online.lexi.com
14.
Aralen (chloroquine phosphate) [package insert]. Bridgewater, NJ: Sanofi-Aventis; 2008. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/006002s045lbl.pdf
15.
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. Published online March 9, 2020. doi:10.1093/cid/ciaa237 PubMedGoogle Scholar
16.
Gautret  P, Lagier  JC, 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. Published online March 20, 2020. doi:10.1016/j.ijantimicag.2020.105949 PubMedGoogle Scholar
17.
Chen  J, Liu  D, Liu  L,  et al.  A pilot study of hydroxychloroquine in treatment of patients with common coronavirus disease-19 (COVID-19).   J Zhejiang Univ (Med Sci). Published online March 6, 2020. doi:10.3785/j.issn.1008-9292.2020.03.03Google Scholar
18.
Hydroxychloroquine [database online]. Hudson, OH: Lexicomp Inc; 2016. Accessed March 17, 2020. http://online.lexi.com
19.
Plaquenil (Hydroxychloroquine sulfate) [package insert]. St Michael, Barbados: Concordia Pharmaceuticals Inc; 2018. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/009768Orig1s051lbl.pdf
20.
Lim  HS, Im  JS, Cho  JY,  et al.  Pharmacokinetics of hydroxychloroquine and its clinical implications in chemoprophylaxis against malaria caused by Plasmodium vivax.   Antimicrob Agents Chemother. 2009;53(4):1468-1475. doi:10.1128/AAC.00339-08 PubMedGoogle ScholarCrossref
21.
Chu  CM, Cheng  VC, Hung  IF,  et al; HKU/UCH SARS Study Group.  Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings.   Thorax. 2004;59(3):252-256. doi:10.1136/thorax.2003.012658 PubMedGoogle ScholarCrossref
22.
de Wilde  AH, Jochmans  D, Posthuma  CC,  et al.  Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture.   Antimicrob Agents Chemother. 2014;58(8):4875-4884. doi:10.1128/AAC.03011-14 PubMedGoogle ScholarCrossref
23.
Cao  B, Wang  Y, Wen  D,  et al.  A trial of lopinavir-ritonavir in adults hospitalized with severe COVID-19.   N Engl J Med. Published online March 18, 2020. doi:10.1056/NEJMoa2001282 PubMedGoogle Scholar
24.
Lopinavir/ritonavir [database online]. Hudson (OH): Lexicomp Inc; 2016. Accessed March 17, 2020. http://online.lexi.com
25.
Kaletra (Lopinavir and ritonavir) [package insert]. North Chicago, IL: Abbvie; 2019. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021226s048lbl.pdf
26.
Department of Health and Human Services Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Accessed March 17, 2020. http://www.aidsinfo.nih.gov/ContentFiles/ AdultandAdolescentGL.pdf
27.
Kadam  RU, Wilson  IA.  Structural basis of influenza virus fusion inhibition by the antiviral drug Arbidol.   Proc Natl Acad Sci U S A. 2017;114(2):206-214. doi:10.1073/pnas.1617020114 PubMedGoogle ScholarCrossref
28.
Khamitov  RA, Loginova  SIa, Shchukina  VN, Borisevich  SV, Maksimov  VA, Shuster  AM.  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(4):9-13.PubMedGoogle Scholar
29.
Wang  Z, Yang  B, Li  Q, Wen  L, Zhang  R.  Clinical Features of 69 cases with coronavirus disease 2019 in Wuhan, China.   Clin Infect Dis. Published online March 16, 2020. doi:10.1093/cid/ciaa272 PubMedGoogle Scholar
30.
Siegel  D, Hui  HC, Doerffler  E,  et al.  Discovery and synthesis of a phosphoramidate prodrug of a pyrrolo[2,1-f][triazin-4-amino] adenine C-nucleoside (GS-5734) for the treatment of Ebola and emerging viruses.   J Med Chem. 2017;60(5):1648-1661. doi:10.1021/acs.jmedchem.6b01594 PubMedGoogle ScholarCrossref
31.
Al-Tawfiq  JA, Al-Homoud  AH, Memish  ZA.  Remdesivir as a possible therapeutic option for the COVID-19.   Travel Med Infect Dis. Published online March 5, 2020. doi:10.1016/j.tmaid.2020.101615 PubMedGoogle Scholar
32.
Sheahan  TP, Sims  AC, Leist  SR,  et al.  Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV.   Nat Commun. 2020;11(1):222. doi:10.1038/s41467-019-13940-6 PubMedGoogle ScholarCrossref
33.
Hayden  FG, Shindo  N.  Influenza virus polymerase inhibitors in clinical development.   Curr Opin Infect Dis. 2019;32(2):176-186. doi:10.1097/QCO.0000000000000532 PubMedGoogle ScholarCrossref
34.
Avigan (favipiravir) [package insert]. Tokyo, Japan: Taisho Toyama Pharmaceutical Co Ltd; 2017, 4th version. Accessed March 25, 2020.
35.
Xu X, Han M, Li T, et al. Effective treatment of severe COVID-19 patients with tocilizumab. chinaXiv. Preprint posted March 5, 2020. doi:10.12074/202003.00026
36.
Actemra (tocilizumab) [package insert]. South San Francisco, CA: Genentech, Inc; 2019. Accessed March 17, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/125276s127,125472s040lbl.pdf
37.
Stockman  LJ, Bellamy  R, Garner  P.  SARS: systematic review of treatment effects.   PLoS Med. 2006;3(9):e343. doi:10.1371/journal.pmed.0030343 PubMedGoogle Scholar
38.
Morra  ME, Van Thanh  L, Kamel  MG,  et al.  Clinical outcomes of current medical approaches for Middle East respiratory syndrome: a systematic review and meta-analysis.   Rev Med Virol. 2018;28(3):e1977. doi:10.1002/rmv.1977 PubMedGoogle Scholar
39.
Gao  J, Tian  Z, Yang  X.  Breakthrough: chloroquine phosphate has shown apparent efficacy in treatment of COVID-19 associated pneumonia in clinical studies.   Biosci Trends. 2020;14(1):72-73. doi:10.5582/bst.2020.01047 PubMedGoogle ScholarCrossref
40.
ClinicalTrials.gov. Accessed March 18, 2020. https://clinicaltrials.gov/
41.
Kalil  AC.  Treating COVID-19—off-label drug use, compassionate use, and randomized clinical trials during pandemics.   JAMA. Published March 24, 2020. doi:10.1001/jama.2020.4742 PubMedGoogle Scholar
42.
Interview with David Juurlink.  Coronavirus (COVID-19) update: chloroquine/hydroxychloroquine and azithromycin.   JAMA. March 24, 2020. Accessed April 3, 2020. https://edhub.ama-assn.org/jn-learning/audio-player/18337225Google Scholar
43.
Osadchy  A, Ratnapalan  T, Koren  G.  Ocular toxicity in children exposed in utero to antimalarial drugs: review of the literature.   J Rheumatol. 2011;38(12):2504-2508. doi:10.3899/jrheum.110686 PubMedGoogle ScholarCrossref
44.
Dong  L, Hu  S, Gao  J.  Discovering drugs to treat coronavirus disease 2019 (COVID-19).   Drug Discov Ther. 2020;14(1):58-60. doi:10.5582/ddt.2020.01012 PubMedGoogle ScholarCrossref
45.
Yao  TT, Qian  JD, Zhu  WY, Wang  Y, Wang  GQ.  A systematic review of lopinavir therapy for SARS coronavirus and MERS coronavirus-A possible reference for coronavirus disease-19 treatment option.  [published online February 27, 2020].  J Med Virol. 2020. doi:10.1002/jmv.25729 PubMedGoogle Scholar
46.
Chan  KS, Lai  ST, Chu  CM,  et al.  Treatment of severe acute respiratory syndrome with lopinavir/ritonavir: a multicentre retrospective matched cohort study.   Hong Kong Med J. 2003;9(6):399-406.PubMedGoogle Scholar
47.
Wu  C, Chen  X, Cai  Y,  et al.  Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China.   JAMA Intern Med. Published online March 13, 2020. PubMedGoogle Scholar
48.
Foolad  F, Aitken  SL, Shigle  TL,  et al.  Oral versus aerosolized ribavirin for the treatment of respiratory syncytial virus infections in hematopoietic cell transplant recipients.   Clin Infect Dis. 2019;68(10):1641-1649. doi:10.1093/cid/ciy760 PubMedGoogle ScholarCrossref
49.
Arabi  YM, 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. Published online June 25, 2019. doi:10.1093/cid/ciz544 PubMedGoogle Scholar
50.
Altınbas  S, Holmes  JA, Altınbas  A.  Hepatitis C virus infection in pregnancy: an update.   Gastroenterol Nurs. 2020;43(1):12-21. doi:10.1097/SGA.0000000000000404 PubMedGoogle ScholarCrossref
51.
Wang  D, Hu  B, Hu  C,  et al.  Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China.   JAMA. Published online February 7, 2020. doi:10.1001/jama.2020.1585 PubMedGoogle Scholar
52.
Totura  AL, Bavari  S.  Broad-spectrum coronavirus antiviral drug discovery.   Expert Opin Drug Discov. 2019;14(4):397-412. doi:10.1080/17460441.2019.1581171 PubMedGoogle ScholarCrossref
53.
Li  G, De Clercq  E.  Therapeutic options for the 2019 novel coronavirus (2019-nCoV).   Nat Rev Drug Discov. 2020;19(3):149-150. doi:10.1038/d41573-020-00016-0 PubMedGoogle ScholarCrossref
54.
Coleman  CM, Sisk  JM, Mingo  RM, Nelson  EA, White  JM, Frieman  MB.  Abelson kinase inhibitors are potent inhibitors of severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome coronavirus fusion.   J Virol. 2016;90(19):8924-8933. doi:10.1128/JVI.01429-16 PubMedGoogle ScholarCrossref
55.
Dyall  J, Gross  R, Kindrachuk  J,  et al.  Middle East respiratory syndrome and severe acute respiratory syndrome: current therapeutic options and potential targets for novel therapies.   Drugs. 2017;77(18):1935-1966. doi:10.1007/s40265-017-0830-1 PubMedGoogle ScholarCrossref
56.
Pfefferle  S, Schöpf  J, Kögl  M,  et al.  The SARS-coronavirus-host interactome: identification of cyclophilins as target for pan-coronavirus inhibitors.   PLoS Pathog. 2011;7(10):e1002331. doi:10.1371/journal.ppat.1002331 PubMedGoogle Scholar
57.
de Wilde  AH, Zevenhoven-Dobbe  JC, van der Meer  Y,  et al.  Cyclosporin A inhibits the replication of diverse coronaviruses.   J Gen Virol. 2011;92(pt 11):2542-2548. doi:10.1099/vir.0.034983-0 PubMedGoogle ScholarCrossref
58.
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(3):269-271. doi:10.1038/s41422-020-0282-0 PubMedGoogle ScholarCrossref
59.
Rossignol  JF.  Nitazoxanide, a new drug candidate for the treatment of Middle East respiratory syndrome coronavirus.   J Infect Public Health. 2016;9(3):227-230. doi:10.1016/j.jiph.2016.04.001 PubMedGoogle ScholarCrossref
60.
Gurwitz  D.  Angiotensin receptor blockers as tentative SARS-CoV-2 therapeutics.   Drug Dev Res. Published online March 4, 2020. doi:10.1002/ddr.21656 PubMedGoogle Scholar
61.
American Heart Association. Patients taking angiotensin converting enzyme inhibitors (ACE-i) or angiotensin receptor blocker (ARB) medications should continue therapy as prescribed [news release]. Published March 17, 2020. Accessed March 18, 2020. https://newsroom.heart.org/news/patients-taking-ace-i-and-arbs-who-contract-covid-19-should-continue-treatment-unless-otherwise-advised-by-their-physician
62.
European Society for Cardiology. Position statement of the ESC Council on Hypertension on ACE-Inhibitors and Angiotensin Receptor Blockers. Published March 13, 2020. Accessed March 18, 2020. https://www.escardio.org/Councils/Council-on-Hypertension-(CHT)/News/position-statement-of-the-esc-council-on-hypertension-on-ace-inhibitors-and-ang
63.
World Health Organization. WHO R&D blueprint: ad-hoc expert consultation on clinical trials for Ebola therapeutics. Published October 2018. Accessed March 20, 2020. https://www.who.int/ebola/drc-2018/summaries-of-evidence-experimental-therapeutics.pdf
64.
Jacobs  M, Rodger  A, Bell  DJ,  et al.  Late Ebola virus relapse causing meningoencephalitis: a case report.   Lancet. 2016;388(10043):498-503. doi:10.1016/S0140-6736(16)30386-5 PubMedGoogle ScholarCrossref
65.
Holshue  ML, DeBolt  C, Lindquist  S,  et al; Washington State 2019-nCoV Case Investigation Team.  First case of 2019 novel coronavirus in the United States.   N Engl J Med. 2020;382(10):929-936. doi:10.1056/NEJMoa2001191 PubMedGoogle ScholarCrossref
66.
Kujawski  SA, Wong  K, Collins  JP,  et al. First 12 patients with coronavirus disease 2019 (COVID-19) in the United States. medRxiv. Preprint posted March 9, 2020. doi:10.1101/2020.03.09.20032896
67.
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(7):449-463. doi:10.2183/pjab.93.027 PubMedGoogle ScholarCrossref
68.
Mentré  F, Taburet  AM, Guedj  J,  et al.  Dose regimen of favipiravir for Ebola virus disease.   Lancet Infect Dis. 2015;15(2):150-151. doi:10.1016/S1473-3099(14)71047-3 PubMedGoogle ScholarCrossref
69.
Sissoko  D, Laouenan  C, Folkesson  E,  et al; JIKI Study Group.  Experimental treatment with favipiravir for Ebola virus disease (the JIKI Trial): a historically controlled, single-arm proof-of-concept trial in Guinea  [published correction appears in PLoS Med. 2016;13(4):e1002009].  PLoS Med. 2016;13(3):e1001967. doi:10.1371/journal.pmed.1001967 PubMedGoogle Scholar
70.
Shiraki  K, Daikoku  T.  Favipiravir, an anti-influenza drug against life-threatening RNA virus infections.  [published online February 22, 2020].  Pharmacol Ther. 2020;107512. doi:10.1016/j.pharmthera.2020.107512 PubMedGoogle Scholar
71.
Chinello  P, Petrosillo  N, Pittalis  S, Biava  G, Ippolito  G, Nicastri  E; INMI Ebola Team.  QTc interval prolongation during favipiravir therapy in an Ebolavirus-infected patient.   PLoS Negl Trop Dis. 2017;11(12):e0006034. doi:10.1371/journal.pntd.0006034 PubMedGoogle Scholar
72.
Kumagai  Y, Murakawa  Y, Hasunuma  T,  et al.  Lack of effect of favipiravir, a novel antiviral agent, on QT interval in healthy Japanese adults.   Int J Clin Pharmacol Ther. 2015;53(10):866-874. doi:10.5414/CP202388 PubMedGoogle ScholarCrossref
73.
Chen  C, Huang  J, Cheng  Z,  et al. Favipiravir versus Arbidol for COVID-19: a randomized clinical trial. medRxiv. Preprint posted March 27, 2020. doi:10.1101/2020.03.17.20037432
74.
Liu  C, Zhou  Q, Li  Y,  et al.  Research and development of therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases.   ACS Cent Sci. 2020;6(3):315-331. doi:10.1021/acscentsci.0c00272 PubMedGoogle ScholarCrossref
75.
Gordon DE, Jang GM, Bouhaddou M, et al. A SARS-CoV-2-human protein-protein interaction map reveals drug targets and potential drug-repurposing. bioRxiv. Preprint posted March 22, 2020. doi:10.1101/2020.03.22.002386
76.
Russell  CD, Millar  JE, Baillie  JK.  Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury.   Lancet. 2020;395(10223):473-475. doi:10.1016/S0140-6736(20)30317-2 PubMedGoogle ScholarCrossref
77.
Arabi  YM, Mandourah  Y, Al-Hameed  F,  et al; Saudi Critical Care Trial Group.  Corticosteroid therapy for critically ill patients with Middle East respiratory syndrome.   Am J Respir Crit Care Med. 2018;197(6):757-767. doi:10.1164/rccm.201706-1172OC PubMedGoogle ScholarCrossref
78.
Ni  YN, Chen  G, Sun  J, Liang  BM, Liang  ZA.  The effect of corticosteroids on mortality of patients with influenza pneumonia: a systematic review and meta-analysis.   Crit Care. 2019;23(1):99. doi:10.1186/s13054-019-2395-8 PubMedGoogle ScholarCrossref
79.
Mehta  P, McAuley  DF, Brown  M, Sanchez  E, Tattersall  RS, Manson  JJ; HLH Across Speciality Collaboration, UK.  COVID-19: consider cytokine storm syndromes and immunosuppression.   Lancet. 2020;395(10229):1033-1034. doi:10.1016/S0140-6736(20)30628-0 PubMedGoogle ScholarCrossref
80.
Zhou  F, Yu  T, Du  R,  et al.  Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study.   Lancet. 2020;395(10229):1054-1062. doi:10.1016/S0140-6736(20)30566-3 PubMedGoogle ScholarCrossref
81.
Sanofi. Sanofi and Regeneron begin global Kevzara (sarilumab) clinical trial program in patients with severe COVID-19 [news release]. Published March 16, 2020. Accessed March 18, 2020. http://www.news.sanofi.us/2020-03-16-Sanofi-and-Regeneron-begin-global-Kevzara-R-sarilumab-clinical-trial-program-in-patients-with-severe-COVID-19
82.
Chen  L, Xiong  J, Bao  L, Shi  Y.  Convalescent plasma as a potential therapy for COVID-19.   Lancet Infect Dis. 2020;20(4):398-400. doi:10.1016/S1473-3099(20)30141-9 PubMedGoogle ScholarCrossref
83.
Soo  YO, Cheng  Y, Wong  R,  et al.  Retrospective comparison of convalescent plasma with continuing high-dose methylprednisolone treatment in SARS patients.   Clin Microbiol Infect. 2004;10(7):676-678. doi:10.1111/j.1469-0691.2004.00956.x PubMedGoogle ScholarCrossref
84.
Arabi  Y, Balkhy  H, Hajeer  AH,  et al.  Feasibility, safety, clinical, and laboratory effects of convalescent plasma therapy for patients with Middle East respiratory syndrome coronavirus infection: a study protocol.   Springerplus. 2015;4:709. doi:10.1186/s40064-015-1490-9 PubMedGoogle ScholarCrossref
85.
Hung  IF, To  KK, Lee  CK,  et al.  Convalescent plasma treatment reduced mortality in patients with severe pandemic influenza A (H1N1) 2009 virus infection.   Clin Infect Dis. 2011;52(4):447-456. doi:10.1093/cid/ciq106 PubMedGoogle ScholarCrossref
86.
Mair-Jenkins  J, Saavedra-Campos  M, Baillie  JK,  et al; Convalescent Plasma Study Group.  The effectiveness of convalescent plasma and hyperimmune immunoglobulin for the treatment of severe acute respiratory infections of viral etiology: a systematic review and exploratory meta-analysis.   J Infect Dis. 2015;211(1):80-90. doi:10.1093/infdis/jiu396 PubMedGoogle ScholarCrossref
87.
Shen  C, Wang  Z, Zhao  F,  et al.  Treatment of 5 critically ill patients with COVID-19 with convalescent plasma.   JAMA. 2020. Published online March 27, 2020. doi:10.1001/jama.2020.4783PubMedGoogle Scholar
88.
Cao  W, Liu  X, Bai  T,  et al.  High-dose intravenous immunoglobulin as a therapeutic option for deteriorating patients with coronavirus disease 2019.   Open Forum Infect Dis. Published online March 21, 2020. doi:10.1093/ofid/ofaa102 Google Scholar
89.
US Food and Drug Administration. Investigational COVID-19 Convalescent plasma: emergency INDs. Updated April 3, 2020. Accessed March 26, 2020. https://www.fda.gov/vaccines-blood-biologics/investigational-new-drug-ind-or-device-exemption-ide-process-cber/investigational-covid-19-convalescent-plasma-emergency-inds
90.
Wang C, Li W, Drabek D, et al. A human monoclonal antibody blocking SARS-CoV-2 infection. bioRxiv. Preprint posted March 11, 2020. doi:10.1101/2020.03.11.987958.2020
91.
Huang  C, Wang  Y, Li  X,  et al.  Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China.   Lancet. 2020;395(10223):497-506. doi:10.1016/S0140-6736(20)30183-5 PubMedGoogle ScholarCrossref
92.
Chen  N, Zhou  M, Dong  X,  et al.  Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study.   Lancet. 2020;395(10223):507-513. doi:10.1016/S0140-6736(20)30211-7 PubMedGoogle ScholarCrossref
93.
Yang  X, Yu  Y, Xu  J,  et al.  Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study.   Lancet Respir Med. Published online February 24, 2020. doi:10.1016/S2213-2600(20)30079-5 PubMedGoogle Scholar
94.
Young  BE, Ong  SWX, Kalimuddin  S,  et al; Singapore 2019 Novel Coronavirus Outbreak Research Team.  Epidemiologic features and clinical course of patients infected with SARS-CoV-2 in Singapore.   JAMA. Published online March 3, 2020. doi:10.1001/jama.2020.3204 PubMedGoogle Scholar
95.
Guan  WJ, Ni  ZY, Hu  Y,  et al; China Medical Treatment Expert Group for Covid-19.  Clinical Characteristics of Coronavirus Disease 2019 in China.   N Engl J Med. Published online February 28, 2020. doi:10.1056/NEJMoa2002032 PubMedGoogle Scholar
96.
Centers for Disease Control and Prevention. Coronavirus disease 2019 (COVID-19) clinical care. Updated March 30, 2020. Accessed March 18, 2020. https://www.cdc.gov/coronavirus/2019-ncov/hcp/clinical-guidance-management-patients.html
97.
World Health Organization. Clinical management of severe acute respiratory infection when COVID-19 is suspected. Updated March 13, 2020. Accessed March 18, 2020. https://www.who.int/publications-detail/clinical-management-of-severe-acute-respiratory-infection-when-novel-coronavirus-(ncov)-infection-is-suspected
98.
Kupferschmidt  K, Cohen  J. WHO launches global megatrial of the four most promising coronavirus treatments. Science. Published March 22, 2020. Accessed March 23, 2020. https://www.sciencemag.org/news/2020/03/who-launches-global-megatrial-four-most-promising-coronavirus-treatments#