Abstract
Novel 2019 coronavirus (severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2]) and coronavirus disease 2019 (COVID-19), the respiratory syndrome it causes, have shaken the world to its core by infecting and claiming the lives of many people since originating in December 2019 in Wuhan, China. World Health Organization and several states have declared a pandemic situation and state of emergency, respectively. As there is no treatment for COVID-19, several research institutes and pharmaceutical companies are racing to find a cure. Advances in computational approaches have allowed the screening of massive antiviral compound libraries to identify those that may potentially work against SARS-CoV-2. Antiviral agents developed in the past to combat other viruses are being repurposed. At the same time, new vaccine candidates are being developed and tested in preclinical/clinical settings. This review provides a detailed overview of select repurposed drugs, their mechanism of action, associated toxicities, and major clinical trials involving these agents.
Keywords: COVID-19, severe acute respiratory syndrome coronavirus 2, remdesivir, hydroxychloroquine, angiotensin-converting enzyme 2, lopinavir
Impact statement
In the wake of the coronavirus disease 2019 (COVID-19) pandemic that originated in late December 2019, antiviral therapies are urgently needed for better patient outcomes and to reduce the burden on healthcare systems across the world. While the vaccines have been developed to mitigate this crisis, there are significant delays in their distribution and administration due to the holiday season, lack of sufficient hospital staff, and short half-life of messenger RNA vaccines. In this review, we have discussed the antiviral agents that inhibit key enzymes facilitating viral propagation. We now know that viral spike protein is primed by the transmembrane protease, serine 2 (TMPRSS2) enzyme, and it binds to the angiotensin-converting enzyme 2 (ACE2) receptor for entry into the host cell. TMPRSS2 and ACE2 blockers have been outlined here as potential strategies to prevent viral entry into cells.
Introduction
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in a seafood market located in Wuhan, China in December 2019 and is the cause of novel coronavirus disease 2019 (COVID-19). 1 Within a short span of 10 months, >106 million people were infected and >2.3 million died due to the virus worldwide. 2 Having descended from bats, SARS-CoV-2 possesses similarities with bat coronaviruses, SARS-CoV, and Middle East respiratory syndrome coronavirus (MERS-CoV), in that order, with possible involvement of a seemingly unknown intermediate host.3,4
Early, rapid sequencing of the viral genome allowed the identification of possible drug targets and prompted subsequent efforts to develop potential intervention strategies. According to Clinicaltrials.gov and World Health Organization (WHO) Trial Registry Network, over 3700 clinical trials in total (including interventional and observational studies) are currently ongoing at various hospitals across the world (Figure 1). In this review, we discuss select interventional strategies against COVID-19, mechanism of action/adverse effects of drugs, and relevant clinical trials. Some classes of agents described here include (1) antiviral agents that may improve survival odds for a patient, (2) receptor blockers that may prevent viral entry into the host cell, and (3) corticosteroids that may reduce inflammatory response by the body, thereby lowering the risk of lung injury. 5
Figure 1.
Clinical trials involving pharmacological treatments against COVID-19. Clinical trials posted on clinicaltrials.gov were sorted by month based on the “First posted” date. The logarithmic scale shows the number of ongoing clinical trials for each repurposed drug. (A color version of this figure is available in the online journal.)
ACEi: angiotensin-converting enzyme inhibitor; ARBs: angiotensin II receptor blockers.
Antiviral agents
Remdesivir
Remdesivir interferes with viral RNA replication by serving as a nucleotide monophosphate analog and inhibiting RNA-dependent RNA polymerase (RdRP) activity. 6 Originally developed during the Ebola outbreak, remdesivir (GS-5734) has shown profound activity against RNA viruses such as SARS-CoV and MERS-CoV.7,8 Upon remdesivir treatment, reduced lung viral titers and lung damage were observed in rhesus macaque and murine models of MERS-CoV.9,10 Remdesivir became a popular treatment option for viral infection due to its extremely low half-maximal effective concentration (EC50) against Ebola (86 nM; in macrophages), 11 and now against SARS-CoV-2 (0.77 μM; in in vitro models).12,13
Clinical trials to determine the safety and efficacy of remdesivir in COVID-19 patients have been completed and demonstrated efficacy. The federally funded Adaptive COVID-19 Treatment Trial (ACTT-1) (NCT04280705; Table 1) assessed the potential of remdesivir to reduce the time to recovery in COVID-19 patients. 6 The patients in the treated group were administered a 200-mg loading dose and subsequent daily doses of 100 mg intravenously. After 10 days of following this regime, the time to recovery was found to be 11 days in the treated group as opposed to 15 days in the placebo group. 6 Given the demonstrated efficacy, subsequent trials of antiviral therapies were evaluated on the background of remdesivir treatment. 14 An additional trial (NCT04292899; Table 1) showed 5-day treatment was equivalent to 10-day treatment of remdesivir in hospitalized COVID-19 patients. 15 Other clinical trials tested variable doses of remdesivir (3–225 mg) and reported a half-life of >35 h within the cell, without any evident toxicity. 16 An international study with over 11,000 patients led by the WHO Solidarity Trial Consortium looked to evaluate 4 separate antiviral therapies in hospitalized COVID-19 patients and found no benefit in mortality of any of the treatments in this intention-to-treat trial though it was not well-structured to assess clinical improvement. 17 With the additional review of adverse events observed such as anemia, acute kidney injury, pyrexia, hyperglycemia, and elevated aminotransferase levels, the U.S. Food and Drug Administration (FDA) ultimately issued an emergency use authorization (EUA) for the use of remdesivir in patients over the age of 12 for the treatment of COVID-19 requiring hospitalization on 22 October 2020, becoming the first therapy approved for the treatment of COVID-19.
Table 1.
Overview of select COVID-19 treatments in clinical development.
| Class | Intervention | Mechanism of action | Clinical trial identifiera |
|---|---|---|---|
| Antiviral agents | Remdesivir | RNA-dependent RNA polymerase inhibitor | NCT04431453, NCT04409262, NCT04280705 |
| Chloroquine/hydroxychloroquine | Viral entry blockade by preventing host cell surface receptor glycosylation, reduction of cytokine secretion, endosomal acidification, autophagy inhibition | NCT04308668, NCT04303507, ChiCTR2000031174 | |
| Lopinavir/Ritonavir | Aspartic protease inhibitor | ChiCTR2000031196, NCT04350684 | |
| Ribavirin | RNA-dependent RNA polymerase inhibitor | NCT04254874, ChiCTR2000029308 | |
| Umifenovir | Prevents membrane fusion by disrupting interaction between viral spike protein and ACE2 receptor | NCT04260594, NCT04254874, ChiCTR2000029993 | |
| Darunavir/Cobicistat | Aspartic protease inhibitor | NCT04252274, ChiCTR2000029541 | |
| Favipiravir | RNA-dependent RNA polymerase inhibitor | NCT04359615, ChiCTR2000030254 | |
| Oseltamivir | Neuraminidase inhibitor | NCT04261270, ChiCTR2000029603 | |
| Enzyme inhibitors/receptor blockers | Camostat mesylate | Viral entry blockade via TMPRSS2 inhibition | NCT04353284, NCT04355052 |
| ACEi/ARBs | Elevation of ACE2 expression | NCT04351581, NCT04357535, NCT01597635, NCT04382950 |
RNA: ribonucleic acid; ACE2: angiotensin-converting enzyme 2; TMPRSS2: transmembrane protease, serine 2; ACEi: angiotensin-converting enzyme inhibitor; ARBs: angiotensin II receptor blockers.
aclinicaltrials.gov/chictr.org.cn.
Chloroquine/hydroxychloroquine
For decades, chloroquine (CQ) and hydroxychloroquine (HCQ) were used to treat diseases like rheumatoid arthritis, malaria, and systemic lupus erythematosus (SLE). 18 Although these relatively inexpensive agents are known to play a role in the reduction of proinflammatory cytokine secretion, endosomal acidification, preventing glycosylation and binding of angiotensin-converting enzyme 2 (ACE2) to the viral spike protein, and autophagy inhibition in host cells,19,20 they did not have much success against SARS-CoV and MERS-CoV. Interestingly, CQ was found to be effective against other viruses such as Zika, human immunodeficiency virus (HIV), dengue virus (DENV), West Nile virus (WNV) infection, influenza virus, and Japanese encephalitis virus (JEV), due to its ability to inhibit pH-dependent steps in the viral replication cycle.21–24
There is a paucity of data pertaining to the benefit of CQ for impacting SARS-CoV-2 infection. A few studies have observed growth inhibition of SARS-CoV-2 in response to CQ.13,25 A study conducted in China suggested improved viral clearance and slower disease progression when COVID-19 patients were treated with CQ. 26 HCQ showed better in vitro antiviral activity against SARS-CoV-2 than CQ at 24 h, as evidenced by lower EC50 values for HCQ. 25 Enhanced virologic clearance was reported in COVID-19 patients (70%) on the sixth day when they were administered a 200-mg dose of HCQ orally thrice a day compared to those in the control group (12.5%). Combinatorial therapy with azithromycin imparted additional benefit (100%) compared to that observed with monotherapy (57%). 27 These studies however had a small starting sample size (n = 36) that became even smaller due to a halt in the treatment of some patients (6) experiencing adverse events, lack of safety outcomes, disparities in basal viral loads among treatment arms, and toxicities associated with combination therapies, making it difficult to make significant conclusions of efficacy. On the contrary, a similar study showed no change in viral clearance when a double dose (400 mg) of HCQ was administered daily to patients in the treatment group, while the control group received standard of care. 28 A clinical trial (NCT04308668; Table 1) assessing the postexposure prophylactic potential of HCQ revealed no difference in the incidence of COVID-19 between the group of people exposed to a known case of COVID-19 and treated with HCQ, and those receiving placebo. 29 A large randomized controlled trial evaluating HCQ with and without azithromycin in hospitalized COVID-19 patients 30 found no improvement in an ordinal scale with HCQ or HCQ with azithromycin, while in the RECOVERY trial, HCQ was found to have no impact on mortality or rate of mechanical ventilation. 31
SLE and malaria patients can display adverse events such as retinopathy, QTc prolongation, neuropsychiatric effects, and hypoglycemia, upon administration of CQ/HCQ and require monitoring of these adverse events while on CQ/HCQ. 32 Patients taking QT-interval prolonging medications and severely ill patients are particularly prone to arrhythmia when treated with CQ/HCQ, generating potential concern for harm with CQ/HCQ treatment. 33
FDA issued an EUA for CQ/HCQ based on a few studies showing that the drugs improve viral clearance and decrease viral shedding. This EUA has since been revoked because the above findings could not be reproduced in randomized controlled trials. 34
Lopinavir/ritonavir
Originally developed to fight HIV, FDA-approved lopinavir/ritonavir combination is a potent inhibitor of aspartic proteases key to viral propagation, thereby resulting in the production of non-functional and immature virions. 35 The two drugs are complementary to each other in that ritonavir inhibits an enzyme called cytochrome P450 3A4 (CYP3A4), which is involved in the lopinavir metabolism in the liver. 36 Lopinavir/ritonavir is primarily excreted via feces with minimal urinary excretion. While lopinavir rarely crosses the placenta, ritonavir can increase the incidence of placental transfer. Studies have reported developmental defects in fetuses upon treatment of pregnant rats with high doses of these drugs. 37 Currently, there is a dearth of adequate data relating to the use of lopinavir/ritonavir in pregnant women, and therefore, the drugs should only be administered in light of significant benefit compared to risks.
A limited amount of in vitro data suggests that lopinavir possesses antiviral activity against SARS-CoV, MERS-CoV, human coronavirus hCoV-229E, 38 and SARS-CoV-2.39 Most clinical studies conducted at the time were either observational or retrospective and, therefore, inconclusive. Molecular docking analyses involving lopinavir/ritonavir revealed that these drugs can strongly bind to SARS-CoV-2 protease. 40 However, a previous study using lopinavir/ritonavir failed to demonstrate satisfactory outcomes for severe COVID-19 patients. Patients were administered 400 and 100 mg of lopinavir and ritonavir, respectively, two times a day over the course of 14 days. No difference was observed in the incidence of death, time to improvement, or the viral clearance in the group receiving lopinavir/ritonavir plus standard care compared to that receiving standard care alone. 41 Combination of lopinavir–ritonavir with interferon beta-1b has shown benefit in a small, randomized control trial for MERS. 42 The WHO Solidarity trial found no improvement in mortality for COVID-19 patients treated with lopinavir/ritonavir. 17 Drug delivery during an earlier stage of the viral life cycle may be necessary since studies involving delayed treatment showed no improvement in clinically significant outcomes.43,44 Adverse reactions have been reported by many studies41,45,46 and include gastrointestinal discomfort, skin eruptions, QT prolongation, liver injury as well as pancreatitis.
Darunavir and cobicistat
Like lopinavir/ritonavir, the darunavir/cobicistat combination belongs to the class of protease inhibitors and is known to have lesser adverse reactions compared to the former. 47 Being a structural analog of ritonavir, cobicistat has a similar mechanism of action to ritonavir in that it also inhibits CYP3A, CYP2D6, p-glycoprotein, and drug transporters such as organic anion transport protein (OATP1B1) and OATP1B3.47 While cobicistat has not shown an antiviral effect in vitro, darunavir worked well against SARS-CoV-2. However, clinically relevant data are currently unavailable.
A randomized Phase III clinical trial is currently ongoing to compare the efficacy of darunavir/cobicistat plus standard of care with the standard of care alone (NCT04252274; Table 1). Various outcomes including the rate of viral clearance on the seventh day and critical illness as well as mortality rate at two weeks are recorded. Another randomized controlled trial evaluating the safety and efficacy of darunavir/cobicistat with the standard of care consisting of thymosin α1 in adults is underway in Wuhan (ChiCTR2000029541; Table 1). Outcomes such as mortality, length of stay in the hospital/ICU, improvement of disease symptoms and lung CT findings, etc. will be evaluated. Some commonly observed adverse effects include nausea, diarrhea, headache, and muscle spasms. 47
Favipiravir
Similar to remdesivir, favipiravir (T-705) inhibits the activity of RdRP, a pivotal enzyme in the viral replication process, by acting as a guanine analog. 48 Favipiravir has shown activity against RNA viruses such as influenza, Ebola, Lassa, rabies,48,49 and more recently, SARS-CoV-2 at the preclinical level, although dosing regimens need to be adjusted depending upon the type of infection. 13 The drug has been licensed to treat influenza in several countries including Japan.
Lower concentrations of favipiravir have been reported to inhibit influenza, while higher doses are needed to treat COVID-19 and Ebola. 50 A loading dose of 2400–3000 mg twice a day and a subsequent dose of 1200–1800 mg twice a day have been suggested for administration. Pharmacokinetic values for the half-life period lie between 4.8 and 5.6 h. 49 Incidence and severity of adverse reactions (such as nausea, vomiting, uric acid elevation in serum) are much lower in favipiravir-treated patients than those treated with lopinavir/ritonavir. 51 Randomized controlled trials are underway to determine the potential of favipiravir to effectively treat COVID-19, in combination with other drugs such as HCQ (NCT04359615; Table 1). The efficacy of favipiravir, when administered with umifenovir, was determined in a clinical trial (ChiCTR2000030254; Table 1). No significant differences were found in the rate of clinical recovery in the two groups on day 7. While the latency was shortened for pyrexia and cough, there was no difference in the rate of oxygen therapy or mechanical ventilation. 52 These data support further investigation with RCTs of the efficacy of favipiravir for the treatment of COVID-19. Of late, Glenmark Pharmaceuticals has launched a Phase III clinical trial to test the efficacy of favipiravir in treating COVID-19 patients. The results of the trial are highly anticipated by August 2020.
Ribavirin
Ribavirin interferes with viral replication by inhibiting the RdRP enzyme. While commonly used for treating hepatitis C, ribavirin has shown a reduction in the mortality rate when administered in combination with lopinavir/ritonavir and a corticosteroid to SARS-CoV patients suffering from acute respiratory distress syndrome (ARDS) but only at high doses (e.g. 1200–2400 mg thrice a day orally). 53 Although a similar combination involving ribavirin, lopinavir/ritonavir, and an interferon-α did not prove effective against MERS-CoV,54,55 reduction in blood viral titers was observed. 56 No differences in antiviral activity were observed with various routes of drug administration. 57 Ribavirin has demonstrated the inhibition of viral replication in SARS-CoV-2 in vitro models established by the Bojkova group. 58 The study revealed the ability of the virus to reprogram key intracellular pathways and opened new avenues in terms of identifying small molecule inhibitors targeting these pathways, thereby inhibiting viral replication. 58 Multiple clinical trials to evaluate the efficacy of ribavirin and interferon-α in COVID-19 patients are underway (NCT04254874, ChiCTR2000029308; Table 1).
Ribavirin administration at high doses was associated with adverse reactions such as hemolytic anemia, transaminase elevations as well as liver toxicity 59 and safety issues such as the need for blood transfusions. 60 Ribavirin is not recommended for use in pregnant women as it is known to cause birth defects. 61
Umifenovir
Umifenovir or arbidol hydrochloride disrupts the interaction between viral spike protein and the ACE2 receptor, thereby preventing the process of membrane fusion. 62 Originally developed in Russia, umifenovir has shown efficacy in the prevention and cure of influenza. 63 Additionally, the antiviral agent has demonstrated activity against a range of viruses, including Zika, WNV, adenovirus, hepatitis B, hantavirus, and respiratory syncytial virus. 64 Preclinical data demonstrated inhibition of SARS-CoV reproduction in cultured cells in response to arbidol and ribavirin. 65
The recommended dosing regimen for influenza consists of oral administration of 200 mg arbidol three times a day. This regimen is currently being tested in combination with conventional treatment in a clinical trial involving ∼380 COVID-19 patients (NCT04260594; Table 1). Evidence also suggests that a nine-day arbidol treatment reduces mortality and increases discharge rates in COVID-19 patients in Wuhan, China compared to the control group. 66 Umifenovir is currently under investigation to determine its potential as a monotherapy or combination therapy candidate (NCT04254874, ChiCTR2000029993; Table 1).
Oseltamivir
Oseltamivir inhibits neuraminidase, a crucial enzyme facilitating the release of newly formed virions via the removal of sialic acid residues holding the viral particles on the host cell surface. 67 It has shown broad activity against various strains of influenza.68,69 If administered early following the onset of symptoms, oseltamivir can reduce the overall duration and severity of the disease. Oseltamivir should be converted to its carboxylate form to be active. 70
While oseltamivir has not shown antiviral activity in vitro, multiple clinical trials are evaluating its efficacy in combination with other agents such as favipiravir, ritonavir, CQ, and ASC09F (NCT04261270, ChiCTR2000029603; Table 1). Patient comorbidities and contraindications should be considered while deciding the optimal dosing regimen. There is an indication that oseltamivir may prove useful as a COVID-19 treatment due to its ability to bind to viral protease. 40 Oseltamivir-associated adverse reactions include nausea, vomiting, skin reactions, neuropsychiatric effects in children, 71 and sometimes even death. 72
Enzyme inhibitors/receptor blockers
Camostat mesylate
Camostat mesylate, a serine protease inhibitor commonly prescribed for pancreatitis in Japan and South Korea, has previously been shown to prevent the viral spread mediated by a serine protease in a mouse model of SARS-CoV infection and may potentially have a similar role in MERS-CoV pathogenesis. 73 More recently, it was demonstrated that the transmembrane protease, serine 2 (TMPRSS2) is involved in SARS-CoV-2 spike protein priming. 74 They also reported the blockade of the SARS-CoV-2 entry into the lung cells by camostat mesylate via TMPRSS2 inhibition. 74 Camostat mesylate (600 mg total, thrice a day) may reduce the severity of COVID-19 in patients. 75 The drug is safe for oral administration. Adverse effects associated with camostat mesylate include lightheadedness, pruritus, and increased appetite/thirst. 76 Multiple clinical trials are ongoing to determine the efficacy with which camostat mesylate reduces the viral burden in the upper respiratory tract in COVID-19 patients as a monotherapy (NCT04353284; Table 1).
ACE inhibitors, angiotensin II receptor blockers, and recombinant ACE2
SARS-CoV-2 infects the host cell upon binding of viral spike protein to ACE2.74 Binding and internalization of viral-ACE2 complex lead to increased ACE2 cleavage from cell membrane. Decreased ACE2 causes an increase in angiotensin II by ACE and a lower rate of conversion of angiotensin II (vasoconstrictor, proinflammatory, profibrotic) and a decrease in angiotensin1–7 (vasodilator, anti-inflammatory, antifibrotic) exacerbating lung injury. 77 In vivo studies in mice have shown ACE2 to be protective in acid-induced lung injury. 78 ACE inhibitors (ACEi) and angiotensin II receptor blockers (ARBs), medications to treat hypertension, have been shown to elevate ACE2 expression79,80 and could be protective in SARS-CoV-2 infection. However, ACE2 may act as a double-edged sword as the increased ACE2 expression could increase viral entry into the host cells.
A multitude of retrospective studies has looked at the impact of ACEi/ARBs on the risk of SARS-CoV-2 infection, COVID-19 disease severity, and COVID-19 mortality, the overwhelming majority finding no effect of ACEi/ARB use. Two recent randomized controlled trials looked at continued or switching ACEi/ARB use upon hospitalization with COVID-19 and found no impact on hospitalization outcomes.81,82 Adverse events associated with ACEi/ARBs include cough, angioedema, hypotension, hyperkalemia, and acute kidney injury. 83
Recombinant ACE2 (rACE2) therapy has previously shown promise in reducing angiotensin II and IL-6 levels in ARDS patients. 84 The absence of antibodies and adverse events was reported following the intravenous administration of the protein (NCT01597635; Table 1). The effect of similar therapy in COVID-19 patients remains to be seen though a case report suggested the potential benefit of the use of rACE2 with improvement in outcome and decrease in inflammatory cytokines and viral load in association rACE2.85 However, clinical trials are ongoing to evaluate the potential of rACE2 therapy (NCT04382950; Table 1) in COVID-19 outcomes.
Corticosteroids
Corticosteroids such as methylprednisolone, hydrocortisone, and dexamethasone are being used to treat COVID-19 due to their role as immunosuppressants. Historically, the role of corticosteroids has been debated. Observational studies conducted to determine their efficacy in treating viral infections indicate the association between deregulated viral clearance and complications in SARS-CoV and MERS-CoV, and higher secondary infection rates as well as mortality in influenza. 86 More recently, a COVID-19 study from Wuhan showed reduced mortality risk with methylprednisolone in patients with ARDS. Similarly, dexamethasone reduced mortality with a 6-mg dose administered for 10 days in COVID-19 patients on oxygen support, while no mortality benefit was observed in those who did not need it. 87 Hydrocortisone showed a similar mortality benefit. 88 Corticosteroid treatment is usually accompanied by adverse reactions such as psychosis and muscle weakness. In addition, steroids are prothrombotic, as evidenced by the autopsy of COVID-19 patients. 87 Their use at higher doses (>6 mg/day) and for longer than two weeks should be carefully weighed, as it could lead to increased mortality. 88
Conclusions
COVID-19 pandemic has led to several challenges across the world. It has challenged the scientific community to understand the pathogenesis of SARS-CoV-2 to inform potential therapeutic targets. It has challenged the medical community to characterize COVID-19 illness and mechanisms of disease to suggest hopeful therapeutic approaches. The ability of the scientific and medical communities to come together to build upon previous knowledge and apply it to the current pandemic has allowed for rapid evaluation of a number of developed medications. In some cases, this has brought promise (remdesivir), while in others, it allowed us to discount previously hopeful therapies (HCQ/CQ). Currently, remdesivir remains the only antiviral compound approved by FDA and can be combined with baricitinib to treat hospitalized COVID-19 patients, both adults and ≥ 2-year-old children. Despite the development of efficacious vaccines, repurposed therapeutics to improve outcomes in COVID-19 remain important, given ongoing challenges with the vaccine distribution.
Supplemental Material
Supplemental material, sj-pdf-1-ebm-10.1177_1535370221999989 for COVID-19: Antiviral agents and enzyme inhibitors/receptor blockers in development by Manasi P Jogalekar, Anurag Veerabathini and Ankit B Patel in Experimental Biology and Medicine
Footnotes
Authors’ contributions: MPJ, AV, and ABP contributed to the conception, writing, and discussion of this review manuscript. MPJ, AV, and ABP wrote the initial draft of the manuscript. The final version of the manuscript was approved by all authors.
Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: There was no funding to support this minireview.
ORCID iD: Manasi P Jogalekar https://orcid.org/0000-0003-1307-4829
Supplemental material: Supplemental material for this article is available online.
References
- 1.Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med 2020; 382:727–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Dong E, Du H, Gardner L. An interactive web-based dashboard to track COVID-19 in real time. Lancet Infect Dis 2020; 20:533–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jogalekar MP, Veerabathini A, Gangadaran P. Novel 2019 coronavirus: genome structure, clinical trials, and outstanding questions. Exp Biol Med (Maywood) 2020; 245:964–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, Wang W, Song H, Huang B, Zhu N. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 2020; 395:565–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic treatments for coronavirus disease 2019 (COVID-19): a review. JAMA 2020; 323:1824–36 [DOI] [PubMed] [Google Scholar]
- 6.Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, Hohmann E, Chu HY, Luetkemeyer A, Kline S. Remdesivir for the treatment of Covid-19. N Engl J Med 2020; 383:1813–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lo MK, Feldmann F, Gary JM, Jordan R, Bannister R, Cronin J, Patel NR, Klena JD, Nichol ST, Cihlar T. Remdesivir (GS-5734) protects African green monkeys from Nipah virus challenge. Sci Transl Med 2019; 11:eaau9242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Madelain V, Baize S, Jacquot F, Reynard S, Fizet A, Barron S, Solas C, Lacarelle B, Carbonnelle C, Mentré F. Ebola viral dynamics in nonhuman primates provides insights into virus immuno-pathogenesis and antiviral strategies. Nat Commun 2018; 9:4013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.de Wit E, Feldmann F, Cronin J, Jordan R, Okumura A, Thomas T, Scott D, Cihlar T, Feldmann H. Prophylactic and 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sheahan TP, Sims AC, Leist SR, Schäfer A, Won J, Brown AJ, Montgomery SA, Hogg A, Babusis D, Clarke MO. Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat Commun 2020; 11:222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Siegel D, Hui HC, Doerffler E, Clarke MO, Chun K, Zhang L, Neville S, Carra E, Lew W, Ross B. 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; 60:1648–61 [DOI] [PubMed] [Google Scholar]
- 12.Al-Tawfiq JA, Al-Homoud AH, Memish ZA. Remdesivir as a possible therapeutic option for the COVID-19. Travel Med Infect Dis 2020; 34:101615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, Shi Z, Hu Z, Zhong W, Xiao G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res 2020; 30:269–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, Marconi VC, Ruiz-Palacios GM, Hsieh L, Kline S. Baricitinib plus remdesivir for hospitalized adults with Covid-19. N Engl J Med 2021;384:795–807 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Goldman JD, Lye DC, Hui DS, Marks KM, Bruno R, Montejano R, Spinner CD, Galli M, Ahn M-Y, Nahass RG. Remdesivir for 5 or 10 days in patients with severe Covid-19. N Engl J Med 2020; 383:1827–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.World Health Organization. WHO R&D blueprint: ad-hoc expert consultation on clinical trials for Ebola therapeutics, https://www.who.int/ebola/drc-2018/treatments-approved-for-compassionate-use-update/en/ (2018, accessed 21 June 2020).
- 17.WHO Solidarity Trial Consortium. Repurposed antiviral drugs for COVID-19—interim WHO SOLIDARITY trial results. N Engl J Med 2020; 384:497–511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.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; 3:722–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Devaux CA, Rolain J-M, Colson P, Raoult D. New insights on the antiviral effects of chloroquine against coronavirus: what to expect for COVID-19? Int J Antimicrob Agents 2020; 55:105938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Zhou D, Dai S-M, Tong Q. COVID-19: a recommendation to examine the effect of hydroxychloroquine in preventing infection and progression. J Antimicrob Chemother 2020; 75:1667–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Delvecchio R, Higa LM, Pezzuto P, Valadão AL, Garcez PP, Monteiro FL, Loiola EC, Dias AA, Silva FJ, Aliota MT. Chloroquine, an endocytosis blocking agent, inhibits Zika virus infection in different cell models. Viruses 2016; 8:322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Farias KJS, Machado PRL, da Fonseca BAL. Chloroquine inhibits dengue virus type 2 replication in vero cells but not in C6/36 cells. ScientificWorldJournal 2013; 2013:282734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ooi EE, Chew JSW, Loh JP, Chua RC. In vitro inhibition of human influenza a virus replication by chloroquine. Virol J 2006; 3:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tsai W-P, Nara PL, Kung H-F, Oroszlan S. Inhibition of human immunodeficiency virus infectivity by chloroquine. AIDS Res Hum Retroviruses 1990; 6:481–9 [DOI] [PubMed] [Google Scholar]
- 25.Yao X, Ye F, Zhang M, Cui C, Huang B, Niu P, Liu X, Zhao L, Dong E, Song C. In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Clin Infect Dis 2020; 71:732–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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; 14:72–3 [DOI] [PubMed] [Google Scholar]
- 27.Gautret P, Lagier J-C, Parola P, Meddeb L, Mailhe M, Doudier B, Courjon J, Giordanengo V, Vieira VE, Dupont HT. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents 2020; 56:105949. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 28.Chen J, LD, LL, LP, Xu Q XL, Ling Y, Huang D Song S, Zhang D. A pilot study of hydroxychloroquine in treatment of patients with common coronavirus disease-19 (COVID-19). J Zhejiang Univ (Med Sci) 2020; 49:215–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Boulware DR, Pullen MF, Bangdiwala AS, Pastick KA, Lofgren SM, Okafor EC, Skipper CP, Nascene AA, Nicol MR, Abassi M. A randomized trial of hydroxychloroquine as postexposure prophylaxis for Covid-19. N Engl J Med 2020; 383:517–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cavalcanti AB, Zampieri FG, Rosa RG, Azevedo LC, Veiga VC, Avezum A, Damiani LP, Marcadenti A, Kawano-Dourado L, Lisboa T. Hydroxychloroquine with or without azithromycin in mild-to-moderate Covid-19. N Engl J Med 2020; 383:2041–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.RECOVERY Collaborative Group. Effect of hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med 2020; 383:2030–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kalil AC. Treating COVID-19—off-label drug use, compassionate use, and randomized clinical trials during pandemics. JAMA 2020; 323:1897–8 [DOI] [PubMed] [Google Scholar]
- 33.Lu ZK, Yuan J, Li M, Sutton SS, Rao GA, Jacob S, Bennett CL. Cardiac risks associated with antibiotics: azithromycin and levofloxacin. Expert Opin Drug Saf 2015; 14:295–303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Thomson K, Nachlis H. Emergency use authorizations during the COVID-19 pandemic: lessons from hydroxychloroquine for vaccine authorization and approval. JAMA 2020; 324:1282–3 [DOI] [PubMed] [Google Scholar]
- 35.Chan JF-W, Yao Y, Yeung M-L, Deng W, Bao L, Jia L, Li F, Xiao C, Gao H, Yu P. Treatment with lopinavir/ritonavir or interferon-β1b improves outcome of MERS-CoV infection in a nonhuman primate model of common marmoset. J Infect Dis 2015; 212:1904–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Oldfield V, Plosker GL. Lopinavir/ritonavir: a review of its use in the management of HIV infection. Drugs 2006; 66:1275–99 [DOI] [PubMed] [Google Scholar]
- 37.Cunha A, Hagemann C, Simoes R, Oliveira-Filho R, Simões M, Soares J Jr, Maciel G, Oliveira F, Amed A, Kulay L Jr. Effects of lopinavir–ritonavir combined therapy during the rat pregnancy: morphological and biochemical aspects. Eur J Obstet Gynecol Reprod Biol 2007; 133:60–3 [DOI] [PubMed] [Google Scholar]
- 38.De Wilde AH, Jochmans D, Posthuma CC, Zevenhoven-Dobbe JC, Van Nieuwkoop S, Bestebroer TM, Van Den Hoogen BG, Neyts J, Snijder EJ. 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; 58:4875–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Choy K-T, Wong AY-L, Kaewpreedee P, Sia S-F, Chen D, Hui KPY, Chu DKW, Chan MCW, Cheung PP-H, Huang X. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antiviral Res 2020; 178:104786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Muralidharan N, Sakthivel R, Velmurugan D, Gromiha MM. Computational studies of drug repurposing and synergism of lopinavir, oseltamivir and ritonavir binding with SARS-CoV-2 protease against COVID-19. J Biomol Str Dyn 2020;1–6 [DOI] [PubMed] [Google Scholar]
- 41.Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G, Ruan L, Song B, Cai Y, Wei M. A trial of lopinavir–ritonavir in adults hospitalized with severe Covid-19. N Engl J Med 2020; 382:1787–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Arabi YM, Asiri AY, Assiri AM, Balkhy HH, Al Bshabshe A, Al Jeraisy M, Mandourah Y, Azzam MH, Bin Eshaq AM, Al Johani S. Interferon beta-1b and lopinavir–ritonavir for Middle East respiratory syndrome. N Engl J Med 2020; 383:1645–56 [DOI] [PubMed] [Google Scholar]
- 43.Que T, Wong V, Yuen K. Treatment of severe acute respiratory syndrome with lopinavir/ritonavir: a multicentre retrospective matched cohort study. Hong Kong Med J 2003; 9:399–406 [PubMed] [Google Scholar]
- 44.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. J Med Virol 2020; 92:556–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Cooper C, Van Heeswijk R, Gallicano K, Cameron D. A review of low-dose ritonavir in protease inhibitor combination therapy. Clin Infect Dis 2003; 36:1585–92 [DOI] [PubMed] [Google Scholar]
- 46.Wu C, Chen X, Cai Y, Zhou X, Xu S, Huang H, Zhang L, Zhou X, Du C, Zhang Y. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med 2020; 180:934–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kakuda TN, Van De Casteele T, Petrovic R, Neujens M, Salih H, Opsomer M, Hoetelmans RM. Bioequivalence of a darunavir/cobicistat fixed-dose combination tablet versus single agents and food effect in healthy volunteers. Antivir Ther 2014; 19:597–606 [DOI] [PubMed] [Google Scholar]
- 48.Furuta Y, Komeno T, Nakamura T. Favipiravir (T-705), a broad spectrum inhibitor of viral RNA polymerase. Proc Jpn Acad Ser B Phys Biol Sci 2017; 93:449–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Shiraki K, Daikoku T. Favipiravir, an anti-influenza drug against life-threatening RNA virus infections. Pharmacol Ther 2020; 209:107512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Agrawal U, Raju R, Udwadia ZF. Favipiravir: a new and emerging antiviral option in COVID-19. Med J Armed Forces India 2020; 76:370–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Yanai H. Favipiravir: a possible pharmaceutical treatment for COVID-19. J Endocrinol Metab 2020; 10:33–4 [Google Scholar]
- 52.Shrestha DB, Budhathoki P, Khadka S, Shah PB, Pokharel N, Rashmi P. Favipiravir versus other antiviral or standard of care for COVID-19 treatment: a rapid systematic review and meta-analysis. Virol J 2020; 17:141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chu C, Cheng V, Hung I, Wong M, Chan K, Chan K, Kao R, Poon L, Wong C, Guan Y. Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings. Thorax 2004; 59:252–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Al-Tawfiq JA, Momattin H, Dib J, Memish ZA. Ribavirin and interferon therapy in patients infected with the Middle East respiratory syndrome coronavirus: an observational study. Int J Infect Dis 2014; 20:42–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Omrani AS, Saad MM, Baig K, Bahloul A, Abdul-Matin M, Alaidaroos AY, Almakhlafi GA, Albarrak MM, Memish ZA, Albarrak AM. Ribavirin and interferon alfa-2a for severe Middle East respiratory syndrome coronavirus infection: a retrospective cohort study. Lancet Infect Dis 2014; 14:1090–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Spanakis N, Tsiodras S, Haagmans BL, Raj VS, Pontikis K, Koutsoukou A, Koulouris NG, Osterhaus AD, Koopmans MP, Tsakris A. Virological and serological analysis of a recent Middle East respiratory syndrome coronavirus infection case on a triple combination antiviral regimen. Int J Antimicrob Agents 2014; 44:528–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Foolad F, Aitken SL, Shigle TL, Prayag A, Ghantoji S, Ariza-Heredia E, Chemaly RF. Oral versus aerosolized ribavirin for the treatment of respiratory syncytial virus infections in hematopoietic cell transplant recipients. Clin Infect Dis 2019; 68:1641–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bojkova D, Klann K, Koch B, Widera M, Krause D, Ciesek S, Cinatl J, Münch C. Proteomics of SARS-CoV-2-infected host cells reveals therapy targets. Nature 2020; 583:469–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Stockman LJ, Bellamy R, Garner P. SARS: systematic review of treatment effects. PLoS Med 2006; 3:e343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Arabi YM, Shalhoub S, Mandourah Y, Al-Hameed F, Al-Omari A, Al Qasim E, Jose J, Alraddadi B, Almotairi A, Al Khatib K. Ribavirin and interferon therapy for critically ill patients with Middle east respiratory syndrome: a multicenter observational study. Clin Infect Dis 2020; 70:1837–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Altinbas S, Holmes JA, Altinbas A. Hepatitis C virus infection in pregnancy: an update. Gastroenterol Nurs 2020; 43:12–21 [DOI] [PubMed] [Google Scholar]
- 62.Kadam RU, Wilson IA. Structural basis of influenza virus fusion inhibition by the antiviral drug arbidol. Proc Natl Acad Sci U S A 2017; 114:206–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Boriskin Y, Leneva I, Pecheur E-I, Polyak S. Arbidol: a broad-spectrum antiviral compound that blocks viral fusion. Curr Med Chem 2008; 15:997–1005 [DOI] [PubMed] [Google Scholar]
- 64.Haviernik J, Štefánik M, Fojtíková M, Kali S, Tordo N, Rudolf I, Hubálek Z, Eyer L, Ruzek D. Arbidol (umifenovir): a broad-spectrum antiviral drug that inhibits medically important arthropod-borne flaviviruses. Viruses 2018; 10:184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Khamitov R, Loginova S, Shchukina V, Borisevich S, Maksimov V, Shuster A. Antiviral activity of arbidol and its derivatives against the pathogen of severe acute respiratory syndrome in the cell cultures. Vopr Virusol 2008; 53:9–13 [PubMed] [Google Scholar]
- 66.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 2020; 71:769–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Moscona A. Neuraminidase inhibitors for influenza. N Engl J Med 2005; 353:1363–73 [DOI] [PubMed] [Google Scholar]
- 68.Leneva IA, Roberts N, Govorkova EA, Goloubeva OG, Webster RG. The neuraminidase inhibitor GS4104 (oseltamivir phosphate) is efficacious against a/Hong Kong/156/97 (H5N1) and a/Hong Kong/1074/99 (H9N2) influenza viruses. Antiviral Res 2000; 48:101–15 [DOI] [PubMed] [Google Scholar]
- 69.Tumpey TM, García-Sastre A, Mikulasova A, Taubenberger JK, Swayne DE, Palese P, Basler CF. Existing antivirals are effective against influenza viruses with genes from the 1918 pandemic virus. Proc Natl Acad Sci U S A 2002; 99:13849–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Davies BE. Pharmacokinetics of oseltamivir: an oral antiviral for the treatment and prophylaxis of influenza in diverse populations. J Antimicrob Chemother 2010; 65:ii5–ii10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Toovey S, Prinssen EP, Rayner CR, Thakrar BT, Dutkowski R, Koerner A, Chu T, Sirzen-Zelenskaya A, Britschgi M, Bansod S. Post-marketing assessment of neuropsychiatric adverse events in influenza patients treated with oseltamivir: an updated review. Adv Ther 2012; 29:826–48 [DOI] [PubMed] [Google Scholar]
- 72.Hoffman KB, Demakas A, Erdman CB, Dimbil M, Doraiswamy PM. Neuropsychiatric adverse effects of oseltamivir in the FDA adverse event reporting system, 1999-2012. BMJ 2013; 347:f4656. [DOI] [PubMed] [Google Scholar]
- 73.Zhou Y, Vedantham P, Lu K, Agudelo J, Carrion R, Jr, Nunneley JW, Barnard D, Pöhlmann S, McKerrow JH, Renslo AR. Protease inhibitors targeting coronavirus and filovirus entry. Antiviral Res 2015; 116:76–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu N-H, Nitsche A. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020; 181:271–80.e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Uno Y. Camostat mesilate therapy for COVID-19. Intern Emerg Med 2020; 15:1577–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ramsey ML, Nuttall J, Hart PA. A phase 1/2 trial to evaluate the pharmacokinetics, safety, and efficacy of NI-03 in patients with chronic pancreatitis: study protocol for a randomized controlled trial on the assessment of camostat treatment in chronic pancreatitis (TACTIC). Trials 2019; 20:501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gurwitz D. Angiotensin receptor blockers as tentative SARS-CoV-2 therapeutics. Drug Dev Res 2020; 81:537–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, Leong-Poi H. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 2005; 436:112–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Furuhashi M, Moniwa N, Mita T, Fuseya T, Ishimura S, Ohno K, Shibata S, Tanaka M, Watanabe Y, Akasaka H. Urinary angiotensin-converting enzyme 2 in hypertensive patients may be increased by olmesartan, an angiotensin II receptor blocker. Am J Hypertens 2015; 28:15–21 [DOI] [PubMed] [Google Scholar]
- 80.Klimas J, Olvedy M, Ochodnicka ‐Mackovicova K, Kruzliak P, Cacanyiova S, Kristek F, Krenek P, Ochodnicky P. Perinatally administered losartan augments renal ACE 2 expression but not cardiac or renal mas receptor in spontaneously hypertensive rats. J Cell Mol Med 2015; 19:1965–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Cohen JB, Hanff TC, William P, Sweitzer N, Rosado-Santander NR, Medina C, Rodriguez-Mori JE, Renna N, Chang TI, Corrales-Medina V. Continuation versus discontinuation of renin–angiotensin system inhibitors in patients admitted to hospital with COVID-19: a prospective, randomised, open-label trial. Lancet Respir Med 2021;9:275–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lopes RD, Macedo AV, Silva P, Moll-Bernardes RJ, dos Santos TM, Mazza L, Feldman A, Arruda GDAS, DC, Camiletti AS. Effect of discontinuing vs continuing angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers on days alive and out of the hospital in patients admitted with COVID-19: a randomized clinical trial. JAMA 2021; 325:254–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Turner JM, Kodali R. Should angiotensin-converting enzyme inhibitors ever be used for the management of hypertension? Curr Cardiol Rep 2020; 22:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Khan A, Benthin C, Zeno B, Albertson TE, Boyd J, Christie JD, Hall R, Poirier G, Ronco JJ, Tidswell M. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit Care 2017; 21:234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zoufaly A, Poglitsch M, Aberle JH, Hoepler W, Seitz T, Traugott M, Grieb A, Pawelka E, Laferl H, Wenisch C. Human recombinant soluble ACE2 in severe COVID-19. Lancet Respir Med 2020; 8:1154–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Russell CD, Millar JE, Baillie JK. Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury. Lancet 2020; 395:473–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mishra GP, Mulani J. Corticosteroids for COVID-19: the search for an optimum duration of therapy. Lancet Respir Med 2021; 9:e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Prescott HC, Rice TW. Corticosteroids in COVID-19 ARDS: evidence and hope during the pandemic. JAMA 2020; 324:1292–5 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental material, sj-pdf-1-ebm-10.1177_1535370221999989 for COVID-19: Antiviral agents and enzyme inhibitors/receptor blockers in development by Manasi P Jogalekar, Anurag Veerabathini and Ankit B Patel in Experimental Biology and Medicine

