Next Article in Journal
Thiophene-Based Compounds with Potential Anti-Inflammatory Activity
Next Article in Special Issue
Prevalence and Significance of Hypermetabolic Lymph Nodes Detected by 2-[18F]FDG PET/CT after COVID-19 Vaccination: A Systematic Review and a Meta-Analysis
Previous Article in Journal
Dietary Effects of Anthocyanins in Human Health: A Comprehensive Review
Previous Article in Special Issue
Current Status of Baricitinib as a Repurposed Therapy for COVID-19
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Update on Functional Inhibitors of Acid Sphingomyelinase (FIASMAs) in SARS-CoV-2 Infection

by
Gwenolé Loas
1,2,* and
Pascal Le Corre
3,4,5
1
Department of Psychiatry, Hôpital Erasme, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
2
Research Unit (ULB 266), Hôpital Erasme, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
3
Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU de Rennes, 35033 Rennes, France
4
Irset (Institut de Recherche en Santé, Environnement et Travail)-Inserm UMR 1085, University of Rennes, CHU Rennes, INSERM, EHESP, 35000 Rennes, France
5
Laboratoire de Biopharmacie et Pharmacie Clinique, Faculté de Pharmacie, Université de Rennes 1, 35043 Rennes, France
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2021, 14(7), 691; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14070691
Submission received: 30 June 2021 / Revised: 15 July 2021 / Accepted: 16 July 2021 / Published: 18 July 2021
(This article belongs to the Special Issue COVID-19 in Pharmaceuticals)

Abstract

:
The SARS-CoV-2 outbreak is characterized by the need of the search for curative drugs for treatment. In this paper, we present an update of knowledge about the interest of the functional inhibitors of acid sphingomyelinase (FIASMAs) in SARS-CoV-2 infection. Forty-nine FIASMAs have been suggested in the treatment of SARS-CoV-2 infection using in silico, in vitro or in vivo studies. Further studies using large-sized, randomized and double-blinded controlled clinical trials are needed to evaluate FIASMAs in SARS-CoV-2 infection as off-label therapy.

1. Introduction

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is identified as the disease-causing pathogen of Coronavirus disease 2019 (COVID-19). Up to June 2021, the number of confirmed cases worldwide exceeded 179 million, with more than 3.9 million deaths. Some encouraging results have been observed using dexamethasone and remdesivir in the treatment of COVID-19 in patients requiring supplemental oxygen, mechanical ventilation or extracorporeal membrane oxygenation, but there is still no drug preventing host cell infection or cytokine release syndrome.
SARS-CoV-2 is a family of enveloped viruses that enter into host cells by receptor binding and membrane fusion [1], and acid sphingomyelinase (ASM) and ceramide play a prominent role in receptor signaling and infection cycle [1]. There are two forms of ASM: (i) lysosomal ASM that is transported to the endosomal compartment and anchored to the inner lysosomal membrane, and (ii) secretory ASM that is transported to the outer leaflet of the plasma membrane.
Some organic molecules, including currently marketed drugs, have the potential to functionally inhibit the activity of ASM from reversible and additive manner. These molecules identified by the acronym FIASMAs (i.e., functional inhibitors of acid sphingomyelinase) have the potential to inhibit this enzyme, notably the lysosomal ASM, and therefore disrupt the entry of viruses into cells [2]. In cell culture models, inhibition of acid sphingomyelinase activity by amitriptyline has been shown to prevent the infection of cells with SARS-CoV-2 and pseudoviral SARS-CoV-2 in an ex vivo model [3]. Hence, the impact of chronic exposure of patients to a drug or a group of drugs with FIASMA properties on the clinical course of patients infected with SARS-CoV-2 may be questioned.
At least sixty-four drugs classified as FIASMAs reduce ASM activity by at least 50% at 10 µM concentration. The distribution of FIASMAs with respect to their ATC code revealed that specific therapeutic groups were over-represented: C08 (calcium channel blockers; amlodipine), D04 (antipruritics; promethazin), N04 (anti-Parkinson’s; benztropine), R06 (antihistamines for systemic use; astemizole), N06 (psychoanaleptics; fluvoxamine) and N05 (psycholeptics; chlorpromazine).
Repurposing of drugs in COVID-19 is being investigated using in silico, in vitro or ex vivo activity-based studies, as well on in vivo activity-based drug repurposing in animal models and human studies (epidemiological, clinical studies, case reports).
Recently (9 January 2021), we reviewed [4] all the corresponding studies (N = 27), including the pre-print publications, and found that thirty-two FIASMAs could be considered as potential drugs for treating SARS-CoV-2.
The aim of the present overview is firstly to present a brief history of the interest of FIASMAs in SARS-CoV-2 infection, and secondly, to provide an updated overview of studies on FIASMAs in this infection.

2. Brief History of the Studies of FIASMAs in the SARS-CoV-2 Infection

In 2005, a study reported the role of increased ASM activity in organ failure of patients with severe sepsis [5].
In 2010, compounds including marketed drugs with a potential to inhibit ASM activity in vitro were called “Functional inhibitors of ASM” (FIASMAs). These compounds are cationic amphiphilic molecules with a relative heterogeneity in terms of chemical structure. They typically are polycyclic molecules, with at least 1 basic nitrogen atom (pKa > 4, which corresponds to a partially protonated functional group at acidic pH) and show moderate to high lipophilicity (logp > 3) [2].
In 2014, two different in vitro studies reported that several FIASMAs were active against both MERS-CoV and SARS-CoV [6,7]. In these two studies, inhibition of ASM was not mentioned, and other active mechanisms were suggested as inhibitors of clathrin-mediated endocytosis for chlorpromazine.
In 2019, Andrews [8] suggested that “It is becoming increasingly clear that many pathogens that produce membrane damaging also trigger a repair mechanism involving exocytosis of lysosomal ASM, generating ceramide-enriched cell surface domains that facilitate cell invasion”.
Independently of the ASM pathway, viruses usually take advantage of the endocytosis mechanisms to penetrate the cytosol of cells, and different mechanisms of internalization are involved. These are clathrin-mediated endocytosis, macropinocytosis, caveolar/lipid raft-mediated endocytosis, as well as several less well-characterized clathrin- and caveolin/lipid raft-independent mechanisms [9].
Markus Blaess in a pre-print (May 5, 2020) [10] strongly suggested that lysosomotropic compounds could protect against COVID-19 infection in a concealed way, and cited a set of 34 clinically approved lysosomotropic compounds, 30 of them being FIASMAs. The author suggested that these compounds could be used off label using either local (inhalative) or systematic administration. Later, the author [11] developed the repurposing of lysosomotropic drugs (including numerous FIASMAs) in COVID-19 infection.
In 2020, among the 23 in silico, in vitro or ex vivo studies reporting activity of FIASMAs against SARS-CoV-2 [4], only two cited the inhibition of ASM as the principal mechanism of action of the drugs [3,12].
In 2021, Chung and Claus [13] reviewed the function of ASM to explore the question whether ASM is a friend or foe in the course of sepsis and severe infection (not limited to COVID-19 infection). The interest of FIASMAs repurposing and particularly amitriptyline were underlined by the authors.
In 2021, a study [14] used SARS-CoV-2 pseudoviruses to infect human angiotensin-converting enzyme 2 (ACE2)-expressing HEK293T cells and evaluated virus infection. SARS-CoV-2 entry was dependent on ACE2 and sensitive to pH endosome/lysosome in HEK293T cells. Moreover, the infection of SARS-CoV-2 pseudoviruses was independent of dynamin, clathrin, caveolin, endophilin as well as micropinocytosis. Cholesterol-rich lipids rafts and endosomal acidification are key steps of SARS-CoV-2 required for infection of host cells.

3. Update (16 June 2021) on Studies with FIASMAs in the SARS-CoV-2 Infection

To review the extensive evidence about FIASMAs as a therapeutic modality for COVID-19, authors attempted to answer the following key questions. First, would FIASMAs provide benefits in relation to COVID-19? Second, should FIASMAs be used as early intervention in COVID-19 disease?
We manually searched two electronic databases, PubMed and Google Scholar, for English-language titles and abstracts using the terms “Alverine OR… OR… Trimipramine” (64 drugs, see [2]) and “COVID-19 OR SARS-CoV-2”
Among the 231 articles (PubMed and Google Scholar), 91 were retained [3,12,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103]. Twenty-seven have been reviewed in our previous article [4] (23 in silico, in vitro or in vivo studies, 5 human studies (one common with in vitro studies)) and 64 were new studies highlighting the interest of the scientific community for FIASMAs. Among these new studies, there were 52 in silico, in vitro or in vivo studies, and 14 were human studies (2 of them also reporting in silico or in vitro studies)
Forty-nine FIASMAs have been suggested as potential treatment in SARS-CoV-2 infection and nine of them (benztropine, chlorpromazine, clomipramine, emetine, fluphenazine, loperamide, promethazine, tamoxifene and triflupromazine) were active on the three coronaviruses (SARS, SARS-MERS, SARS-CoV-2) (see Table 1).
Fifteen FIASMAs [20,25,29,30,31,32,46,53,54,73,75,76,77,78,82,88,101,102,103] have been studied using epidemiological-clinical studies or case reports (amiodarone, amitriptyline, amlodipine, carvedilol, chlorpromazine, clomipramine, desloratadine, fluoxetine, fluvoxamine, hydroxyzine, loperamide, loratadine, melatonine, paroxetine, and sertraline). Among the 15 FIASMAs, only amlodipine [32] and fluvoxamine [75] were studied using randomized double-blind clinical studies (see Table 2).
Among the 19 human studies, there were two case reports on amiodarone [20] or loratadine [88], four retrospective studies that have explored the association between one FIASMAs (carvedilol [25,53], hydroxyzine [77,101], loratadine [77], melatonine [25,53], paroxetine [53]) and the negativity or positivity on a PCR test, four prospective studies on fluvoxamine [75,76], melatonine [102] or amlodipine [32] including two randomized clinical trial [32,75], 4 retrospective studies on mortality in hospitalized COVID-19 patients on amlodipine [29,30,31] or melatonine [82], one observational study on low rate of COVID-19 infection in psychiatric patients treated by antipsychotics comparatively to nurses or physicians [73], 4 retrospective studies on mortality or intubation on hospitalized COVID-19 patients on amiodarone, amitriptyline, amlodipine, chlorpromazine, clomipramine, desloratadine, fluoxetine, hydroxyzine, paroxetine and sertraline [46,54,78,103].
Several methodological aspects of the abovementioned studies deserve to be discussed. First, the choice of the primary end point can be questioned in some studies, especially those based on clinical deterioration (e.g., percentage of admission in ICU, time to intubation, or percentage of intubation …) or those using a composite criterion as opposed to those based only on mortality rate. Indeed, the former studies can display significant variability as a function of time (bed occupancy rate, number of other cases requiring intubation, pandemia severity …) and also according to hospitals, thus potentially leading to problems of reproducibility in the criteria in multicenter retrospective studies [46,54,78,103]. Hence, studies based on a mortality criterion should be considered more relevant. The second point to be considered is the choice of the FIASMAs and the duration of their prescription within the study. Indeed, this latter parameter influences the accumulation in pulmonary tissue, and more specifically in lysosomes, to achieve the desired inhibition of acid sphingomyelinase [11]. The highest accumulation in plasma and also into the lysosome compartments (i.e., called steady-state) requires around 7 times the apparent elimination half-life of the drug to occur. This can lead to a quite long delay depending on the drug (e.g., from 7 to 14 days for fluoxetine), especially for an infection where deterioration and mortality arise quite rapidly in patients with severe symptoms. Besides the time to reach the highest accumulation depending on the elimination half-life, the magnitude of the tissue distribution has to be considered. As lipophilic and amine drugs, most FIASMAs have a very high volume of distribution [4], so that they have the potential to distribute freely in some body organs. More precisely, some of these drugs have been called “pneumophilic drugs” as a result of their ability to distribute in the lungs (e.g., amiodarone, verapamil, tricyclic antidepressants like imipramine and amitriptyline, and phenothiazines like chlorpromazine, β-blockers like propranolol, and local anesthetics like lidocaine). The mechanism of this lung accumulation currently remains unclear but carrier-mediated pathways have been suggested. Even though their volume of distribution may be quite large, these drugs are also very highly bound to plasma proteins (>95%) so that drug-free levels in the body should be quite low. Hence, in order to estimate the potential effect of a drug on the course of COVID-19 infection in patients from retrospective studies, it would be necessary to make sure that the patients included in the study have previously been exposed to the drug(s) being studied at the time of inclusion, ideally by a chronic prescription. Given these pharmacokinetic features, in our previous paper [4] we have underlined that reaching a steady state to obtain a maximal effect would require a delay (seven times the half-life), and alternatively that a loading dose may be used to rapidly reach the steady state [11].
The direct administration through the pulmonary tract may help in reaching pulmonary targets, and should be an option to consider since COVID is a respiratory disease (11). Ambroxol, a lipophilic cationic molecule, not formally categorized as FIASMA, is approved as a mucolytic drug, and can be used by inhalation. In a recent ex vivo study, it has been shown that ambroxol reduced ASM activity in human nasal epithelial cells infected with pp-SARS-CoV-2 spike [100].
When examining the results of retrospective studies in COVID-19 hospitalized patients, it is important to keep in mind that the mean length of hospital stay is 13 days with around an 8-day delay from symptom onset to admission, and 3-day delay from admission to ICU (ISARIC clinical data report 4 October 2020 on 102,959 individuals from 566 sites across 42 countries).
In other terms, if the study baseline was defined as the date of hospital admission, and notably within the first 48 h, then the steady state is unlikely to be obtained for most drugs within the natural course of the disease except for the FIASMAs having a short half-life (melatonine: 7 half-lives: 6–7 h, alverine: 7 half-lives: 6 h, cloperastine: 7 half-lives: 24 h).
Among the different retrospective studies on COVID-19 hospitalized patients, two studies have included patients without mentioning the duration of the prescription of the drugs [29,30], one study explored the interest of melatonine [82], only one study has included patients with FIASMAs taken for at least 7 half-lives [31], and four studies have taken into account only FIASMAs prescribed after the hospitalization. More precisely, for these four studies, the beginning of the prescription was: the date of the first prescription (during hospitalization) of chlorpromazine [46] or hydroxyzine [78],—within the first 48 h of hospital admission for receiving any antidepressants [54],—within the first 24 h of hospital admission for receiving at least one FIASMAs [103]. However, among the clinical studies, negative results were reported for amlodipine, amiodarone, amitriptyline, clomipramine, chlorpromazine, desloratadine, fluoxetine, hydroxyzine, paroxetine, sertraline [32,46,54,103] (see Table 2). These negative results could be explained first by the acute drug prescription, second by underpowered studies (an insufficient power of the statistical tests) explained by low samples size, and third by selection biases.
Regarding the two case reports, those [20] reporting the use of amiodarone during 5 days, although the half-life of the drug is around 50 days, calls into question the FIASMA activity of the drug.
Regarding the prospective studies, and notably the randomized clinical studies using either fluvoxamine or amlodipine, the protocol required the drug intake after the randomization to patients having a positive RT-PCR.
For the randomized double-blind clinical study exploring the potential effect of fluvoxamine against placebo [75], the main criterion was clinical deterioration within 15 days so that the half-life of the drug (17–22 h, steady-state: 5–6 days) allowed to test for ASM activity.
For the randomized double-blind clinical study [32] exploring the potential effect of amlodipine against losartan, the authors had two principal criteria: 30-day mortality and length of hospital stay. They reported no significant difference on the two criteria, but in their study, a stratified randomization of age has not been done, and the mean age of the groups were different: 67.3 years and 60.1 years for losartan and amlodipine, respectively. The half-lives of amlodipine (30 to 60 h) and losartan and its carboxylic active metabolite (about 2 h and 6–9 h, respectively) were also different. The difference in age and in half-life of the drugs studied appeared to be serious limitations of the study for a fair comparison between the drugs.
Unfortunately, there is, as yet, no published study using randomized double-blind clinical study exploring one FIASMA against placebo with mortality as principal criterion.

4. Conclusions

The present review highlights the particular interest of the repurposing of FIASMAs for the inhibition of SARS-CoV-2 entry, and potential limitations of some published retrospective or prospective studies.
Future studies should pay better attention to the pharmacokinetic properties of the drugs investigated to allow the control of potential confounding factors found in retrospective studies; and to optimize the design of prospective controlled clinical trials (e.g., relevance of a loading dose for drugs with prolonged elimination half-lives). The present review highlights the particular interest of the repurposing of FIASMAs for the inhibition of SARS-CoV-2 entry. Very recent review highlights the interest of inhibition of ASM in SARS-CoV-2 infection, and suggests that serum level of shingosine-1-phosphate (S1P), one of the shingolipids, could be a prognostic factor for COVID-19 severity [104].
However, a recent research article has highlighted that phospholipidosis was a shared mechanism underlying the SARS-CoV-2 antiviral activity of many repurposed drugs without mentioning ASM inhibition [105].
Besides the pharmacokinetic properties, the choice of a FIASMA should consider the target population. Studies implemented in subjects just after a positive RT-PCR test should consider FIASMA with a low rate of undesirable adverse effects. Thus, antipsychotics (e.g., chlorpromazine) should not be considered and could be contraindicated. Studies implemented in subjects with a recent infection requiring hospitalization should ideally consider FIASMA combining other mechanisms of action to reduce excessive inflammatory response during sepsis or to attenuate neurological complications. For example, amitriptyline exerts neuroprotection via tropomyosin receptor kinase [106], and fluvoxamine via the Sigma-1 receptor activity may modulate SARS-CoV-2 induced cytokine storm [107].

Author Contributions

G.L. and P.L.C. conceived the idea and write the manuscript. Both authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zheng, Y.; Shang, J.; Yang, Y.; Liu, C.; Wan, Y.; Geng, Q.; Wang, M.; Baric, R.; Li, F. Lysosomal proteases are a determinant of coronavirus tropism. J. Virol. 2018, 92, e01504–e01518. [Google Scholar] [CrossRef] [Green Version]
  2. Kornhuber, J.; Tripal, P.; Reichel, M.; Mühle, C.; Rhein, C.; Muehlbacher, M.; Groemer, T.W.; Gulbins, E. Functional Inhibitors of Acid Sphingomyelinase (FIASMAs): A novel pharmacological group of drugs with broad clinical applications. Cell. Physiol. Biochem. 2010, 26, 9–20. [Google Scholar] [CrossRef]
  3. Carpinteiro, A.; Edwards, M.J.; Hoffmann, M.; Kochs, G.; Gripp, B.; Weigang, S.; Adams, C.; Carpinteiro, E.; Gulbins, A.; Keitsch, S.; et al. Pharmacological inhibition of acid sphingomyelinase prevents uptake of SARS-CoV-2 by epithelial cells. Cell. Rep. Med. 2020, 1, 100142. [Google Scholar] [CrossRef] [PubMed]
  4. Le Corre, P.; Loas, G. Repurposing functional inhibitors of acid sphingomyelinase (fiasmas): An opportunity against SARS-CoV-2 infection? J. Clin. Pharm. Ther. 2021, 1. [Google Scholar] [CrossRef]
  5. Claus, R.A.; Bunck, A.C.; Bockmeyer, C.L.; Brunkhorst, F.M.; Lösche, W.; Kinscherf, R.; Deigner, H.P. Role of increased sphingomyelinase activity in apoptosis and organ failure of patients with severe sepsis. FASEB J. 2005, 19, 1719–1721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Dyall, J.; Coleman, C.M.; Hart, B.J.; Venkataraman, T.; Holbrook, M.R.; Kindrachuk, J.; Johnson, R.F.; Olinger, G.G., Jr.; Jahrling, P.B.; Laidlaw, M.; et al. Repurposing of clinically developed drugs for treatment of Middle East Respiratory Syndrome coronavirus infection. Antimicrob. Agents Chemother. 2014, 58, 4885–4893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. de Wilde, A.H.; Jochmans, D.; Posthuma, C.C.; Zevenhoven-Dobbe, J.C.; van Nieuwkoop, S.; Bestebroer, T.M.; van den Hoogen, B.G.; Neyts, J.; Snijder, E.J. 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, 4875–4884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Andrews, N.W. Solving the secretory acid sphingomyelinase puzzle: Insights from lysosome-mediated parasite invasion and plasma membrane repair. Cell. Microbiol. 2019, 21, e13065. [Google Scholar] [CrossRef]
  9. Mercer, J.; Schelhaas, M.; Helenius, A. Virus entry by endocytosis. Annu. Rev. Biochem. 2010, 79, 803–833. [Google Scholar] [CrossRef] [Green Version]
  10. Blaess, M.; Kaiser, L.; Sauer, M.; Deigner, H. Lysosomotropic active compounds—Hidden protection against COVID-19/SARS-CoV-2 Infection? Preprints 2020. [Google Scholar] [CrossRef]
  11. Blaess, M.; Kaiser, L.; Sommerfeld, O.; Csuk, R.; Deigner, H.P. Drugs, metabolites, and lung accumulating small lysosomotropic molecules: Multiple targeting impedes SARS-CoV-2 infection and progress to COVID-19. Int. J. Mol. Sci. 2021, 22, 1797. [Google Scholar] [CrossRef] [PubMed]
  12. Schloer, S.; Brunotte, L.; Goretzko, J.; Mecate-Zambrano, A.; Korthals, N.; Gerke, V.; Ludwig, S.; Rescher, U. Targeting the endolysosomal host-SARS-CoV-2 interface by clinically licensed functional inhibitors of acid sphingomyelinase (FIASMA) including the antidepressant fluoxetine. Emerg. Microbes. Infect. 2020, 9, 2245–2255. [Google Scholar] [CrossRef]
  13. Chung, H.Y.; Claus, R.A. Keep your friends close, but your enemies closer: Role of acid sphingomyelinase during infection and host response. Front. Med. 2021, 21, 616500. [Google Scholar] [CrossRef] [PubMed]
  14. Li, X.; Zhu, W.; Fan, M.; Zhang, J.; Peng, Y.; Huang, F.; Wang, N.; He, L.; Zhang, L.; Holmdahl, R.; et al. Dependence of SARS-CoV-2 infection on cholesterol-rich lipid raft and endosomal acidification. Comput. Struct. Biotechnol. J. 2021, 19, 1933–1943. [Google Scholar] [CrossRef] [PubMed]
  15. Cava, C.; Bertoli, G.; Castiglioni, I. In silico discovery of candidate drugs against Covid-19. Viruses 2020, 12, 404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Mirabelli, C.; Wotring, J.W.; Zhang, C.J.; McCarty, S.M.; Fursmidt, R.; Frum, T.; Kadambi, N.S.; Amin, A.T.; O’Meara, T.R.; Pretto, C.D.; et al. RMorphological cell profiling of SARS-CoV-2 infection identifies drug repurposing candidates for COVID-19. bioRxiv 2020. [Google Scholar] [CrossRef]
  17. Yang, L.; Pei, R.J.; Li, H.; Ma, X.N.; Zhou, Y.; Zhu, F.H.; He, P.L.; Tang, W.; Zhang, Y.C.; Xiong, J.; et al. Identification of SARS-CoV-2 entry inhibitors among already approved drugs. Acta Pharmacol. Sin. 2020, 28, 1–7. [Google Scholar] [CrossRef] [PubMed]
  18. Chen, C.Z.; Shinn, P.; Itkin, Z.; Eastman, R.T.; Bostwick, R.; Rasmussen, L.; Huang, R.; Shen, M.; Hu, X.; Wilson, K.M.; et al. Drug repurposing screen for compounds inhibiting the cytopathic effect of SARS-CoV-2. Front Pharmacol. 2021, 11, 592737. [Google Scholar] [CrossRef]
  19. Villoutreix, B.O.; Krishnamoorthy, R.; Tamouza, R.; Leboyer, M.; Beaune, P. Chemoinformatic analysis of psychotropic and antihistaminic drugs in the light of experimental anti-SARS-CoV-2 activities. Adv. Appl. Bioinform. Chem. 2021, 14, 71–85. [Google Scholar] [CrossRef] [PubMed]
  20. Castaldo, N.; Aimo, A.; Castiglione, V.; Padalino, C.; Emdin, M.; Tascini, C. Safety and efficacy of amiodarone in a patient with COVID-19. JACC Case Rep. 2020, 2, 1307–1310. [Google Scholar] [CrossRef]
  21. Arya, R.; Das, A.; Prashar, V.; Kumar, M. Potential inhibitors against papain-like protease of novel coronavirus (SARS-CoV-2) from FDA approved drugs. ChemRxiv Preprint 2020. [Google Scholar] [CrossRef]
  22. Sencanski, M.; Perovic, V.; Pajovic, S.B.; Adzic, M.; Paessler, S.; Glisic, S. Drug repurposing for candidate SARS-CoV-2 main protease inhibitors by a novel in silico method. Molecules 2020, 25, 3830. [Google Scholar] [CrossRef]
  23. Dayer, M.R. Old drugs for newly emerging viral disease, COVID-19: Bioinformatic prospective. arXiv 2020, arXiv:2003.04524. [Google Scholar]
  24. Dayer, M.R. Old drugs for JAK-STAT pathway inhibition in COVID-19. arXiv 2020, arXiv:2010.12332. [Google Scholar]
  25. Zhou, Y.; Hou, Y.; Shen, J.; Mehra, R.; Kallianpur, A.; Culver, D.A.; Gack, M.U.; Farha, S.; Zein, J.; Comhair, S.; et al. A network medicine approach to investigation and population-based validation of disease manifestations and drug repurposing for COVID-19. PLoS Biol. 2020, 18, e3000970. [Google Scholar] [CrossRef]
  26. Hsieh, K.; Wang, Y.; Chen, L.; Zhao, Z.; Savitz, S. Drug repurposing for COVID-19 using graph neural network with genetic, mechanistic, and epidemiological validation. Res. Sq. 2020. [Google Scholar] [CrossRef]
  27. Straus, M.R.; Bidon, M.; Tang, T.; Whittaker, G.; Daniel, S. FDA approved calcium channel blockers inhibit SARS-CoV-2 infectivity in epithelial lung cells. bioRxiv 2020. [Google Scholar] [CrossRef]
  28. 28 Xiao, X.; Wang, C.; Chang, D.; Wang, Y.; Dong, X.; Jiao, T.; Zhao, Z.; Ren, L.; Dela Cruz, C.S.; Sharma, L.; et al. Identification of potent and safe antiviral therapeutic candidates against SARS-CoV-2. Front. Immunol. 2020, 11, 586572. [Google Scholar] [CrossRef]
  29. Zhang, L.; Sun, Y.; Zeng, H.L.; Peng, Y.; Jiang, X.; Shang, W.J. Calcium channel blocker amlodipine besylate is associated with reduced case fatality rate of COVID-19 patients with hypertension. Cell Discov. 2020, 6, 96. [Google Scholar] [CrossRef] [PubMed]
  30. Solaimanzadeh, I. Nifedipine and amlodipine are associated with improved mortality and decreased risk for intubation and mechanical ventilation in elderly patients hospitalized for COVID-19. Cureus 2020, 12, e8069. [Google Scholar] [PubMed]
  31. Darquennes, G.; Le Corre, P.; Le Moine, O.; Loas, G. Association between functional inhibitors of acid sphingomyelinase (FIASMAs) and reduced risk of death in COVID-19 patients: A retrospective cohort study. Pharmaceuticals 2021, 14, 226. [Google Scholar] [CrossRef]
  32. Nouri-Vaskeh, M.; Kalami, N.; Zand, R.; Soroureddin, Z.; Varshochi, M.; Ansarin, K.; Rezaee, H.; Taghizadieh, A.; Sadeghi, A.; Ahangari Maleki, M.; et al. A comparison of losartan and amlodipine effects on the outcomes of patient with COVID-19 and primary hypertension: A randomised clinical trial. Int. J. Clin. Pract. 2021, e14124. [Google Scholar] [CrossRef]
  33. Vatansever, E.C.; Yang, K.S.; Drelich, A.K.; Kratch, K.C.; Cho, C.C.; Kempaiah, K.R.; Hsu, J.C.; Mellott, D.M.; Xu, S.; Tseng, C.K.; et al. Bepridil is potent against SARS-CoV-2 in vitro. Proc. Natl. Acad. Sci. USA 2021, 118, e2012201118. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, Y.; Hou, Y.; Shen, J.; Huang, Y.; Martin, W.; Cheng, F. Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2. Cell Discov. 2020, 16, 14. [Google Scholar] [CrossRef] [Green Version]
  35. Sauvat, A.; Ciccosanti, F.; Colavita, F.; Di Rienzo, M.; Castilletti, C.; Capobianchi, M.R.; Kepp, O.; Zitvogel, L.; Fimia, G.M.; Piacentini, M.; et al. On-target versus off-target effects of drugs inhibiting the replication of SARS-CoV-2. Cell. Death Dis. 2020, 11, 656. [Google Scholar] [CrossRef]
  36. Jeon, S.; Ko, M.; Lee, J.; Choi, I.; Byun, S.Y.; Park, S.; Shum, D.; Kim, S. Identification of antiviral drug candidates against SARS-CoV-2 from FDA-approved drugs. Antimicrob. Agents Chemother. 2020, 64, e00819–e00820. [Google Scholar] [CrossRef]
  37. Chen, C.Z.; Xu, M.; Pradhan, M.; Gorshkov, K.; Petersen, J.D.; Straus, M.R.; Zhu, W.; Shinn, P.; Guo, H.; Shen, M.; et al. Identifying SARS-CoV-2 entry inhibitors through drug repurposing screens of SARS-S and MERS-S pseudotyped particles. ACS Pharmacol. Transl. Sci. 2020, 3, 1165–1175. [Google Scholar] [CrossRef]
  38. Li, S.; Liu, W.; Chen, Y.; Wang, L.; An, W.; An, X.; Song, L.; Tong, Y.; Fan, H.; Lu, C. Transcriptome analysis of cepharanthine against a SARS-CoV-2-related coronavirus. Brief Bioinform. 2021, 22, 1378–1386. [Google Scholar] [CrossRef] [PubMed]
  39. Ruan, Z.; Liu, C.; Guo, Y.; He, Z.; Huang, X.; Jia, X.; Yang, T. SARS-CoV-2 and SARS-CoV: Virtual screening of potential inhibitors targeting RNA-dependent RNA polymerase activity (NSP12). J. Med. Virol. 2021, 93, 389–400. [Google Scholar] [CrossRef] [PubMed]
  40. Fan, H.H.; Wang, L.Q.; Liu, W.L.; An, X.P.; Liu, Z.D.; He, X.Q.; Song, L.H.; Tong, Y.G. Repurposing of clinically approved drugs for treatment of coronavirus disease 2019 in a 2019-novel coronavirus-related coronavirus model. Chin. Med. J. 2020, 133, 1051–1056. [Google Scholar] [CrossRef] [PubMed]
  41. Ginex, T.; Garaigorta, U.; Ramírez, D.; Castro, V.; Nozal, V. Host-directed FDA-approved drugs with antiviral activity against SARS-CoV-2 identified by hierarchical in silico/in vitro screening method. Pharmaceuticals 2021, 14, 332. [Google Scholar] [CrossRef]
  42. Weston, S.; Haupt, R.; Logue, J.; Matthews, K.; Frieman, M. BFDA approved drugs with broad anti-coronaviral activity inhibit SARS-CoV-2 in vitro. BioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  43. Plaze, M.; Attali, D.; Prot, M.; Petit, A.C.; Blatzer, M.; Vinckier, F.; Levillayer, L.; Chiaravalli, J.; Perin-Dureau, F.; Cachia, A.; et al. Inhibition of the replication of SARS-CoV-2 in human cells by the FDA-approved drug chlorpromazine. Int. J. Antimicrob. Agents 2021, 57, 106274. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, Y.; Chan, W.K.B.; Wang, Z.; Hur, J.; Xie, J.; Yu, H.; He, Y. Ontological and bioinformatic analysis of anti-coronavirus drugs and their implication for drug repurposing against COVID-19. Preprints 2020. [Google Scholar] [CrossRef]
  45. Weston, S.; Coleman, C.M.; Haupt, R.; Logue, J.; Matthews, K.; Li, Y.; Reyes, H.M.; Weiss, S.R.; Frieman, M.B. Broad anti-coronavirus activity of food and drug administration-approved drugs against SARS-CoV-2 in vitro and SARS-CoV in vivo. J. Virol. 2020, 94, e01218–e01220. [Google Scholar] [CrossRef] [PubMed]
  46. Hoertel, N.; Sánchez-Rico, M.; Vernet, R.; Jannot, A.S.; Neuraz, A.; Blanco, C.; Lemogne, C.; Airagnes, G.; Paris, N.; Daniel, C.; et al. AP-HP/Universities/INSERM COVID-19 research collaboration and AP-HP COVID CDR initiative. Observational study of chlorpromazine in hospitalized patients with COVID-19. Clin. Drug. Investig. 2021, 41, 221–233. [Google Scholar] [CrossRef] [PubMed]
  47. Gordon, D.E.; Jang, G.M.; Bouhaddou, M.; Xu, J.; Obernier, K.; O’Meara, M.J.; Guo, J.Z.; Swaney, D.L.; Tummino, T.A.; Huttenhain, R.; et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 2020. [Google Scholar] [CrossRef]
  48. Norinder, U.; Tuck, A.; Norgren, K.; Munic Kos, V. Existing highly accumulating lysosomotropic drugs with potential for repurposing to target COVID-19. Biomed. Pharmacother. 2020, 130, 110582. [Google Scholar] [CrossRef]
  49. Ke, Y.Y.; Peng, T.T.; Yeh, T.K.; Huang, W.Z.; Chang, S.E.; Wu, S.H.; Hung, H.C.; Hsu, T.A.; Lee, S.J.; Song, J.S.; et al. Artificial intelligence approach fighting COVID-19 with repurposing drugs. Biomed. J. 2020, 43, 355–362. [Google Scholar] [CrossRef]
  50. Wan, W.; Zhu, S.; Li, S.; Shang, W.; Zhang, R.; Li, H.; Liu, W.; Xiao, G.; Peng, K.; Zhang, L. High-throughput screening of an FDA-approved drug library identifies inhibitors against arenaviruses and SARS-CoV-2. ACS Infect. Dis. 2020. [Google Scholar] [CrossRef]
  51. Yuan, S.; Yin, X.; Meng, X.; Chan, J.F.; Ye, Z.W.; Riva, L.; Pache, L.; Chan, C.C.; Lai, P.M.; Chan, C.C.; et al. Clofazimine broadly inhibits coronaviruses including SARS-CoV-2. Nature 2021, 593, 418–423. [Google Scholar] [CrossRef]
  52. Xiong, H.L.; Cao, J.L.; Shen, C.G.; Ma, J.; Qiao, X.Y.; Shi, T.S.; Ge, S.X.; Ye, H.M.; Zhang, J.; Yuan, Q.; et al. Several FDA-approved drugs effectively inhibit SARS-CoV-2 infection in vitro. Front. Pharmacol. 2021, 11, 609592. [Google Scholar] [CrossRef] [PubMed]
  53. Jehi, L.; Ji, X.; Milinovich, A.; Erzurum, S.; Rubin, B.P.; Gordon, S.; Young, J.B.; Kattan, M.W. Individualizing risk prediction for positive coronavirus disease 2019 testing: Results from 11,672 patients. Chest 2020, 158, 1364–1375. [Google Scholar] [CrossRef]
  54. Hoertel, N.; Sánchez-Rico, M.; Vernet, R.; Beeker, N.; Jannot, A.S.; Neuraz, A.; Salamanca, E.; Paris, N.; Daniel, C.; Gramfort, A.; et al. Association between antidepressant use and reduced risk of intubation or death in hospitalized patients with COVID-19: Results from an observational study. Mol. Psychiatry 2021, 1–14. [Google Scholar] [CrossRef]
  55. Fred, M.S.; Kuivanen, S.; Ugurlu, H.; Casarotto, P.C. Antidepressant and antipsychotic drugs reduce viral infection by SARS-CoV-2 and fluoxetine show antiviral activity against the novel variants in vitro. BioRxiv 2021. [Google Scholar] [CrossRef]
  56. Hou, Y.; Ge, S.; Li, X.; Wang, C.; He, H.; He, L. Testing of the inhibitory effects of loratadine and desloratadine on SARS-CoV-2 spike pseudotyped virus viropexis. Chem. Biol. Interact. 2021, 338, 109420. [Google Scholar] [CrossRef] [PubMed]
  57. Gelemanović, A.; Vidović, T.; Stepanić, V.; Trajković, K. Identification of 37 heterogeneous drug candidates for treatment of COVID-19 via a rational transcriptomics-based drug repurposing approach. Pharmaceuticals 2021, 14, 87. [Google Scholar] [CrossRef]
  58. Ge, S.; Wang, X.; Hou, Y.; Lv, Y.; Wang, C.; He, H. Repositioning of histamine H(1) receptor antagonist: Doxepin inhibits viropexis of SARS-CoV-2 Spike pseudovirus by blocking ACE2. Eur. J. Pharmacol. 2021, 896, 173897. [Google Scholar] [CrossRef]
  59. Das, S.; Sarmah, S.; Lyndem, S.; Singha Roy, A. An investigation into the identification of potential inhibitors of SARS-CoV-2 main protease using molecular docking study. J. Biomol. Struct. Dyn. 2020, 13, 1–11. [Google Scholar] [CrossRef]
  60. Choy, K.T.; Wong, A.Y.L.; Kaewpreedee, P.; Sia, S.F.; Chen, D. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antivir. Res. 2020, 178, 104786. [Google Scholar] [CrossRef]
  61. Ianevski, A.; Yao, R.; Fenstad, M.H.; Biza, S.; Zusinaite, E.; Reisberg, T.; Lysvand, H.; Løseth, K.; Landsem, V.M.; Fossum, J.; et al. Potential antiviral options against SARS-CoV-2 infection. Viruses 2020, 12, 642. [Google Scholar] [CrossRef] [PubMed]
  62. Yang, C.W.; Peng, T.T.; Hsu, H.Y.; Lee, Y.Z.; Wu, S.H.; Lin, W.H.; Ke, Y.Y.; Hsu, T.A.; Yeh, T.K.; Huang, W.Z.; et al. Repurposing old drugs as antiviral agents for coronaviruses. Biomed. J. 2002, 4, 368–374. [Google Scholar] [CrossRef] [PubMed]
  63. Kumar, R.; Afsar, M.; Khandelwal, N.; Chander, Y.; Riyesh, T.; Dedar, R.K.; Gulati, B.R.; Pal, Y.; Barua, S.; Tripathi, B.N.; et al. Emetine suppresses SARS-CoV-2 replication by inhibiting interaction of viral mRNA with eIF4E. Antivir. Res. 2021, 189, 105056. [Google Scholar] [CrossRef] [PubMed]
  64. Huang, C.T.; Chao, T.L.; Kao, H.C.; Pang, Y.H.; Lee, W.H.; Hsieh, C.H.; Chang, S.Y.; Huang, H.C.; Juan, H.F. Enhancement of the IFN-beta-induced host signature informs repurposed drugs for COVID-19. Heliyon 2020, 6, e05646. [Google Scholar] [CrossRef] [PubMed]
  65. Gurung, A.B.; Ali, M.A.; Lee, J.; Farah, M.A.; Al-Anazi, K.M. The potential of Paritaprevir and Emetine as inhibitors of SARS-CoV-2 RdRp. Saudi J. Biol. Sci. 2021, 28, 1426–1432. [Google Scholar] [CrossRef]
  66. Hajjo, R.; Tropsha, A. A systems biology workflow for drug and vaccine repurposing: Identifying small-molecule BCG mimics to reduce or prevent COVID-19 mortality. Pharm. Res. 2020, 37, 212. [Google Scholar] [CrossRef]
  67. Sisakht, M.; Mahmoodzadeh, A.; Darabian, M. Plant-derived chemicals as potential inhibitors of SARS-CoV-2 main protease (6LU7), a virtual screening study. Phytother. Res. 2021. [Google Scholar] [CrossRef]
  68. Zimniak, M.; Kirschner, L.; Hilpert, H.; Seibel, J.; Bodem, J. The serotonin reuptake inhibitor Fluoxetine inhibits SARS-CoV-2. Sci. Rep. 2021, 11, 5890. [Google Scholar] [CrossRef]
  69. Creeden, J.F.; Imami, A.S.; Eby, H.M.; Gillman, C.; Becker, K.N.; Reigle, J.; Andari, E.; Pan, Z.K.; O’Donovan, S.M.; McCullumsmith, R.E.; et al. Fluoxetine as an anti-inflammatory therapy in SARS-CoV-2 infection. Biomed. Pharmacother. 2021, 138, 111437. [Google Scholar] [CrossRef]
  70. Duarte Rodrigo, R.R.; Copertino Jr Dennis, C.; Iñiguez Luis, P.; Marston Jez, L.; Nixon, D.F.; Powell Timothy, R. Repurposing FDA-approved drugs for COVID-19 using a data-driven approach. Chem. Rxiv. Preprint 2020. [Google Scholar] [CrossRef]
  71. O’Donovan, S.M.; Imami, A.; Eby, H.; Henkel, N.D.; Creeden, J.F.; Asah, S.; Zhang, X.; Wu, X.; Alnafisah, R.; Taylor, R.T.; et al. Identification of candidate repurposable drugs to combat COVID-19 using a signature-based approach. Sci. Rep. 2021, 11, 4495. [Google Scholar] [CrossRef] [PubMed]
  72. Dechaumes, A.; Nekoua, M.P.; Belouzard, S.; Sane, F.; Engelmann, I.; Dubuisson, J.; Alidjinou, E.K.; Hober, D. Fluoxetine can inhibit SARS-CoV-2 in vitro. Microorganisms 2021, 9, 339. [Google Scholar] [CrossRef] [PubMed]
  73. Plaze, M.; Attali, D.; Petit, A.C.; Blatzer, M.; Simon-Loriere, E.; Vinckier, F.; Cachia, A.; Chrétien, F.; Gaillard, R. Repurposing chlorpromazine to treat COVID-19: The recovery study. Encephale 2020, S35–S39. [Google Scholar] [CrossRef] [PubMed]
  74. Nazeam, J.; Mohammed, E.Z.; Raafat, M.; Houssein, M.; Elkafoury, A.; Hamdy, D.; Jamil, L. Based on principles and insights of COVID-19 epidemiology, genome sequencing, and pathogenesis: Retrospective analysis of sinigrin and ProlixinRX (Fluphenazine) provides off-label drug candidates. SLAS Discov. 2020, 25, 1123–1140. [Google Scholar] [CrossRef] [PubMed]
  75. Lenze, E.J.; Mattar, C.; Zorumski, C.F.; Stevens, A.; Schweiger, J.; Nicol, G.E.; Miller, J.P.; Yang, L.; Yingling, M.; Avidan, M.S.; et al. Fluvoxamine vs placebo and clinical deterioration in outpatients with symptomatic COVID-19: A randomized clinical trial. JAMA 2020, 324, 2292–2300. [Google Scholar] [CrossRef]
  76. Seftel, D.; Boulware, D.R. Prospective cohort of fluvoxamine for early treatment of coronavirus disease 19. Open Forum Infect. Dis. 2021, 8, ofab050. [Google Scholar] [CrossRef] [PubMed]
  77. Reznikov, L.R.; Norris, M.H.; Vashisht, R.; Bluhm, A.P.; Li, D.; Liao, Y.J.; Brown, A.; Butte, A.J.; Ostrov, D.A. Identification of antiviral antihistamines for COVID-19 repurposing. Biochem. Biophys. Res. Commun. 2021, 538, 173–179. [Google Scholar] [CrossRef] [PubMed]
  78. Hoertel, N.; Sánchez, M.; Vernet, R.; Beeker, N.; Neuraz, A.; Blanco, C.; Olfson, M.; Lemogne, C.; Meneton, P.; Daniel, C.; et al. Association between hydroxyzine use and reduced mortality in patients hospitalized for coronavirus disease 2019: Results from a multicenter observational study. medRxiv 2020. [Google Scholar] [CrossRef]
  79. Artigas, L.; Coma, M.; Matos-Filipe, P.; Aguirre-Plans, J.; Farrés, J.; Valls, R.; Fernandez-Fuentes, N.; de la Haba-Rodriguez, J.; Olvera, A.; Barbera, J.; et al. In-silico drug repurposing study predicts the combination of pirfenidone and melatonin as a promising candidate therapy to reduce SARS-CoV-2 infection progression and respiratory distress caused by cytokine storm. PLoS ONE 2020, 15, e0240149. [Google Scholar] [CrossRef]
  80. Hazra, S.; Chaudhuri, A.G.; Tiwary, B.K.; Chakrabarti, N. Matrix metallopeptidase 9 as a host protein target of chloroquine and melatonin for immunoregulation in COVID-19: A network-based meta-analysis. Life Sci. 2020, 257, 118096. [Google Scholar] [CrossRef]
  81. Cheng, F.; Rao, S.; Mehra, R. COVID-19 treatment: Combining anti-inflammatory and antiviral therapeutics using a network-based approach. Cleve. Clin. J. Med. 2020. [Google Scholar] [CrossRef] [PubMed]
  82. Ramlall, V.; Zucker, J.; Tatonetti, N. Melatonin is significantly associated with survival of intubated COVID-19 patients. medRxiv 2020. [Google Scholar] [CrossRef]
  83. Marinho, E.M.; Batista de Andrade Neto, J.; Silva, J.; Rocha da Silva, C.; Cavalcanti, B.C.; Marinho, E.S.; Nobre Júnior, H.V. Virtual screening based on molecular docking of possible inhibitors of Covid-19 main protease. Microb. Pathog. 2020, 148, 104365. [Google Scholar] [CrossRef]
  84. Salas Rojas, M.; Silva Garcia, R.; Bini, E.; Pérez de la Cruz, V.; León Contreras, J.C.; Hernández Pando, R.; Bastida Gonzalez, F.; Davila-Gonzalez, E.; Orozco Morales, M.; Gamboa Domínguez, A.; et al. Quinacrine, an antimalarial drug with strong activity inhibiting SARS-CoV-2 viral replication in vitro. Viruses 2021, 13, 121. [Google Scholar] [CrossRef]
  85. Han, Y.; Duan, X.; Yang, L.; Nilsson-Payant, B.E.; Wang, P.; Duan, F.; Tang, X.; Yaron, T.M.; Zhang, T.; Uhl, S.; et al. Identification of SARS-CoV-2 inhibitors using lung and colonic organoids. Nature 2021, 589, 270–275. [Google Scholar] [CrossRef]
  86. Puhl, A.C.; Fritch, E.J.; Lane, T.R.; Tse, L.V.; Yount, B.L.; Sacramento, C.Q.; Fintelman-Rodrigues, N.; Tavella, T.A.; Maranhão Costa, F.T.; Weston, S.; et al. Repurposing the Ebola and Marburg virus inhibitors tilorone, quinacrine, and pyronaridine: In vitro activity against SARS-CoV-2 and potential mechanisms. ACS Omega 2021, 6, 7454–7468. [Google Scholar] [CrossRef]
  87. Udrea, A.M.; Avram, S.; Nistorescu, S.; Pascu, M.L.; Romanitan, M.O. Laser irradiated phenothiazines: New potential treatment for COVID-19 explored by molecular docking. J. Photochem. Photobiol. B 2020, 211, 111997. [Google Scholar] [CrossRef]
  88. Birlutiu, V.; Birlutiu, R.M.; Iancu, G.M. Pityriasis rosea Gibert triggered by SARS-CoV-2 infection: A case report. Medicine 2021, 100, e25352. [Google Scholar] [CrossRef]
  89. Janabi, A.H.D. Molecular docking analysis of anti-severe acute respiratory syndrome-coronavirus 2 ligands against spike glycoprotein and the 3-chymotrypsin-like protease. J. Med. Signals Sens. 2021, 11, 31–36. [Google Scholar] [CrossRef] [PubMed]
  90. Werner, J.; Kronberg, R.M.; Stachura, P.; Ostermann, P.N.; Müller, L.; Schaal, H.; Bhatia, S.; Kather, J.N.; Borkhardt, A.; Pandyra, A.A.; et al. Deep transfer learning approach for automatic recognition of drug toxicity and inhibition of SARS-CoV-2. Viruses 2021, 13, 610. [Google Scholar] [CrossRef] [PubMed]
  91. Jade, D.; Ayyamperumal, S.; Tallapaneni, V.; Joghee Nanjan, C.M.; Barge, S.; Mohan, S.; Nanjan, M.J. Virtual high throughput screening: Potential inhibitors for SARS-CoV-2 PLPRO and 3CLPRO proteases. Eur. J. Pharmacol. 2021, 901, 174082. [Google Scholar] [CrossRef]
  92. Liu, D.Y.; Liu, J.C.; Liang, S.; Meng, X.H.; Greenbaum, J.; Xiao, H.M.; Tan, L.J.; Deng, H.W. Drug repurposing for COVID-19 treatment by integrating network pharmacology and transcriptomics. Pharmaceutics 2021, 13, 545. [Google Scholar] [CrossRef]
  93. Wang, X.; Lu, J.; Ge, S.; Hou, Y.; Hu, T.; Lv, Y.; Wang, C.; He, H. Astemizole as a drug to inhibit the effect of SARS-COV-2 in vitro. Microb. Pathog. 2021, 156, 104929. [Google Scholar] [CrossRef]
  94. Ohashi, H.; Watashi, K.; Saso, W.; Shionoya, K.; Iwanami, S.; Hirokawa, T.; Shirai, T.; Kanaya, S.; Ito, Y.; Kim, K.S.; et al. Potential anti-COVID-19 agents, cepharanthine and nelfinavir, and their usage for combination treatment. iScience 2021, 24, 102367. [Google Scholar] [CrossRef]
  95. Le, B.L.; Andreoletti, G.; Oskotsky, T.; Vallejo-Gracia, A.; Rosales, R.; Yu, K.; Kosti, I.; Leon, K.E.; Bunis, D.G.; Li, C.; et al. Transcriptomics-based drug repositioning pipeline identifies therapeutic candidates for COVID-19. Sci. Rep. 2021, 11, 12310. [Google Scholar] [CrossRef] [PubMed]
  96. Imamura, K.; Sakurai, Y.; Enami, T.; Shibukawa, R.; Nishi, Y.; Ohta, A.; Shu, T.; Kawaguchi, J.; Okada, S.; Hoenen, T.; et al. iPSC screening for drug repurposing identifies anti-RNA virus agents modulating host cell susceptibility. FEBS Open Bio. 2021, 11, 1452–1464. [Google Scholar] [CrossRef] [PubMed]
  97. Gorshkov, K.; Chen, C.Z.; Bostwick, R.; Rasmussen, L.; Tran, B.N.; Cheng, Y.S.; Xu, M.; Pradhan, M.; Henderson, M.; Zhu, W.; et al. The SARS-CoV-2 cytopathic effect is blocked by lysosome alkalizing small molecules. ACS Infect Dis. 2021, 7, 1389–1408. [Google Scholar] [CrossRef] [PubMed]
  98. Ren, P.X.; Shang, W.J.; Yin, W.C.; Ge, H.; Wang, L.; Zhang, X.L.; Li, B.Q.; Li, H.L.; Xu, Y.C.; Xu, E.H.; et al. A multi-targeting drug design strategy for identifying potent anti-SARS-CoV-2 inhibitors. Acta Pharmacol. Sin. 2021, 27, 1–11. [Google Scholar] [CrossRef]
  99. Eberle, R.J.; Olivier, D.S.; Amaral, M.S.; Gering, I.; Willbold, D.; Arni, R.K.; Coronado, M.A. The repurposed drugs suramin and quinacrine cooperatively inhibit SARS-CoV-2 3CL(pro) in vitro. Viruses 2021, 13, 873. [Google Scholar] [CrossRef]
  100. Carpinteiro, A.; Gripp, B.; Hoffmann, M.; Pöhlmann, S.; Hoertel, N.; Edwards, M.J.; Kamler, M.; Kornhuber, J.; Becker, K.A.; Gulbins, E. Inhibition of acid sphingomyelinase by ambroxol prevents SARS-CoV-2 entry into epithelial cells. J. Biol. Chem. 2021, 296, 100701. [Google Scholar] [CrossRef]
  101. Thompson, L.A.; Gurka, M.J.; Filipp, S.L.; Schatz, D.A.; Mercado, R.E.; Ostrov, D.A.; Atkinson, M.A.; Rasmussen, S.A. The influence of selection bias on identifying an association between allergy medication use and SARS-CoV-2 infection. E Clin. Med. 2021, 4, 100936. [Google Scholar] [CrossRef]
  102. Chavarría, A.P.; Vázquez, R.R.V.; Cherit, J.G.D.; Bello, H.H.; Suastegui, H.C.; Moreno-Castañeda, L.; Alanís Estrada, G.; Hernández, F.; González-Marcos, O.; Saucedo-Orozco, H.; et al. Antioxidants and pentoxifylline as coadjuvant measures to standard therapy to improve prognosis of patients with pneumonia by COVID-19. Comput. Struct. Biotechnol. J. 2021, 19, 1379–1390. [Google Scholar] [CrossRef]
  103. Hoertel, N.; Sánchez-Rico, M.; Gulbins, E.; Kornhuber, J.; Carpinteiro, A.; Lenze, E.J.; Reiersen, A.M.; Abellán, M.; De La Muela, P.; Vernet, R.; et al. Association between FIASMAs and reduced risk of intubation or death in individuals hospitalized for severe COVID-19: An observational multicenter study. Clin. Pharmacol. Ther. 2021. [Google Scholar] [CrossRef] [PubMed]
  104. Törnquist, K.; Asghar, M.Y.; Srinivasan, V.; Korhonen, L.; Lindholm, D. Sphingolipids as modulators of SARS-CoV-2 infection. Front. Cell. Dev. Biol. 2021, 9, 689854. [Google Scholar] [CrossRef] [PubMed]
  105. Tummino, T.A.; Rezelj, V.V.; Fischer, B.; Fischer, A.; O’Meara, M.J.; Monel, B.; Vallet, T.; White, K.M.; Zhang, Z.; Alon, A.; et al. Drug-induced phospholipidosis confounds drug repurposing for SARS-CoV-2. Science 2021, eabi4708. [Google Scholar] [CrossRef]
  106. Zhang, L.; Peng, X.; Ai, Y.; Li, L.; Zhao, S.; Liu, Z.; Peng, Q.; Deng, S.; Huang, Y.; Mo, Y.; et al. Amitriptyline reduces sepsis-induced brain damage through TrkA signaling pathway. J. Mol. Neurosci. 2020, 70, 2049–2057. [Google Scholar] [CrossRef]
  107. Sukhatme, V.P.; Reiersen, A.M.; Vayttaden, S.J.; Sukhatme, V.V. Fluvoxamine: A review of its mechanism of action and its role in COVID-19. Front. Pharmacol. 2021, 12, 652688. [Google Scholar] [CrossRef]
Table 1. Functional inhibitors of acid sphingomyelinase (FIASMAs) with activity against SARS-CoV-2 on in silico, or/and in vitro models or/and in vivo models (N = 49).
Table 1. Functional inhibitors of acid sphingomyelinase (FIASMAs) with activity against SARS-CoV-2 on in silico, or/and in vitro models or/and in vivo models (N = 49).
FIASMAsIn Silico(References)In Vitro (References)In Vivo(References)
Ambroxol [100]
Amiodarone□□□[15,19,26]■■■[12,17,18]❖❖†[20,103]
Amitriptyline □□□□□□[19,21,22,23,24,25][3]❖†❖†[54,103]
Amlodipine □□□□□[19,23,24,26,89]■■■[27,28,29]❖❖❖❖†❖[29,30,31,32,103]
Astemizole[93]
Benztropine ■■[42,45]
Bepridil□□[19,22][33]
Carvedilol□□□[19,25,34] ❖❖[25,53]
Cepharanthine□□[35,39]■■■■■■[36,37,38,40,41,94]
Chlorpromazine□□□□[19,26,35,43]■■■■■[42,43,44,45,55]❖❖❖†[45,46,73]
Chlorprothixene[19][18]
Clemastine□□□□[19,35,47,48]■■[17,18]
Clofazimine□□[19,91]■■■■■■[28,41,49,50,51,95][51]
Clomiphene ■■[52,96]
Clomipramine[19]■■■■[42,45,55,97]❖†[54]
Cloperastine□□[35,47]
Cyclobenzaprine[19]
Cyproheptadine[19]
Desipramine[19]■■[3,55]
Desloratadine□□[19,48][56]
Dicycloverine [95]
Dilazep[57]
Doxepine [58]
Emetine□□□□□□ [35,59,65,66,67,90]■■■■■■[60,61,62,63,64,98]
Flunarizine[19][18]
Fluoxetine□□□□□[19,57,69,70,71]■■■■■■[3,12,18,55,68,72]❖❖†[54,103]
Fluphenazine□□[19,74]■■[42,45]
Flupenthixol[70]■■[17,55]
Fluvoxamine [55]❖❖[75,76]
Hydroxyzine[48][77]❖❖❖†❖†[77,78,101,103]
Imipramine□□[19,22]■■■[3,12,55]
Loperamide ■■[18,36]❖†[103]
Loratadine[19][56]❖❖[77,88]
Maprotiline[19]■■[3,18]
Melatonine□□□□□□[25,26,34,79,80,81] ❖❖❖❖[25,53,82,102]
Nortriptyline[57]
Paroxetine[34][43]❖❖❖†[53,54,103]
Perphenazine[19]
Pimozide ■■[33,55]
Promazine[19]
Promethazine□□□[19,48,92][45]
Protriptyline[19]
Quinacrine□□□□[34,48,71,83]■■■■[84,85,86,99]
Sertraline ■■[13,28]❖†[54]
Tamoxifene□□[48,92]■■[45,96]
Thioridazine□□[71,87][28]
Trifluoperazine□□[19,70][28]
Triflupromazine[19]
Trimipramine□□[19,22][37]
In bold: 9 drugs active against the 3 coronaviruses; in silico (□), in vitro (■), in vivo (❖) and negative result (†).
Table 2. Functional inhibitors of acid sphingomyelinase (FIASMAs) (N = 15) with activity against SARS-CoV-2 in epidemiological, clinical studies, or case reports.
Table 2. Functional inhibitors of acid sphingomyelinase (FIASMAs) (N = 15) with activity against SARS-CoV-2 in epidemiological, clinical studies, or case reports.
FIASMA (Reference)Study DesignSample SizeFIASMA PrevalenceOutcome
Amiodarone [20]* Case report1100%Case report of a 74-year-old man affected by respiratory failure related to COVID-19 who recovered after only supportive measures and amiodarone lasted 5 days.
[103]* Retrospective26021.27%Mortality or intubation on hospitalized COVID-19 patients (N = 33 on amiodarone, N = 2569 without FIASMAs) HR = 1.26 (p = 0.14).
Amitriptyline [54]* Retrospective69240.56%Mortality or intubation on hospitalized COVID-19 patients (N = 39 on amitriptyline, N = 6885 without antidepressants) HR = 0.85 (p = 0.59).
[103]* Retrospective25890.77%Mortality or intubation on hospitalized COVID-19 patients (N = 20 on amitriptyline, N = 2569 without FIASMAs) HR = 0.54 (p = 0.12).
Amlodipine [29]Retrospective9619.8%Mortality on COVID-19 inpatients with hypertension as the only comorbidity. Patients on amlodipine (N = 19) or non-amlodipine (N = 77) had lower mortality (0% vs. 19.5%, p = 0.037).
[30]Retrospective6536.9% ?Mortality on elderly patients hospitalized for COVID-19; 24 were on amlodipine or nifedipine and 41 were not, 50% survived in the amlodipine or nifedipine group and 14.6% in the other group (p = 0.0036).
[31]Retrospective31718.9%Mortality on hospitalized COVID-19 patients; 60 were on amlodipine and 257 were not. Multiple logistic regression found lower mortality on patients on amlodipine (OR = 0.24, p = 0.0031).
[32]* Prospective randomized8048.7%Mortality. Losartan (N = 41) and amlodipine (N = 39) on patients with COVID-19 and primary hypertension. No significant difference of 30-day mortality rate.
[103]* Retrospective26663.64%Mortality or intubation on hospitalized COVID-19 patients (N = 97 on amlodipine, N = 2569 without FIASMAs) HR = 0.7 (p = 0.037).
Carvedilol [25]Retrospective 26,7792.93%PCR-positive. Patients tested for COVID-19 in Cleveland Clinic Health System; Carvedilol use (N = 785) was significantly associated with reduced likelihood of PCR positive to SARS-CoV-2 (OR = 0.74; p < 0.05).
[53]Retrospective 11,6722.96%PCR-negative. Patients tested for COVID-19 in Cleveland Clinic Health System. Among 346 subjects on Carvedilol, 333 (96.2%) were PCR-negative and 13 (3.8%) were PCR-positive (p = 0.022).
Chlorpromazine [73]Observational Prevalence of COVID-19. Low rate (4%) of symptomatic COVID-19 infection in patients treated by antipsychotics than the rate (14%) observed in nurses or physicians in the same departments of psychiatry.
[46]* Retrospective 14,3400.38%Mortality or intubation on hospitalized COVID-19 patients (N = 55 on chlorpromazine, N = 14,285 without chlorpromazine); 23.6% deaths on chlorpromazine and 9% deaths on subjects without chlorpromazine HR = 2.01 (p = 0.16).
Clomipramine [54]* Retrospective68940.13%Mortality or intubation on hospitalized COVID-19 patients (N = 9 on clomipramine, N = 6885 without antidepressants) HR = 0.44 (p = 0.4).
Desloratadine [103]* Retrospective 25760.27%Mortality or intubation on hospitalized COVID-19 patients (N = 7 on desloratadine, N = 2569 without FIASMAs) HR = 0.68 (p = 0.44).
Fluoxetine [54]* Retrospective 69150.43%Mortality or intubation on hospitalized COVID-19 patients (N = 30 on fluoxetine, N = 6885 without antidepressants) HR = 0.37 (p = 0.049).
[103]* Retrospective25830.54%Mortality or intubation on 2583 hospitalized COVID-19 patients (N = 14 on fluoxetine, N = 2569 without FIASMAs) HR = 0.3 (p = 0.082).
Fluvoxamine [75]Double-blind randomized 152 52.6%Clinical deterioration within 15 days. Fluvoxamine (N = 80) vs. placebo (N = 72) on non-hospitalized adults. Less clinical deterioration within 15 days of randomization in fluvoxamine group (0/80) than in placebo group (6 /72) (log-rank p = 0.009).
[76]Prospective 11357.5%Incidence of hospitalization was 0% (0 of 65) with fluvoxamine and 12.5% (6 of 48) without fluvoxamine (p = 0.005).
Hydroxyzine [77]Retrospective 219,0000.12%Incidence PCR-positive. Prior usage of hydroxyzine (N = 269) was associated with reduced incidence of positive SARS-CoV-2 in individuals 61 years and above.
[78]* Retrospective 73451.88%Mortality or intubation on hospitalized COVID-19 patients (N = 138) on hydroxyzine), (N = 7207) without hydroxyzine; HR = 0.42 (p = 0.001).
[101]Retrospective 230,3761.7%Incidence PCR-negative. Prior usage of hydroxyzine (N = 3909) was not associated with increased incidence of negative SARS-CoV-2 in individuals. Adjusted OR = 0.76 (p > 0.05).
[103]* Retrospective26001.19%Mortality or intubation on hospitalized COVID-19 patients (N = 31 on hydroxyzine, N = 2569 without FIASMAs) HR = 0.43 (p = 0.04).
Loperamide [103]* Retrospective25780.35%Mortality or intubation on hospitalized COVID-19 patients (N = 9 on loperamide, N = 2569 without FIASMAs) HR = 0.25 (p = 0.1).
Loratadine [77]Retrospective219,0000.13%Incidence PCR-positive. Prior usage of loratadine (N = 284) was associated with reduced incidence of positive SARS-CoV-2 in individuals 61 years and above.
[88]Case report 1100%Case report (54-year-old female) of pityriasis rosea gibert associated with COVID-19 infection hospitalized and treated with 200 mg/day hydrocortisone hemisuccinate and loratadine 20 mg/day. Two weeks after admission, the patient was discharged with a negative RT-PCR and without respiratory symptoms.
Melatonine [25]Retrospective 26,7793.94%Incidence PCR-positive. Patients tested for COVID-19 in Cleveland Clinic Health System. Melatonine use (n = 1055) was significantly associated with reduced likelihood of PCR-positive to SARS-CoV-2 (OR = 0.72; p < 0.05).
[53]Retrospective11,6724.53%Incidence PCR-positive. Patients tested for COVID-19 in Cleveland Clinic Health System. Among 529 subjects on melatonin, 513 (97%) were PCR-negative and 16 (3%) were PCR-positive (p = 0.001).
[82]Retrospective 791 Survival rate. Patients with COVID-19 infection. Melatonin exposure was associated with survival in COVID-19 patients.
[102]Prospective longitudinal (before-after) 11020%Survival scores. Five groups of 22 patients were receiving pentoxifylline and one group had also 5 mg of melatonine every 12 h for 5 days. The medications improved the survival scores, and several inflammation markers (CRP…) were diminished at the end of the treatment
Paroxetine [53]Retrospective11,672 Incidence PCR-positive. Patients tested for COVID-19 in Cleveland Clinic Health System (7% PCR+). Among subjects on paroxetine, there was significant higher PCR-.
[54]* Retrospective 69480.91%Mortality or intubation in hospitalized COVID-19 patients (N = 63 on paroxetine, N = 6885 without antidepressants) HR = 0.52 (p = 0.0006).
[103]* Retrospective26101.57%Mortality or intubation on 2610 hospitalized COVID-19 patients (N = 41 on paroxetine, N = 2569 without FIASMAs) HR = 0.66 (p = 0.13).
Sertraline [54]* Retrospective69070.32%Mortality or intubation in hospitalized COVID-19 patients (N = 22 on sertraline N = 6885 without antidepressants) HR = 0.68 (p = 0.13).
[103]* Retrospective25900.81%Mortality or intubation on 2590 hospitalized COVID-19 patients (N = 21 on sertraline, N = 2569 without FIASMAs) HR = 0.57 (p = 0.11).
* Studies exploring acute and not chronic intake of FIASMAs asking the question of non-obtaining the steady state allowing a maximal ASM inhibition; HR: hazard ratio; OR: odds ratio.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Loas, G.; Le Corre, P. Update on Functional Inhibitors of Acid Sphingomyelinase (FIASMAs) in SARS-CoV-2 Infection. Pharmaceuticals 2021, 14, 691. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14070691

AMA Style

Loas G, Le Corre P. Update on Functional Inhibitors of Acid Sphingomyelinase (FIASMAs) in SARS-CoV-2 Infection. Pharmaceuticals. 2021; 14(7):691. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14070691

Chicago/Turabian Style

Loas, Gwenolé, and Pascal Le Corre. 2021. "Update on Functional Inhibitors of Acid Sphingomyelinase (FIASMAs) in SARS-CoV-2 Infection" Pharmaceuticals 14, no. 7: 691. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14070691

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop