IMR Press / FBL / Volume 27 / Issue 9 / DOI: 10.31083/j.fbl2709257
Open Access Review
Combination Therapies against COVID-19
Show Less
1 School of Basic Medical Sciences, Nanchang University, 330047 Nanchang, Jiangxi, China
2 Queen Marry College, Nanchang University, 330047 Nanchang, Jiangxi, China
*Correspondence: luoqunfeng@ncu.edu.cn (Qunfeng Luo); zhangxiaokong@hotmail.com (Jin Zhang)
Academic Editor: Sang Heui Seo
Front. Biosci. (Landmark Ed) 2022, 27(9), 257; https://doi.org/10.31083/j.fbl2709257
Submitted: 23 June 2022 | Revised: 14 July 2022 | Accepted: 25 July 2022 | Published: 6 September 2022
(This article belongs to the Special Issue Vaccine and anti-viral drug development for SARS-CoV2)
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of Coronavirus disease 2019 (COVID-19), which was announced as a pandemic leading to devastating economic and medical burden worldwide. The virus attacks the organ system across the body by binding to its receptor (for example, angiotensin converting enzyme 2) on the surface of the host cell of various organs. The patients present with a variety of pathological symptoms ranging from fever, cough and cytokine storm to acute respiratory distress syndrome (ARDS). Many combination therapies have been developed to combat the disease, via blocking one or more processes of the viral life cycle and/or relieving host complications simultaneously. In this review, the progress of those combination therapies containing at least one small molecule is updated. We believe it’ll provide significant inspiration for further development of treatment strategy against SARS-CoV-2, especially its mutant variants.

Keywords
combination therapy
anti-SARS-CoV-2
complication
antiviral
anti-inflammation
1. SARS-CoV-2 Life Cycle

COVID-19 is an infectious disease caused by SARS-CoV-2 virus. It had killed more than 6.12 million of persons worldwide as of 29 March 2022. While the majority of patients with COVID-19 will recover without special treatment, a minority will manifest severe symptoms requiring hospitalization and even suffer serious complications, including ARDS, which may induce multi-organ dysfunction. Considering SARS-CoV-2 is a unprecedented virus and radical treatment against the resulting disease remain to be developed, a clear interpretation of the viral life cycle is essential for designing prophylactic and/or therapeutic strategies that target one or more processes of its life cycle. SARS-CoV-2 affects human starting from entering the nasopharynx by attaching to angiotensin converting enzyme 2 (ACE2) receptor-enriched epithelial cells of the nasal and oral mucosa. Then it goes down and attacks the lung, where the infection mainly takes place. The following processes include endocytosis, replication, transcription, assembly and release of virus [1].

SARS-CoV-2 invades human cells through interaction of the receptor binding domain (RBD) of its spike (S) protein with the host ACE2 on the cell surface [2]. The SARS-CoV-2 S protein belongs to class 1 viral fusion protein and it means that the S protein should be cleaved into S1 and S2 subunits by a fusion protein before functioning normally. S1 contains the receptor binding domain (RBD), which directly binds to the peptidase domain (PD) of ACE2, whereas S2 is responsible for the fusion between the viral envelope and the host cell membrane and internalization by endocytosis with ACE2 [3, 4]. Various proteases including cathepsins, trypsin, human airway trypsin-like proteases, furin and transmembrane protease serine protease (2/4) (TMPRSS) have been reported to cleave S protein in a proteolytic way in those harmful coronaviruses since 2003 [5, 6, 7, 8]. That a specific furin-like cleavage site was discovered in the S-protein genome sequence of SARS-CoV-2 indicates furin and furin-like proteases may be involved in the S protein cleavage [9]. We have known that SARS-CoV-2 uses TMPRSS2 as well as furin or furin-like proteases, for its interaction with the ACE2 receptor and entry into the host cell. The study by Hoffmann et al. [2] proposed a rational role of TMPRSS2 in proteolysis of S protein to S1 and S2 subunits for S protein priming and camostat mesylate, an inhibitor of TMPRSS2, blocked SARS-CoV-2 infection of lung cells. Furthermore, the data showed that both furin and TMPRSS2 could not replace each other functionally and suppression of either of them might interfere virus to bind to host cells. Indeed, many experimental pieces of evidence recently have made clear that these two proteins act synergistically in viral entry and infectivity and shed light on the combination of furin and TMPRSS2 inhibitors as potent antivirals against SARS-CoV-2 [10]. Further processing that the S1 subunit is removed in host cell endosomes is promoted by cathepsins, which eventually assists the fusion of viral envelope with the host membranes, viral RNA release, and replication [11]. Overall, virus S protein, ACE2 and the host proteases play the essential roles in SARS-CoV-2 entry in host cells. Other than ACE2, several other receptors were also discovered including DC-SIGN (also known as CD209), L-SIGN (also known as CD209L or CLEC4M), SIGLEC1 (also known as CD169, sialoadhesin or Siglec-1) [12, 13] and neuropilin-1 (NRP1, known to bind furin-cleaved substrates) [14].

In case of the release of the viral genome into the host cell, the SARS-CoV-2 RNA acts as messenger RNA (mRNA) and undergoes translation by using host cell machinery [15]. The viral genome comprises a positive-sense, single-stranded RNA molecule with about 29,900 nucleotides which encode about 31 proteins: nonstructural proteins (NSPs; 16 proteins), structural proteins (4 proteins), and accessory proteins (11 proteins) [16]. The structural proteins consist of S protein, envelope (E) and membrane (M) proteins which form the viral envelope, and nucleocapsid (N) protein which connects with the virus genome. After entering host cell, the NSPs domain is first translated into two polypeptides, both of which are further processed to produce papain like protease (PLpro) (NSP3), main protease (Mpro) (also known as 3-chymotrypsin-like protease (3CLpro); NSP5) [17], and RNA-dependent RNA polymerase (RdRp; NSP12) [18]. Interestingly, the process is promoted by host proteases at the first stage, and then, is strengthened by PLpro and Mpro. The viral RdRp is key for the replication of genetic material. The viral genome and the N structural protein are produced in the host cell cytoplasm, while other viral structural proteins including S, M, and E are finally synthesized in the endoplasmic reticulum and transferred to the Golgi apparatus. The viral RNA–N complex and S, M, and E proteins are then packaged together to form a virion by a budding process. Following packaging, the complete virus particles are transported to the cell surface in vesicles and released by exocytosis [19, 20]. These newly released virions continue their life cycle in other healthy cells.

2. Concomitant Symptoms Following Virus Infection

The shortest and the longest average incubation period of SARS-CoV-2 in China are 1.8 days and 12.8 days, respectively, mostly ranging from 3 to 7 days [21, 22]. As those with SARS and MERS, most patients with COVID-19 have a specific ground glass appearance on chest computed tomographic (CT) scans [23]. Besides, patients with COVID-19 exhibit similar biopsy features to that seen in SARS-CoV and MERS-CoV patients [24]. Presentations of SARS-CoV-2 frequently emerged include fever (>37.3 °C), coughing, fatigue, muscles ache, runny nose, haemoptysis, headache, diarrhea, and shortness of breath [25]. Most patients showed mild and moderate symptoms, however, some cases in severe condition may suffer ARDS, multi-organ failure, and even death [26]. The statistic data revealed that COVID-19 mortality ranges from 1.4% [27] to 4.3% [28] in different areas or hospitals.

As aforementioned above, SARS-CoV-2 enters into host cells via binding to the receptor ACE2. ACE2 exists generally in many organs and tissues, including nasal epithelium, oral mucosa, kidney, brain, lungs, etc., which indicates SARS-CoV-2 can attack multiple organs aside from the lung [29]. Indeed, SARS-CoV-2 causes a wide variety of symptoms across organ systems in patients with COVID-19. The involvement of the lungs by SARS-CoV-2 might result in ARDS, requiring intubation and admission to the intensive care unit. Individuals with life-threatening SARS-CoV-2 disease demonstrate related cytokine release syndrome (CRS) [30]. CRS appears to be a hazardous factor involved in different inflammatory pathways hastening lung parenchymal impairment and thromboembolism [31]. Severe lung involvement and mortality may be predicted early through lymphocytopenia and elevated signs of inflammatory factors [32]. Particularly, neurogenic pulmonary edema can be seen in invalids with extreme COVID-19 pneumonia. It is also defined as a non-cardiogenic interstitial pulmonary edema with a distribution of the peripheral lung zone that can be found in viral pneumonia [33].

SARS-CoV-2 is a potent inducer of inflammatory cytokines. The virus can activate immune cells and induce the secretion of inflammatory cytokines and chemokines directing to pulmonary vascular endothelial cells. Surge of cytokines can cause a cytokine storm and extensive inflammation all over the body of patients. Organ damage following SARS-CoV-2 invasion may be attributed to the cytokine storm or cytokine cascade [34]. Although respiratory failure from ARDS is the utmost reason for death, several death cases were reported to die from circulatory failure as a result of myocardial damage, which indicated the infection of SARS-CoV-2 might cause fulminant myocarditis [35]. Neurological syndromes caused by COVID-19 include anosmia and dysgeusia, ataxia, headache, dizziness, and unconsciousness [36]. The neuron-invading potency and possible role of SARS-CoV-2 in the patients with acute respiratory failure was first proposed by Li et al. [37]. Although COVID-19 is characterized by respiratory presentations, the relative virus can also attack the alimentary system. Most patients suffer diarrhea, nausea, or vomiting [38]. SARS-CoV-2 is also considered a causative agent for thyroiditis or thyrotoxicosis. And SARS-CoV-2 may be a potential trigger for autoimmune thyroid disease [39]. Once infected by SARS-CoV-2, various key organs across the body can be attacked, and possibly result in multiple organ involvement. More complications about this viral infection can be read in another review [33].

3. Combination Therapy as a Treatment Strategy Blocking One or More Processes of the Viral Life Cycle and Relieving the Complications

The whole viral lifetime involves attachment and entry to the host cell, translation, replication and release, during which time the patients manifest various symptoms ranging from cough to cytokine storm, even multi-organ dysfunction, etc. In order to combat the disease COVID-19 efficiently, not only the stages of the viral life cycle, but the complications should be considered for potential therapeutic intervention. As long as immunologic complications like macrophage activation syndrome (MAS) occur, anti-viral monotherapy is not enough and additional anti-inflammatory treatment should be added. Early detection and proper treatment of MAS and cytokine storm will reduce the incidence and mortality in COVID-19 patients. Therefore, a combination therapy with dual or multiple drugs encompassing one or more targets could be given more favor. Ideally, the combined use of drugs should have at least dual functions: inhibiting or killing the virus and relieving the complicated symptoms of infected patients. The former function is performed by anti-viral drugs which block RNA synthesis and virus invasion, and bind to receptor proteins on the surface of cells, and cell cycle protein, etc. The latter function is served mainly by anti-inflammatory drugs which control cytokine production, break down the basement membrane, regulate outer mitochondrial membrane permeability, stimulate activated B-cell and T-cell proliferation, etc. Other drugs serving the latter function include anti-oxidant, immunomodulator and relating symptom-relieving drugs.

Combination therapies have the advantage to improve treatment efficiency while decreasing concentration of individual drug. For example, remdesivir at 5.05 μM combined with omipalisib at 0.25 μM exhibited 79% viral inhibition, while the concentrations of single drug, remdesivir or omipalisib to achieve the same inhibition rate were about 20.18 μM or 1.97 μM, respectively [40]. And the lower drug concentration may reduce the adverse effect of each drug emerged when used alone. The limited efficacy of some drugs may also be enhanced by combination therapy. Furthermore, if the drugs selected for combined use target different objects (exhibit different mode of actions), combination of these drugs might enhance the overall activity by simultaneously engaging two or more pathways [40].

Drug combination therapy has been proved useful in treating virus infection disease [41]. Recently it also showes some application potential on COVID-19 patients. Some studies demonstrate that combination therapy for COVID-19 outpatients might decrease hospitalization and death by 89%. Particularly, in some cases, combination therapy displays excellent outcome. COVID-19 patients sometimes may suffer several devastating conditions, such as cytokine cascade, organ damage, and thrombosis. McCullough and co-authors recommended that combination therapy should be a vital standard for management of those with these devastating conditions [42]. It is exciting that combination therapy is effective against some new SARS-CoV-2 variants, which makes this treatment strategy invaluable especially when the SARS-CoV-2 virus presents fast-mutating characteristics.

In this review, we aim to summarize the outcome of the combination therapies against COVID-19. We focus on the combination therapies that contain at least one small molecule, such as remdesivir, umifenovir, or hydroxychloroquine (HCQ). While those combination therapies with both or more biomacromolecules, like antibody, nanobody, convalescent plasma or some other therapeutic proteins like interferon, are not included. You may read another relative review for this kind of combination therapies [43]. We deem that this review would provide an option for the scientific and rational therapeutic alliance against COVID-19.

4. Progress of Combination Therapy with at Least One Small Molecule
4.1 Combination Therapy with Small Molecules

In this section, we will summarize those combination therapies with two or more small molecules. Each combination therapy may comprise two virus-directed antivirals, one virus-directed antiviral and another host-directed, one antiviral and another complication-relieving drug (anti-inflammatory, antioxidant, etc.), one antiviral and its pharmacokinetic enhancer, or other antimicrobials, etc. And according to the composition, they are categorized to 6 groups, all of which will be presented in the next 6 chapters, respectively. Many studies of combination therapy involve remdesivir, so we give a brief introduction of it first. Remdesivir is a nucleotide analog and the high resemblance between its triphosphate form and adenosine triphosphate (ATP) enables it to function as a competing inhibitor of RNA synthesis (RdRp inhibitor). Remdesivir shows great potential in inhibting all the coronaviruses including SARS-CoV-2 [44]. Beigel et al. [45] completed a double-blind trial and they found that about half patients (total 1062 participants) treated with remdesivir had a shorter recovery time. Their data also indicate that remdesivir may prevent deterioration of the disease. Given the positive results of the trial, remdesivir became the first antiviral to be authorized by the Food and Drug Association for emergency use for hospitalized adult patients at the risk of serious illness [46]. Following the approval, the clinical performance of the drug is also closely supervised and updated with the new evidence [47]. It was found that remdesivir could lead to renal failure or liver dysfunction during therapeutic process of COVID-19 [48, 49]. Furthermore, a solidarity trial guided by the World Health Organization (WHO) demonstrated remdesivir had little or no benefit on hospitalized patients with COVID-19 [50]. Thus, on 20 November 2020, WHO recommended against its use in spite of state of illness of hospitalized patients. Nonetheless, recently, on 22 April 2022 WHO updated the conditional recommendation for the use of remdesivir in patients with non-severe COVID-19 at the highest risk of hospitalization. There have been many clinical trials on remdesivir in a completed, terminated or recruiting stage, which were designed in combination with other agents such as Interferon beta-1b, Interferon beta-1a, Tocilizumab, Lopinavir/Ritonavir, Merimepodib, DWJ1248, baricitinib, or dexamethasone (https://www.clinicaltrials.gov/ct2/results?cond=COVID19&term=remdesivir&cntry=&state=&city=&dist=).

4.1.1 Antivirals Acting on SARS-CoV-2

SARS-CoV-2 proteases Mpro and PLpro are promising targets for antiviral drug development for their vital roles in coronavirus replication. MG-101, a Mpro inhibitor, significantly improved the antiviral activity against SARS-CoV-2 Delta variant, when combined with PLpro inhibitor Sitagliptin. Similarly, enhanced anti-SARS-CoV-2 activity was also observed in combination therapy of two Mpro inhibitors, such as MG-101 plus Lycorine HCl or Nelfinavir. MG-101, combined with Lycorine HCl or Nelfinavir mesylate, led to a 3–4 log reduction in virus titer at 1 μM concentration of each drug compared with blank group. Overall, these experimental data suggest that the double suppression of Mpro and PLpro or suppressing the same protease with multiple drugs is an attractive approach to combat COVID-19 [51] (Table 1, Ref. [51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67]). GC376, a pre-clinical inhibitor against feline infectious peritonitis (corona) virus (FIPV), was shown to be a potent inhibitor of Mpro in Vero cells. Moreover, combination of GC376 with remdesivir could reinforce antiviral activity, indicating additive effect of the combination of RdRp inhibitor and protease inhibitor (Table 1) [52]. Molnupiravir, a prodrug of the nucleoside derivative N-hydroxycytidine (NHC), targets viral RNA polymerase and becomes the first authorized oral antiviral for COVID-19, though some mild adverse effects emerged including nausea, diarrhea, headache and insomnia, etc. [68, 69]. Most recently, it was found to exhibit synergisitic antiviral activity against Omicron infection in Calu-3 cells when combined with nirmatrelvir, a SARS-CoV-2 Mpro inhibitor [53]. Hattori et al. [54] discovered a small molecule compound with an indole moiety targeting Mpro. This compound blocked virus replication without viral breakthrough and exerted synergistic anti-SARS-CoV-2 activity in combination with remdesivir. Corilagin is an ellagitannin with a hexahydroxydiphenoyl group bridging over the 3-O and 6-O of the glucose core, as a medicinal herbal agent found in Euphorbia fischeriana, Euphorbia hyssopifolia, and other organisms. It was reported to function as non-nucleoside inhibitor of SARS-CoV-2 RdRp, possibly via inhibiting the conformational change of RdRp, a different mechanism compared with remdesivir. When combined with remdesivir, it exhibited additive inhibition against SARS-CoV-2 RdRp [55]. Sofosbuvir (SOF) and daclatasvir (DCV) are clinically approved direct-acting RdRp inhibitors against hepatitis C virus (HCV). Some in silico and in vitro studies suggest that SOF and DCV also have high affinity for SARS-CoV-2 RdRp. While other studies show little or no effect on preventing SARS-CoV-2 infection. Recently, Sadeghi et al. [56] reported promising outcomes in a clinical trial using the combination therapy SOF/DCV on moderate or severe COVID-19 patients. They found that SOF/DCV treatment increased 14-day clinical recovery rates and reduced hospitalization length compared with standard care alone [56]. Two similar SOF/DCV clinical trials, though in notably small scale, were also performed and provided preliminary data favoring the SOF/DCV combination or SOF/DCV/ ribavirin triple therapy [57, 58]. Velpatasvir (VEL) is known as an inhibitor of HCV NS5A protein. Recently it was also reported to be tailored to A chain and B chain active sites of the SARS-CoV-2 3CLpro [59]. A single-center, randomized controlled trial study was conducted to evaluate the efficacy of the SOF/VEL combination plus the national standard of care (hydroxychloroquine and lopinavir/ritonavir as well as supportive care) in patients with moderate to severe COVID-19 illness. And the outcome showed SOF/VEL was safe, however, combining SOF/VEL with the standard of care did not provide any benefit for the clinical improvement or mortality reduction [60]. Linoleic acid (LA) is an inflammatory response modulator, isolated from the traditional meal Vicia faba [61]. Toelzer et al. [62] resolved a 2.85-angstrom cryo–electron microscopy structure in which the receptor binding domains of SARS-CoV-2 S glycoprotein tightly binded LA in three composite binding pockets. LA binding stabilized a locked S conformation, leading to reduced ACE2 interaction in vitro. In human Caco-2 ACE2+ cells, LA supplementation synergized with remdesivir in inhibiting SARS-CoV-2 replication [62]. Recently, Li et al. [63] experimentally proved that glycyrrhizin (GR) inhibited SARS-CoV-2 infection through interaction with S protein and blocks attachment of recombinant S protein to host cells. Also, boswellic acid (BA) is reported to exhibit a high affinity for the functional S protein of SARS-CoV-2 [64]. A randomized clinical trial was conducted to explore the effect of GR/BA versus placebo on hospitalized patients with moderate SARS-CoV-2 infection. And a potent decline in serum C-reactive protein levels was observed in the GR/BA group in comparison with the placebo group, which reflected reduction of systemic inflammation by GR/BA treatment. Though many superior profiles were still observed including the reduced risks of hospitalization and mortality, credible safety, good tolerance, and extensive availability, with this combination, it’s limited by lack of group receiving GR or BA alone to compare with the combination group (ClinicalTrials.gov number, NCT04487964) [65]. The anti-inflammatory drug cepharanthine (CEP) and human immunodeficiency virus (HIV) protease inhibitor nelfinavir (NFV) were discovered more potent than remdesivir and other drugs currently in clinical trial through screening a group of authorized drugs in a SARS-CoV-2 infection cell assay. Further study demonstrated that cepharanthine inhibited SARS-CoV-2 entry through interfering with S protein engagement to its ACE2 receptor, while nelfinavir suppressed viral replication partly by inhibition of SARS-CoV-2 main protease. Consistent with their different modes of action, synergistic effect of this combination therapy (CEP/NFV) to limit SARS-CoV-2 proliferation was highlighted over a wide range of concentrations [66]. Bismuth drugs colloidal bismuth subcitrate (CBS) or bismuth subsalicylate (BSS), usually used in the treatment of gastroduodenal disorders, in combination with N-acetyl-L-cysteine (NAC) significantly diminished the viral load of lung and the pathologic condition in Syrian hamster infection model. The mechanism of this combination involved NAC plays a vital role in preventing the hydrolysis of bismuth drugs via forming stable coordination compound [Bi(NAC)3], and optimizing the pharmacokinetic profiles of CBS. Besides, the cocktail also demonstrated broad-spectrum antiviral activities against key viral cysteine enzymes/proteases such as PLpro, Mpro, helicase (Hel) and ACE2 (Table 1) [67].

Table 1.Combination therapies with antivirals acting on SARS-CoV-2.
Drugs for Combination Targets or action mechanisms Results Reference
MG-101 + Sitagliptin Mpro + PLpro Improved the antiviral effect against SARS-CoV-2 Delta variant [51]
MG-101 + Lycorine or Nelfinavir Mpro Enhanced anti-SARS-CoV-2 activity [51]
GC376 + remdesivir Mpro + RdRp Additive antiviral activity [52]
Molnupiravir + nirmatrelvir RdRp + Mpro Synergisitic antiviral activity [53]
An indole derivative+ remdesivir Mpro Synergistic activity [54]
Corilagin + remdesivir RdRp (with different mechanisms) Additive inhibitory effect [55]
SOF + DCV RdRp Clinical recovery rate was increased and hospitalization length reduced [56]
SOF + DCV; SOF + DCV + ribavirin RdRp Favoring this combination therapy [57, 58]
VEL + SOF + the national standard of care* Mpro + RdRp (1) Safe; (2) did not improve the clinical status or reduce mortality [59, 60]
Linoleic acid (LA) + remdesivir SARS-CoV-2 S glycoprotein Synergistic in inhibiting SARS-CoV-2 replication [61, 62]
Glycyrrhizin (GR) + boswellic acid (BA) SARS-CoV-2 S glycoprotein (1) Reduction of systemic inflammation; (2) reduced risks of hospitalization and mortality, being safe, well tolerated, and widely available [63, 64, 65]
Cepharanthine (CEP) + nelfinavir (NFV) SARS-CoV-2 S protein + Mpro Synergistic to limit SARS-CoV-2 proliferation [66]
NAC + CBS or BSS Cysteine enzymes (proteases) including PLpro, Mpro, helicase (Hel) and ACE2 Significantly diminished the viral load of lung and the pathologic condition [67]
* The national standard of care comprises hydroxychloroquine and lopinavir/ritonavir as well as supportive care.
4.1.2 Antivirals Acting on SARS-CoV-2 and Host Cell

As aforementioned, some host proteases facilitate SARS-CoV-2 entry in host cell and the virus rely on host translation system for replication, thus it’s also vital to block host relative protease and translation associated enzymes for combating SARS-CoV-2. IMU-838, a developmental dihydroorotate dehydrogenase (DHODH) inhibitor in phase II for autoimmune disease, showed enhanced in vitro anti-SARS-CoV-2 activity when combined with remdesivir [70] (Table 2, Ref. [2, 40, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88]). Biering et al. [71] identified compound B02, a human RAD51 inhibitor, exhibiting antiviral synergy with remdesivir after screening a library of FDA-approved and well-studied preclinical and clinical chemicals (Table 2). Besides, a synergy between remdesivir and emetine (an anti-protozoan drug against ameobiasis) was noticed that remdesivir at 6.25 μM in combination with emetine at 0.195 μM may achieve 64.9% inhibition in viral yield [72]. Baricitinib, a Janus kinase inhibitor, plus remdesivir showed better outcomes than remdesivir alone in reducing recovery time and accelerating clinical improvement among COVID-19 patients. Furthermore, severe adverse events were also reduced in the combination group (ClinicalTrials.gov number, NCT04401579) [73]. A living guideline from the WHO published 14 January 2022 strongly recommended the use of baricitinib as an alternative to interleukin-6 (IL-6) receptor blockers, in combination with corticosteroids, in patients with severe or critical COVID-19 (https://www.who.int/teams/health-care-readiness/covid-19/therapeutics). Of note, baricitinib, used alone or as a combination with remdesivir, may bring about some adverse effects, such as liver injury [89] and transient leukocytopenia [90]. Tipifarnib is a mighty farnesyltransferase inhibitor with the potential as an anticancer remedy and has completed phase III clinical trial [74]. Omipalisib (also termed GSK2126458) is a powerful inhibitor of phosphoinositide 3-kinases and mammalian target of rapamycin, and is being developed for curing solid tumors [75]. Jang et al. [40] found three combination therapies (omipalisib/remdesivir, tipifarnib/omipalisib, and tipifarnib/remdesivir) demonstrating strong synergistic effects in curbing SARS-CoV-2, through virtual screening of 6, 218 drugs and cell-based assay. Calpeptin exhibited high antiviral activity against SARS-CoV-2 without apparent cytotoxicity via blocking extracellular vesicles (EVs) biogenesis/release as a cysteine proteinase inhibitor. Interestingly, a cocktail of calpeptin and remdesivir significantly enhanced anti-SARS-CoV-2 activity in comparison with monotherapy [76]. Lenvatinib, as a broad-spectrum host receptor tyrosine kinase (RTK) inhibitor, showed no inhibitory activity against Mpro in vitro, but exhibited remarkable synergistic effect with remdesivir to suppress SARS-CoV-2 replication, albeit selectively in Vero-CCL81 cells. Moreover, time-of-addition experiment revealed that lenvatinib/remdesivir combination remedy probably targeted SARS-CoV-2 replication process at a post-entry step [77]. Androgen was reported to function as a transcriptional regulator of ACE2 and TMPRSS2 in mouse and human cells. Notably, the combination of camostat (a TMPRSS2 serine protease inhibitor) with anti-androgen enzalutamide or androgen receptor degrader ARD-69 was more efficacious in blocking the entry of pseudotype SARS-CoV-2 into the host cells than the single drug [78]. Zhang et al. [79] have shown that heparin/heparan sulfate (HS) binds directly to S protein and promotes the binding of viral particles carrying S protein to the cell surface to assist viral entry. Tilorone used to be a pan-antiviral agent and also prevented SARS-CoV-2 infection in vitro. Raloxifene, a heparin/HS-binding drug, in combination with tilorone, was found to be synergistic against cytopathic issue induced by virus infection. Furthermore, no notable cytotoxicity emerged in the combination regimen even at highest concentrations [79]. Since it was established that SARS-CoV-2 infection could induce the activation of tissue factor-mediated coagulation via activation of acid sphingomyelinase [91], and blockage of acid sphingomyelinase prevented uptake of SARS-CoV-2 by epithelial cells [92], targeting the acid sphingomyelinase is a promising strategy to combat COVID-19. Fluoxetine and fluvoxamine are well studied inhibitors of acid sphingomyelinase and show great therapeutic potential for SARS-CoV-2 infection [93, 94, 95]. Remarkably, fluoxetine, in combination with GS-441524 (a plasma metabolite of remdesivir), exerted synergistic antiviral effects against different SARS-CoV-2 variants in vitro [80]. Despite its small sample size, an open-label non-randomized clinical trial showed that HCQ (an alkalinizing lysosomatropic drug) treatment was extremely efficient for virus clearance in COVID-19 patients and it is synergic when combined with azithromycin [81]. Similarly, a retrospective analysis of larger scale samples (3119 samples, 83.5% of 3737) suggested that early treatment with at least 3 days of HCQ/azithromycin led to a better clinical status and a quicker virus eradication than other treatments [82]. However, Molina et al. [83] pointed out that the combination of HCQ and azithromycin brought no clinical benefit for the severe COVID-19 patients. Notably, treatment with azithromycin combined with HCQ caused a greater QT interval prolongation than treatment with either drug alone [84]. Since the addition of azithromycin doesn’t increase the activity of HCQ, the combination was no longer recommended as treatment or prophylaxis for COVID-19 by the WHO. Further, Kang et al. [85] reported that the in vitro antiviral activity of HCQ at concentrations with little concern of toxicity was minimal, no matter it was used alone or with lopinavir/ritonavir. A randomized, multicenter trial was conducted to probe whether Zinc supplements can enhance the clinical efficacy of HCQ. And no additive effect was observed when zinc supplements were added to HCQ as a combination therapy [86]. As to more combination therapies comprising chloroquine or HCQ and side-effects, mechanisms thereof, you may refer to another review [96].

Table 2.Combination therapies with antivirals acting on SARS-CoV-2 and host cell.
Drugs for Combination Targets or action mechanisms Results Reference
IMU-838 + remdesivir DHODH + RdRp Enhanced in vitro anti-SARS-CoV-2 activity [70]
B02 + remdesivir Human RAD51 + RdRp Antiviral synergy [71]
Emetine + remdesivir A protein synthesis inhibitor + RdRp 64.9% inhibition in viral yield [72]
Baricitinib + remdesivir Janus kinase + RdRp Reducing recovery time and accelerating improvement [73]
Baricitinib + corticosteroids Janus kinaser + anti-inflammation Strongly recommended by WHO https://www.who.int/teams/health-care-readiness/covid-19/therapeutics
Tipifarnib + Omipalisib (GSK2126458) Farnesyltransferase + phosphoinositide 3-kinases Strong synergistic effects [40, 74, 75]
Omipalisib + remdesivir; tipifarnib + remdesivir Phosphoinositide 3-kinases + RdRp; Farnesyltransferase + RdRp Strong synergistic effects [40]
Calpeptin + remdesivir Cysteine proteinase + RdRp Enhanced the anti-SARS-CoV-2 activity [76]
Lenvatinib + remdesivir Host RTK + RdRp Exhibited striking synergistic effect [77]
Camostat + enzalutamide or ARD-69 TMPRSS2 serine protease + anti-androgen or androgen receptor degrader More efficacious in blocking the entry [78]
Raloxifene + tilorone A heparin/HS-binding drug + a pan-antiviral agent Synergistic against SARS-CoV-2-induced cytopathic effect [79]
Fluoxetine + GS-441524 Acid sphingomyelinase + RdRp Synergistic antiviral effect [80]
HCQ + azithromycin An alkalinizing lysosomatropic drug + antibiotic (1) Synergic; (2) a better clinical status and a quicker virus eradication; (3) no clinical benefit for the treatment of the hospitalized patients with severe COVID-19; (4) a greater QT interval [81, 82, 83, 84]
HCQ + lopinavir + ritonavir An alkalinizing lysosomatropic drug + anti-HIV drug Minimal in vitro antiviral activity [85]
HCQ + zinc supplements An alkalinizing lysosomatropic drug + an essential micronutrient No additive effect [86]
MEDS433 + dipyridamole (DPY) DHODH + the pyrimidine salvage pathway Restored the anti-SARS-CoV-2 activity of MEDS433 [87]
Camostat mesylate + E-64d TMPRSS2 + endosomal cysteine proteases cathepsin B and L Complete inhibition of the SARS-CoV-2 cell entry [2, 88]
Camostat mesylate + HCQ TMPRSS2 + an alkalinizing lysosomatropic drug Relative clinical trials have been withdrawn or in an unknown status NCT04355052, NCT04338906

MEDS433 is a new inhibitor of the human dihydroorotate dehydrogenase (hDHODH), a key enzyme of the de novo pyrimidine biosynthesis pathway. The pyrimidine salvage pathway may attenuate the antiviral efficacy of an hDHODH inhibitor through transporting nucleosides from extracellular medium [97]. Thus, a combination strategy was adopted with MEDS433 and dipyridamole (DPY), the latter inhibiting the pyrimidine salvage pathway. And this combination strategy restored the lost anti-SARS-CoV-2 activity of MEDS433 in the presence of exogenous uridine [87]. Camostat mesylate is a clinically efficient serine protease inhibitor and active against TMPRSS2 [88]. Complete inhibition of the SARS-CoV-2 cell entry was realized when camostat mesylate and E-64d, an inhibitor of endosomal cysteine proteases cathepsin B and L (CatB/L), were added [2]. However, two clinical trials (NCT04355052, NCT04338906) exploring the combination remedy of TMPRSS2 inhibitor camostat mesylate and HCQ have been withdrawn or in an unknown status (Table 2).

4.1.3 Antiviral Combined with Pharmacokinetic Enhancer

PF-07321332 (Nirmatrelvir) is an oral antiviral drug developed by Pfizer. Ritonavir is able to slow the metabolism of PF-07321332 by cytochrome enzymes, therefore maintaining higher concentrations of the primary drug. Thus PF-07321332 and ritonavir was used as a combination medication in Phase III studies and was marketed as Paxlovid for the treatment of COVID-19. Now Paxlovid has been utilized in market across the world since it was able to reduce the risk of hospitalization or death by 89% compared with placebo [98] (Table 3, Ref. [98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112]). WHO has strongly recommended the use of nirmatrelvir/ritonavir in patients with non-severe illness at the highest risk of hospitalization, however provided conditional recommendation against the use of them in patients with non-severe illness at a low risk of hospitalization (published 22 April 2022) (https://www.who.int/teams/health-care-readiness/covid-19/therapeutics). Cobicistat is an FDA-approved drug that can boost the activity of major drug via blocking the activity of cytochrome P450-3As (CYP3As) and P-glycoprotein (P-gp). Recently, it was reported to inhibit SARS-CoV-2 replication through suppressing the fusion of the viral S-glycoprotein to the cell membrane. In combination with remdesivir, cobicistat exhibited a synergistic antiviral effect in vitro and decreased viral titers and disease progression in Syrian hamsters [99]. Darunavir, a protease inhibitor and its pharmacokinetic enhancer, cobicistat, work as a whole to treat HIV infection. A pilot study was conducted at Shanghai Public Health Clinical Center (SPHCC) to preliminarily evaluate the efficacy and safety of darunavir/cobicistat in treating COVID-19 pneumonia. There was no any tendency of improvement observed in the darunavir/cobicistat group in comparison with the control group, although it was well tolerated (clinicaltrials.gov: NCT04252274) (Table 3) [100].

Table 3.Combination therapies with antiviral plus pharmacokinetic enhancer or complication-treating drug.
Drugs for Combination Targets or action mechanisms Results Reference
PF-07321332 (Nirmatrelvir) + ritonavir Mpro + CYP3A Decrease the risk of hospitalization or death by 89% [98]
Cobicistat + remdesivir CYP3As + P-gp + S-glycoprotein + RdRp Decreased viral titers and disease progression [99]
Darunavir + cobicistat A protease inhibitor + CYP3As+ P-gp No any trend of improvement [100]
Quercetin + Vitamin C (VC) Anti-inflammatory + a broad spectrum antiviral agent Synergistic antiviral, antioxidant and immunomodulatory effects [101]
Plitidepsin + dexamethasone host cell’s eEF1A + anti-inflammation A phase III trial is underway [102]
Standard of care (SOC) + remdesivir + dexamethasone RdRp + anti-inflammation A reduction in 30-day mortality [103]
Methylprednisolone + remdesivir RdRp (1) Prevented body weight loss and inflammation; (2) dampened viral protein expression and viral load [104]
olfactory rehabilitation + palmitoylethanolamide + luteolin NA* Effective in improving recovery of olfactory function [105]
Cannabidiol (CBD) + terpenes An anti-inflammatory molecule + anti-microbials Demonstrated mild to moderate antivirus effect [106, 107]
Pentoxifylline + oxypurinol TNF-α production + xanthine oxidase Just a suggestion, no experimental data [108]
VD + DPP-4i Anti-inflammatory + immunomodulatory A perspective, no experimental data [109, 110, 111]
Dapsone + doxycycline Suppress production of various cytokines + anti-microbial Just a suggestion, no experimental data [112]
* NA, not available.
4.1.4 One Antiviral and Another Drug Treating Complications

In plants, quercetin is a flavonoid compound, produced from the phenylpropanoid pathway and ultimately derived from phenylalanine. There is a tremendous amount of literature supporting its anti-inflammatory and antiviral properties, especially against several respiratory viruses in both in vitro and in vivo experiments (Table 3) [101]. Vitamin C (VC) is a broad spectrum antiviral agent and an inhibitor of aerobic glycolysis. Treatment with quercetin in combination with VC provided synergistic antiviral, antioxidant and immunomodulatory effects due to overlapping antiviral and immunomodulatory properties and the capacity of VC to regenerate quercetin (Table 3) [101]. Plitidepsin is a cyclic depsipeptide known for its anti-tumor and antiviral activity, mainly acting on isoforms of the host cell’s eukaryotic-translation-elongation-factor-1-alpha (eEF1A). Through blocking eEF1A and therefore translation of essential viral proteins, it exhibits anti-SARS-CoV-2 potential. A phase III trial is underway to compare the plitidepsin/dexamethasone remedy with the standard of care in moderate hospitalized patients (ClinicalTrials.gov Identifier: NCT04784559) [102]. A comparative study of the effectiveness of remdesivir/dexamethasone plus standard of care (SOC) versus SOC alone was proceeded in a clinical trial, and the result showed a reduction in 30-day mortality with the combination treatment [103]. In the hamster model of SARS-CoV-2 infection, treatment with methylprednisolone suppressed viral induction of proinflammatory cytokines but enhanced RNA replication of SARS-CoV-2. Although weight reduction, along with nasal and pulmonary inflammation, was relieved with methylprednisolone monotherapy, both viral loads enhancement and antibody response weakening also accompanied. On the contrary, a combination therapy methylprednisolone/remdesivir not only restrained weight reduction and inflammation, but also dampened viral protein expression and viral loads. Furthermore, the suppression of methylprednisolone on antibody response was also attenuated in this combination therapy [104]. Approximately 30% of COVID-19 patients were reported to have obstinate smell or taste dysfunction as prolonged sequalae of infection. Treatment combining olfactory rehabilitation with oral supplementation with palmitoylethanolamide and luteolin was effective in improving recovery of olfactory function, especially in those patients with longstanding olfactory dysfunction [105]. Cannabidiol (CBD) is widely available as medicinal compounds with various applications, most involved in modulating the inflammation processes [106]. Santos et al. [107] have evaluated the anti-infection effect of the combination of CBD with terpenes, as an anti-inflammatory chemical and anti-microbial, respectively. The virucide effectiveness of CBD and terpene-based six formulations were tested in different cell lines and the result demonstrated mild to moderate antivirus effect [107].

Pentoxifylline is an inhibitor of TNF-α production while oxypurinol an inhibitor of xanthine oxidase. Accordingly, pentoxifylline alone, or combined with oxypurinol, was reported to reduce the systemic inflammation caused by experimentally-induced pancreatitis in rats. Therefore, pentoxifylline in combination with oxypurinol was suggested as an early remedy for COVID-19 patients to prevent the fatal acute respiratory distress syndrome (ARDS) [108]. Pinheiro et al. [111] published a perspective article discussing the synergistic effect of joint application of vitamin D (VD) and dipeptidyl peptidase-4 inhibitors (DPP-4i). After analysis of the relative progress of biological activity of VD and DPP-4i and pathologic profile of COVID-19, they proposed that co-administration of VD and DPP-4i might exert anti-inflammatory and immunomodulatory activity to a greater extent than VD or DPP-4i alone. Besides, VD and DPP-4i might bring about beneficial effect on endothelial dysfunction [109, 110], which was implicated in COVID-19 pathophysiology [113]. Overall, the authors provided us the plausibility for the combination therapy VD/DPP-4i as an immunomodulation strategy to dampen the virulence of SARS-CoV-2, inhibit disease deterioration and modulate the cytokine storm in COVID-19 [111]. Dapsone, belonging to a class of sulfone drugs, suppresses production of various cytokines including interleukin (IL)1α, IL8, IL1β, IL6, and IL8 and tumor necrosis factor-α [114]. Thus, it was suggested to combine with doxycycline to treat severe COVID-19 patients (Table 3) [112].

4.1.5 Multiple Drugs for Combination Use

The pathophysiology of SARS-Cov-2 relates to inflammation, immune dysregulation, coagulopathy, and endothelial dysfunction. No single therapeutic agent can manage all these pathophysiologic conditions. Hence, a randomized open-label trial was initiated to investigate a triple combination remedy (aspirin, atorvastatin, and nicorandil) with anti-inflammatory, antithrombotic, immunomodulatory, and vasodilator properties against COVID-19 in India [115] (Table 4, Ref. [115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132]). Procter et al. [116, 117] have evaluated the effects of multidrug combination therapy on high-risk patients. At least two anti-SARS-CoV-2 agents (zinc, HCQ, ivermectin) and one antibiotic (azithromycin, doxycycline, ceftriaxone) were used as well as inhaled budesonide and/or intramuscular dexamethasone. And it was concluded that early ambulatory multidrug therapy was associated with low rates of hospitalization and death (Table 4) [116, 117]. The outcome of a triple therapy (zinc, azithromycin, and HCQ) was evaluated in COVID-19 patients. No adverse cardiac events and notably fewer hospitalizations were observed [118].

Table 4.Combination therapies with multiple drugs or some specific antimicrobials.
Drugs for Combination Targets or action mechanisms Results Reference
Aspirin + atorvastatin + nicorandil Anti-inflammatory, antithrombotic, immunomodulatory and vasodilator properties NA* [115]
Two antivirals of (zinc, HCQ, ivermectin) + one antibiotic of (azithromycin, doxycycline, ceftriaxone) + budesonide + dexamethasone Multiple targets Low rates of hospitalization and death [116, 117]
Zinc + azithromycin + HCQ Multiple targets No adverse cardiac events and notably fewer hospitalizations [118]
Umifenovir (arbidol) + LPV/r Inhibition of membrane fusion + anti-HIV Viral load vanished in 94% patients of the combination group, compared to 52.9% of the monotherapy group (LPV/r) [119, 120]
Shufeng Jiedu Capsules + umifenovir Multiple targets The more rapid subsidence of a fever and better resolution of pneumonia symptoms [121]
HCQ + VC + VD + Zinc An alkalinizing lysosomatropic drug + broad-spectrum antiviral Ongoing NCT04335084
Clofazimine + remdesivir An anti-tuberculosis drug + RdRp Synergistic antiviral activity [122]
Mefloquine + artesunate, artesunate + amodiaquine, artemether + lumefantrine, artesunate + pyronaridine, or dihydroartemisinin + piperaquine Antimalarial drug Mefloquine/artesunate demonstrated the strongest antiviral activity with % inhibition of 72.1 ± 18.3%, others 27.1 to 34.1% [123]
Mefloquine + Nelfinavir Blocking viral entry + Mpro inhibitor Synergistic antiviral activity [124]
Nitazoxanide + favipiravir A commercial antiprotozoal agent + RdRp Underway NCT04918927
ATV+ RTV An HIV-1 protease inhibitor + Cytochrome P450 3A More potent [125]
LPV/r+azithromycin Anti-HIV + antibiotic The most effective combination group among 8 groups of combination drugs [126]
LPV/r Anti-HIV drug No significant benefit and more gastrointestinal adverse events [127]
DOXY + HCQ Antibiotic + an alkalinizing lysosomatropic drug Reduction in recovery time and mortality and lower rate of transfer to hospital [128]
DOXY+ ivermectin Antibiotic Recovered earlier [129]
DOXY+ VC Antibiotic + broad-spectrum antiviral Suggestion [130]
Minocycline + HCQ Antibiotic + an alkalinizing lysosomatropic drug Just an appeal for further clinical studies [131]
Itraconazole + remdesivir Antifungals + RdRp Synergistically prohibited the production of SARS‐CoV‐2 particles in vitro [132]
* NA, not available.

Umifenovir (also termed as arbidol), an antiviral agent with broad spectrum, functions primarily through inhibition of membrane fusion between the viral envelope and host cell membrane, therefore, suppressing viral entry and infection [119]. Deng et al. [120] conducted a retrospective cohort study to compare arbidol and lopinavir/ritonavir (LPV/r) combination treatment for COVID-19 patients with LPV/r alone. They found that viral load vanished after 14 days in 94% patients of the combination group, compared to 52.9% of the monotherapy group (LPV/r) [120]. Recently, another retrospective cohort study was carried out to understand the clinical effectiveness and safety of Shufeng Jiedu Capsules (a Chinese herbal compound composed of eight medicinal plants [133]) in combination with umifenovir (Arbidol) for common-type COVID-19. The subsidence of a fever was observed more rapidly and the chest CT scan also showed better resolution of pneumonia symptoms in the combination treatment group than that in the control group (treated with arbidol hydrochloride capsules alone) [121]. A Phase II interventional study testing whether treatment with HCQ, Vitamin C, Vitamin D, and Zinc can prevent symptoms of COVID-19 is ongoing (NCT04335084).

4.1.6 Other Antimicrobials for Combination Use against SARS-CoV-2

Clofazimine was discovered as an anti-tuberculosis drug and later used for the treatment of leprosy [134]. Clofazimine, in combination with remdesivir, exhibited synergistic antiviral activity in vitro and in vivo, and restricted viral shedding from the upper respiratory tract (Table 4) [122]. The antiviral activity of several antimalarial artemisinin-based combination therapies (ACT), including mefloquine/artesunate, artesunate/amodiaquine, artemether/lumefantrine, artesunate/pyronaridine, or dihydroartemisinin/piperaquine, was tested in vitro against a SARS-CoV-2 strain (IHUMI-3) in Vero E6 cells. Mefloquine/artesunate demonstrated the strongest antiviral activity with % inhibition of 72.1 ± 18.3% at the relative concentration in malaria treatment. However all the other combinations showed anti-SARS-CoV-2 activity with % inhibition in the same ranges (27.1 to 34.1%) [123]. Mefloquine, an anti-malarial drug, exhibited stronger anti-SARS-CoV-2 activity than HCQ in VeroE6/TMPRSS2 and Calu-3 cells, through blocking viral entry after attachment to the host cell. Joint treatment with Mefloquine and Nelfinavir manifested synergistic antiviral activity in wide concentration ranges [124]. Itraconazole is a member of the triazole group of broad-spectrum antifungals [135]. The in vitro antiviral activities of itraconazole and its metabolite against SARS-CoV-2 were proved in low micromolar level [136]. Furthermore, itraconazole/remdesivir combination prohibited the production of SARS-CoV-2 particles >90% in a synergistic fashion [132]. However, the antiviral effect of itraconazole was lost in vivo [137]. Nitazoxanide, a commercial antiprotozoal agent with broad-spectrum antiviral activity against various viruses including human and animal coronaviruses, was reported to inhibit the SARS-CoV-2 at a low-micromolar concentration in vitro [138]. A proof-of-principle placebo-controlled clinical trial is underway to investigate the effect of early antiviral treatment with nitazoxanide plus or minus favipiravir (RdRp inhibitor) in preventing progression to the later phase of the disease (ClinicalTrials.gov Identifier: NCT04918927). Atazanavir (ATV) is an HIV-1 protease inhibitor currently suggested as a first-line treatment for naive HIV-infected patients. ATV in combination with ritonavir (RTV) was more potent than ATV alone in different cell assays against SARS-CoV-2 replication [125]. Lopinavir/ritonavir (LPV/r) is a combinational antiviral drug commonly used in the treatment of HIV, the causative agent for acquired immunodeficiency syndrome (AIDS) [139]. More and more evidence proved that lopinavir/ritonavir could be considered an efficient redemy for CoVs-induced infections [140]. Purwati et al. [126] have evaluated 8 groups of combination drugs for the anti-SARS-CoV-2 activity in vitro, and found LPV/r/azithromycin is the most effective combination group among them. However, a trial of LPV/r treatment in severe COVID-19 adult patients showed that LPV/r didn’t bring about significant benefit [127]. So was it in another similar randomized trial [141]. Besides, there were more gastrointestinal adverse events including nausea, vomiting, and diarrhea in the LPV/r group than in the standard-care group [127]. Apart from the gastrointestinal side effects, LPV/r use can also be associated with skin rash, hepatitis, metabolic derangements (hypercholesterolemia, hyperglycemia), neutropenia, thrombocytopenia, and QT prolongation [142]. Consequently, due to lack of efficacy, WHO withdrew LPV/r from their solidarity trial and provided strong recommendation against LPV/r in COVID-19 patients of any severity (published 17 December 2020) (https://www.who.int/teams/health-care-readiness/covid-19/therapeutics).

Doxycycline (DOXY) is a semisynthetic, second-generation class of tetracycline with a wide spectrum of antimicrobial activity. The activity of DOXY/HCQ combination therapy was studied in a series of fifty-four high-risk COVID-19 patients. And the clinical experience of this case series indicated a reduction in recovery time and death rate, and lower rate of transfer to hospital after treatment with DOXY/HCQ [128]. A double-blind and randomized interventional trial of a combination of DOXY and ivermectin was carried out with 400 participants. Patients with mild-to-moderate COVID-19 infection treated with ivermectin plus DOXY recovered earlier than those with placebo, were less possible to deteriorate, and were more inclined to be SARS-CoV-2 negative by RT-PCR at the end of the treatment (NCT04523831) [129]. It is suggested that co-administration of doxycycline and vitamin C shows more benefit against COVID-19 [130]. Minocycline is another semisynthetic, second-generation derivative of tetracycline with an activity against lots of microorganisms [143]. It also inhibits many proinflammatory cytokines, which are common in severe and complicated COVID-19 cases [144]. Gautam et al. [131] analyzed the pros and cons of the cocktail HCQ/minocycline in treating moderate to severe COVID-19 patients and called upon public and private healthcare bodies to implement large well-designed clinical studies for generating more convincing suggestions.

4.2 Combination Therapy with Biomacromolecule and Small Molecule

Interferons (IFNs) are glycoproteins with potential immunomodulatory and hormone-like functions [145]. Both IFNα and IFNβ have been considered as a potential therapy against COVID-19, especially combined with ribavirin [146]. In a non-controlled trial, the combination use of IFN-β-1a with HCQ and LPV/r in the management of COVID-19 offered positive results including virus eradication rates, fever subsidence, recovery time and safety characteristics [147] (Table 5, Ref. [147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168]). Retrospective data also revealed oxygenation increase, survival advantage and discharging of sever COVID-19 inpatients by the combination therapy of IFN-β-1a with LPV/r (Table 5) [148]. However, in a phase III trial, the combination regimen of LPV/r and IFN-β-1a demonstrated neither clinical improvement at day 15 nor reduction of SARS-CoV-2 load in respiratory specimens [149]. Aerosol IFN-α2b alone or in combination with arbidol exhibited superior performance in decreasing inflammatory markers in the blood of COVID-19 patients and accelerating viral clearance compared to arbidol treatment alone [150]. As for subcutaneous administration of IFN-α2b, an observational study of its use in combination with LPV/r showed a decline in the length of hospital stay and acceleration of viral eradication in COVID-19 patients [151]. An open-label, randomised, phase Ⅱ trial was conducted to assess the efficacy and safety of triple combination of interferon beta-1b, LPV/r, and ribavirin in the treatment of hospitalized COVID-19 patients. And the outcome manifested the triple antiviral therapy was safe and superior to LPV/r alone in shortening virus shedding, relieving symptoms, and facilitating discharge of patients with mild to moderate COVID-19 (NCT04276688) [152]. It was known that IFNα-induced serpin E1 is a risk factor for thrombosis. Thus, a combination therapy was designed with IFNα and nafamostat, for the anticoagulative properties of nafamostat could compensate for the adverse effects of thrombosis by IFNα administration. And the combination suppressed SARS-CoV-2 infection in an additive fasion by cooperatively targeting host TMPRSS2 [153]. Besides, combinations of IFN-α2a with known SARS-CoV-2 inhibitors remdesivir, EIDD-2801, camostat or cycloheximide, showed a strong synergy in inhibiting SARS-CoV-2 infection [154].

Table 5.Combination therapy with biomacromolecular and small molecular.
Drugs for Combination Targets or action mechanisms Results Reference
IFN-β-1a + HCQ + LPV/r Immunomodulatory + antiviral Viral eradication rates, fever subsidence, recovery time and safety characteristics were improved [147]
IFN-β-1a + LPV/r Immunomodulatory + antiviral Oxygenation increase, survival advantage and discharging of sever COVID-19 [148]
IFN-β-1a + LPV/r Immunomodulatory + antiviral Neither clinical improvement nor reduction of SARS-CoV-2 load [149]
IFN-α2b + arbidol Immunomodulatory + antiviral Exhibited superior performance [150]
IFN-α2b + LPV/r Immunomodulatory + anti-HIV drug A decline in the hospital time and acceleration of viral eradication [151]
IFN-β-1b + LPV/r + ribavirin Immunomodulatory + antivirals Safe and superior to LPV/r alone [152]
IFNα + nafamostat Immunomodulatory + anticoagulant Suppressed SARS-CoV-2 infection in an additive fasion [153]
IFN-α2a + remdesivir; IFN-α2a + EIDD-2801; IFN-α2a + camostat; or IFN-α2a + cycloheximide Immunomodulatory + SARS-CoV-2 inhibitors A strong synergy in inhibiting SARS-CoV-2 infection [154]
Remdesivir + tocilizumab + dexamethasone Antiviral (small molecular and antibody) + anti-inflammation Timely resolution of the cytokine storm and subsequent improvement in ARDS symptoms and eventual recovery [155]
Remdesivir + convalescent plasma or mAbs Antiviral (small molecular and antibody) High viral clearance [156, 157, 158, 159, 160]
Ruxolitinib + eculizumab A Janus kinase (JAK) 1/2 inhibitor + an anti-C5a complement monoclonal antibody Significant improvements in respiratory symptoms and radiographic pulmonary lesions and reduction of circulating D-dimer [161]
Favilavir + tocilizumab Antiviral + an anti–interleukin-6 monoclonal antibody Its status has not been updated for 2 years NCT04310228
Immunoglobulin + steroid pulses Immunoglobulin + anti-inflammation Useful in single-kidney transplanted patient with COVID-19 [162]
Sarilumab + standard of care (including corticosteroids) IL6 alfa-receptor antibody + anti-inflammation Not more effective [163]
Dexamethasone + anti- SARS-CoV-2 mAbs Anti-inflammatory + antiviral antibody Synergistic [164]
GRFT + EK1 An antiviral + spike S2 subunit Strong synergistic effect [165]
Bromelain + acetylcysteine Virus glycoproteins Synergistically weakened the infectivity [166, 167]
Bromelain + curcumin Prevent entry of SARS-CoV-2 into cells and interfere with viral replication A proposal [168]

COVID-19 may developed as a chronic disease in patients with some types of immunodeficiency. In this condition, remdesivir monotherapy is frequently ineffective, but the combination of remdesivir with antibody-based therapeutics holds promise. Remdesivir, combined with tocilizumab (an anti–interleukin-6 monoclonal antibody) and dexamethasone, was administered at the early stage of the disease, resulting in timely resolution of the cytokine storm and subsequent improvement in ARDS symptoms and eventual recovery [155]. Combination of remdesivir with convalescent plasma or anti-SARS-CoV-2 monoclonal antibodies (mAbs) achieved high viral clearance [156, 157]. Successful outcomes with this combination therapy have also been demonstrated in other similar patients, such as with B-cell depleted [158], chronic lymphocytic leukemia [159] and X-linked agammaglobulinemia [160]. Considering that ruxolitinib, a Janus kinase (JAK) 1/2 inhibitor and eculizumab, an anti-C5a complement monoclonal antibody, function by acting on different but correlative pathological pathways, a combination therapy containing both of them was conceived to test its effect on SARS-CoV-2-related ARDS. And the results showed that patients treated with the combination obtained significant improvement in respiratory symptoms and radiographic pulmonary lesions and reduction in circulating D-dimer concentrations compared to the best available therapy group. Since the number of participants was small, only 7 in this study, the authors suggested further clinical studies with larger populations [161]. NCT04310228 is a clinical trial assessing the curative effect and security of favilavir in combination with tocilizumab. But its status has not been updated for 2 years. It was proved useful that high-dose intravenous immunoglobulin plus steroid pulses in treating a case of COVID-19 pneumonia patient with a single-kidney transplanted requiring mechanical ventilation and hemodialysis [162]. Sarilumab is a recombinant human immunoglobulin monoclonal antibody of IL6 alfa-receptor. In hospitalized patients with COVID-19 pneumonia, an early treatment with sarilumab combined with standard of care (including corticosteroids) was not more efficatious than current standard of care alone [163]. We have known that from previous studies glucocorticoid treatment brought beneficial anti-inflammatory effects, with virus replication being rather strengthened. This outcome inspired us to apply glucocorticoid together with virus-neutralizing mAbs. Based on histopathology and bulk and single-cell transcriptomic analysis, Wyler et al. [164] demonstrated the useful therapeutic effects of them and their potential as synergistic combination therapy in hamster models of moderate and severe COVID-19.

griffithsin (GRFT), a lectin isolated from the red alga Griffithsia sp, can recognize mannose with high specifity and has a broad-spectrum antiviral activity [169]. Combining GRFT and EK1, a pan-CoV fusion inhibitor targeting SARS-CoV-2 spike S2 subunit, exhibited strong synergistic effect against pseudotyped and live SARS-CoV-2 infection [165].

Bromelain exists mainly in the stem of the pineapple plant (Ananas comosus) and contains a number of enzymes that enable it to hydrolyze glycosidic bonds. Experimental studies have shown that bromelain exhibits unique immunomodulatory activity through various pathways [168]. Acetylcysteine is a powerful antioxidant and able to reduce disulfide bonds. Bromelain and acetylcysteine combination regimen (BromAc) showed synergistic action against glycoproteins by breakage of glycosidic linkages and disulfide bonds. Thus the combination therapy, BromAc, synergistically weakened the infectivity of SARS-CoV-2 cultured on Vero cells [166]. Further study indicated strong mucolytic and anti-inflammatory effect of BromAc ex vivo in tracheal aspirates from severe patients with COVID-19 [167]. Curcumin (diferuloylmethane) is a natural phenol found in turmeric (Curcuma longa), a member of the ginger family of plants [170]. Interestingly, curcumin has been shown in silico studies to prevent entry of SARS-CoV-2 into cells and interfere with viral replication, while bromelain may also prohibit viral entry demonstrated by a recent experimental study [171]. Notably, bromelain can markedly increase the absorption of curcumin, thereby compensating for its poor absorption drawback. Based on the analysis, Kritis et al. [168] reported to highlight the potential value of the synergistic effect of bromelain and curcumin in the prevention of severe COVID-19.

5. Discussion

The outbreak of COVID-19 has caused devastating economic and medical burden worldwide and seriously affects the living styles of most people. It has been more than two years since the first emergence of SARS-CoV-2 infected cases in Wuhan, China, during which time many scientists are trying to develop effective vaccines and therapies including antibodies and oral small molecules, at an unprecedented scale and pace. And fortunately, there have been several vaccines, antibodies and oral small molecules prove effective as prophylaxis or remedy in combating COVID-19. However, since its outbreak, SARS-CoV-2 has developed a variety of mutations, and different mutants have been identified in all four structural proteins and other viral proteins [172]. Mutations in the SARS-CoV-2 RBD or N-terminal domain (NTD) may endow these strains with enhanced replication and/or transmission ability, which lead to escape from antibody recognition and attenuated neutralizing activity of mAbs [173]. Besides, some new SARS-CoV-2 variants can also infect those people who have been vaccinated once or more. In theory, combination therapy may have additive or synergistic activity in preventing infection by escape mutants compared to monotherapy, thus is highly emphasized. However, the present studies on combination therapy against SARS-CoV-2 mutant strains focus on mAbs, the small molecule drugs scarcely being involved.

In a combination therapy, one drug may endow another more power to counter the disease. Ideally, the pharmacokinetic profiles as well as the pharmacodynamics characteristics of individual drug are improved when combined used. As aforementioned above, effective concentration of each drug would be reduced below the maximal plasma concentration, which therefore attenuates toxicity resulted by high concentration. However, in many cases, there were limited data regarding the pharmacokinetic profiles of individual drug in the combination therapies. We call for more attention on the drug interaction and the resulting pharmacokinetic profiles in the future research.

The virus invades the organ systems across the body, which triggers a variety of complications (concomitant symptoms), ranging from fever to multiple organ dysfunction syndrome, etc. Sometimes, the severe complications are fatal, needing to be tackled as a matter of urgency. Thus it is not enough for a combination therapy to counter just the disease-causing agent. An effective treatment strategy for COVID-19 should take comprehensive consideration of both the host symptom and the pathogenic microorganism. So we are a bit more optimistic about the therapeutic effect of those combination therapies targeting both the virus life cycle and host complications.

This review updates the progress of dual and multiple drug combination therapies (containing at least one small molecule drug) against COVID-19. The antiviral mechanism of each combination, especially the target of each component, and the outcome is highlighted. We can see that these combinations target the same viral enzyme with different action mechanisms, different viral enzymes playing key roles in virus life cycle, different enzymes from virus and host cell facilitating virus survival, or host complication-relating pathways. Also we find that some combination therapies were proposed as medical hypotheses or perspective based on previous knowledge about the pharmacology of individual drug, most of which deserve further exploration by experiment. Others were validated by clinical trials, animal experiments, cell or enzyme assays. Many combination therapies exhibited positive (additive or synergistic) outcome awaiting further efficiency clarification in SARS-CoV-2-relating animal model or clinical trial. Though some combinations turned out to be no effect, even toxic, they are still of guiding value in clinical practice, especially for those combinations of repurposing drugs. In summary, this review is hopeful for facilitating us to select a proper anti-SARS-CoV-2 combination therapy for further research or clinical treatment.

Author Contributions

QL conceived the topic and wrote the manuscript. YZ assisted in writing and organizing the draft. JZ supervised the work and approved the final draft. All authors read and approved the final draft.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

JZ was supported by the Thousand Young Talents Program of China, the National Natural Science Foundation of China (Grant No. 31770795; Grant No. 81974514), and the Jiangxi Province Natural Science Foundation (Grant No. 20181ACB20014).

Conflict of Interest

The authors declare no conflict of interest.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References
[1]
Poduri R, Joshi G, Jagadeesh G. Drugs targeting various stages of the SARS-CoV-2 life cycle: Exploring promising drugs for the treatment of Covid-19. Cellular Signalling. 2020; 74: 109721.
[2]
Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020; 181: 271–280.e8.
[3]
Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020; 367: 1444–1448.
[4]
Brielle ES, Schneidman-Duhovny D, Linial M. The SARS-CoV-2 Exerts a Distinctive Strategy for Interacting with the ACE2 Human Receptor. Viruses. 2020; 12: 497.
[5]
Belouzard S, Chu VC, Whittaker GR. Activation of the SARS coronavirus spike protein via sequential proteolytic cleavage at two distinct sites. Proceedings of the National Academy of Sciences. 2009; 106: 5871–5876.
[6]
Millet JK, Whittaker GR. Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein. Proceedings of the National Academy of Sciences. 2014; 111: 15214–15219.
[7]
Park J, Li K, Barlan A, Fehr AR, Perlman S, McCray PB, et al. Proteolytic processing of Middle East respiratory syndrome coronavirus spikes expands virus tropism. Proceedings of the National Academy of Sciences. 2016; 113: 12262–12267.
[8]
Gierer S, Bertram S, Kaup F, Wrensch F, Heurich A, Krämer-Kühl A, et al. The spike protein of the emerging betacoronavirus EMC uses a novel coronavirus receptor for entry, can be activated by TMPRSS2, and is targeted by neutralizing antibodies. Journal of Virology. 2013; 87: 5502–5511.
[9]
Coutard B, Valle C, de Lamballerie X, Canard B, Seidah NG, Decroly E. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antiviral Research. 2020; 176: 104742.
[10]
Essalmani R, Jain J, Susan-Resiga D, Andréo U, Evagelidis A, Derbali RM, et al. Distinctive Roles of Furin and TMPRSS2 in SARS-CoV-2 Infectivity. Journal of Virology. 2022; 96: e0012822.
[11]
Liu T, Luo S, Libby P, Shi GP. Cathepsin L-selective inhibitors: A potentially promising treatment for COVID-19 patients. Pharmacology & Therapeutics. 2020; 213: 107587.
[12]
Lempp FA, Soriaga LB, Montiel-Ruiz M, Benigni F, Noack J, Park Y, et al. Lectins enhance SARS-CoV-2 infection and influence neutralizing antibodies. Nature. 2021; 598: 342–347.
[13]
Amraei R, Yin W, Napoleon MA, Suder EL, Berrigan J, Zhao Q, et al. CD209L/L-SIGN and CD209/DC-SIGN Act as Receptors for SARS-CoV-2. ACS Central Science. 2021; 7: 1156–1165.
[14]
Cantuti-Castelvetri L, Ojha R, Pedro LD, Djannatian M, Franz J, Kuivanen S, et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science. 2020; 370: 856–860.
[15]
Kim D, Lee J, Yang J, Kim JW, Kim VN, Chang H. The Architecture of SARS-CoV-2 Transcriptome. Cell. 2020; 181: 914–921.e10.
[16]
Redondo N, Zaldívar-López S, Garrido JJ, Montoya M. SARS-CoV-2 Accessory Proteins in Viral Pathogenesis: Knowns and Unknowns. Frontiers in Immunology. 2021; 12: 708264.
[17]
Zhang L, Lin D, Sun X, Curth U, Drosten C, Sauerhering L, et al. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors. Science. 2020; 368: 409–412.
[18]
Gao Y, Yan L, Huang Y, Liu F, Zhao Y, Cao L, et al. Structure of the RNA-dependent RNA polymerase from COVID-19 virus. Science. 2020; 368: 779–782.
[19]
Al-Horani RA, Kar S, Aliter KF. Potential Anti-COVID-19 Therapeutics that Block the Early Stage of the Viral Life Cycle: Structures, Mechanisms, and Clinical Trials. International Journal of Molecular Sciences. 2020; 21: 5224.
[20]
Malik YA. Properties of Coronavirus and SARS-CoV-2. The Malaysian Journal of Pathology. 2020; 42: 3–11.
[21]
Wassie GT, Azene AG, Bantie GM, Dessie G, Aragaw AM. Incubation Period of Severe Acute Respiratory Syndrome Novel Coronavirus 2 that Causes Coronavirus Disease 2019: a Systematic Review and Meta-Analysis. Current Therapeutic Research. 2020; 93: 100607.
[22]
Zhou M, Zhang X, Qu J. Coronavirus disease 2019 (COVID-19): a clinical update. Frontiers of Medicine. 2020; 14: 126–135.
[23]
Azhar EI, Hui DSC, Memish ZA, Drosten C, Zumla A. The Middle East Respiratory Syndrome (MERS). Infectious Disease Clinics of North America. 2019; 33: 891–905.
[24]
Wu F, Zhao S, Yu B, Chen Y, Wang W, Song Z, et al. A new coronavirus associated with human respiratory disease in China. Nature. 2020; 579: 265–269.
[25]
Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. The Lancet. 2020; 395: 497–506.
[26]
Du Y, Tu L, Zhu P, Mu M, Wang R, Yang P, et al. Clinical Features of 85 Fatal Cases of COVID-19 from Wuhan. a Retrospective Observational Study. American Journal of Respiratory and Critical Care Medicine. 2020; 201: 1372–1379.
[27]
Guan W, Ni Z, Hu Y, Liang W, Ou C, He J, et al. Clinical Characteristics of Coronavirus Disease 2019 in China. New England Journal of Medicine. 2020; 382: 1708–1720.
[28]
Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical Characteristics of 138 Hospitalized Patients with 2019 Novel Coronavirus–Infected Pneumonia in Wuhan, China. Journal of the American Medical Association. 2020; 323: 1061–1069.
[29]
Hikmet F, Méar L, Edvinsson Å, Micke P, Uhlén M, Lindskog C. The protein expression profile of ACE2 in human tissues. Molecular Systems Biology. 2020; 16: e9610.
[30]
Faqihi F, Alharthy A, Alodat M, Kutsogiannis DJ, Brindley PG, Karakitsos D. Therapeutic plasma exchange in adult critically ill patients with life-threatening SARS-CoV-2 disease: a pilot study. Journal of Critical Care. 2020; 60: 328–333.
[31]
Alharthy A, Faqihi F, Memish ZA, Karakitsos D. Lung Injury in COVID-19—an Emerging Hypothesis. ACS Chemical Neuroscience. 2020; 11: 2156–2158.
[32]
Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, et al. Baseline Characteristics and Outcomes of 1591 Patients Infected with SARS-CoV-2 Admitted to ICUs of the Lombardy Region, Italy. Journal of the American Medical Association. 2020; 323: 1574–1581.
[33]
Waqar W, Ismail S, Jamil Z, Al-Shehhi A, Imran M, Hetta HF, et al. SARS-CoV-2 associated pathogenesis, immune dysfunction and involvement of host factors: a comprehensive review. European Review for Medical and Pharmacological Sciences. 2021; 25: 7526–7542.
[34]
Jiang F, Deng L, Zhang L, Cai Y, Cheung CW, Xia Z. Review of the Clinical Characteristics of Coronavirus Disease 2019 (COVID-19). Journal of General Internal Medicine. 2020; 35: 1545–1549.
[35]
Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Medicine. 2020; 46: 846–848.
[36]
Baig AM, Sanders EC. Potential neuroinvasive pathways of SARS‐CoV‐2: Deciphering the spectrum of neurological deficit seen in coronavirus disease‐2019 (COVID‐19). Journal of Medical Virology. 2020; 92: 1845–1857.
[37]
Li Y, Bai W, Hashikawa T. The neuroinvasive potential of SARS‐CoV2 may play a role in the respiratory failure of COVID‐19 patients. Journal of Medical Virology. 2020; 92: 552–555.
[38]
Pan L, Mu M, Yang P, Sun Y, Wang R, Yan J, et al. Clinical Characteristics of COVID-19 Patients with Digestive Symptoms in Hubei, China: a Descriptive, Cross-Sectional, Multicenter Study. American Journal of Gastroenterology. 2020; 115: 766–773.
[39]
Edwards K, Hussain I. Two Cases of Severe Autoimmune Thyrotoxicosis Following SARS-CoV-2 Infection. Journal of Investigative Medicine High Impact Case Reports. 2021; 9: 232470962110564.
[40]
Jang WD, Jeon S, Kim S, Lee SY. Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assay. Proceedings of the National Academy of Sciences. 2021; 118: e2024302118.
[41]
Bocci G, Bradfute SB, Ye C, Garcia MJ, Parvathareddy J, Reichard W, Surendranathan S, Bansal S, Bologa CG, Perkins DJ, Jonsson CB, Sklar LA, Oprea TI. Virtual and In Vitro Antiviral Screening Revive Therapeutic Drugs for COVID-19. ACS Pharmacology & Translational Science. 2020; 3: 1278–1292.
[42]
McCullough PA, Alexander PE, Armstrong R, Arvinte C, Bain AF, Bartlett RP, et al. Multifaceted highly targeted sequential multidrug treatment of early ambulatory high-risk SARS-CoV-2 infection (COVID-19). Reviews in Cardiovascular Medicine. 2020; 21: 517–530.
[43]
Al-Hajeri H, Baroun F, Abutiban F, Al-Mutairi M, Ali Y, Alawadhi A, et al. Therapeutic role of immunomodulators during the COVID-19 pandemic– a narrative review. Postgraduate Medicine. 2022; 134: 160–179.
[44]
Saha A, Sharma AR, Bhattacharya M, Sharma G, Lee S, Chakraborty C. Probable Molecular Mechanism of Remdesivir for the Treatment of COVID-19: need to Know more. Archives of Medical Research. 2020; 51: 585–586.
[45]
Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, et al. Remdesivir for the Treatment of Covid-19 — Final Report. New England Journal of Medicine. 2020; 383: 1813–1826.
[46]
Lamb YN. Remdesivir: first Approval. Drugs. 2020; 80: 1355–1363.
[47]
Wilt TJ, Kaka AS, MacDonald R, Greer N, Obley A, Duan-Porter W. Remdesivir for Adults with COVID-19: A Living Systematic Review for American College of Physicians Practice Points. Annals of Internal Medicine. 2021; 174: 209–220.
[48]
Rahimi MM, Jahantabi E, Lotfi B, Forouzesh M, Valizadeh R, Farshid S. Renal and liver injury following the treatment of COVID-19 by remdesivir. Journal of Nephropathology. 2021; 10: e10.
[49]
Gérard AO, Laurain A, Fresse A, Parassol N, Muzzone M, Rocher F, et al. Remdesivir and Acute Renal Failure: a Potential Safety Signal from Disproportionality Analysis of the who Safety Database. Clinical Pharmacology & Therapeutics. 2021; 109: 1021–1024.
[50]
WHO Solidarity Trial Consortium, Pan H, Peto R, Henao-Restrepo AM, Preziosi MP, Sathiyamoorthy V, et al. Repurposed Antiviral Drugs for Covid-19 - Interim WHO Solidarity Trial Results. The New England Journal of Medicine. 2021; 384: 497–511.
[51]
Narayanan A, Narwal M, Majowicz SA, Varricchio C, Toner SA, Ballatore C, et al. Identification of SARS-CoV-2 inhibitors targeting Mpro and PLpro using in-cell-protease assay. Communications Biology. 2022; 5: 169.
[52]
Fu L, Ye F, Feng Y, Yu F, Wang Q, Wu Y, et al. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nature Communications. 2020; 11: 4417.
[53]
Li P, Wang Y, Lavrijsen M, Lamers MM, de Vries AC, Rottier RJ, et al. SARS-CoV-2 Omicron variant is highly sensitive to molnupiravir, nirmatrelvir, and the combination. Cell Research. 2022; 32: 322–324.
[54]
Hattori S, Higashi-Kuwata N, Hayashi H, Allu SR, Raghavaiah J, Bulut H, et al. A small molecule compound with an indole moiety inhibits the main protease of SARS-CoV-2 and blocks virus replication. Nature Communications. 2021; 12: 668.
[55]
Li Q, Yi D, Lei X, Zhao J, Zhang Y, Cui X, et al. Corilagin inhibits SARS-CoV-2 replication by targeting viral RNA-dependent RNA polymerase. Acta Pharmaceutica Sinica B. 2021; 11: 1555–1567.
[56]
Sadeghi A, Ali Asgari A, Norouzi A, Kheiri Z, Anushirvani A, Montazeri M, et al. Sofosbuvir and daclatasvir compared with standard of care in the treatment of patients admitted to hospital with moderate or severe coronavirus infection (COVID-19): a randomized controlled trial. Journal of Antimicrobial Chemotherapy. 2020; 75: 3379–3385.
[57]
Eslami G, Mousaviasl S, Radmanesh E, Jelvay S, Bitaraf S, Simmons B, et al. The impact of sofosbuvir/daclatasvir or ribavirin in patients with severe COVID-19. Journal of Antimicrobial Chemotherapy. 2020; 75: 3366–3372.
[58]
Abbaspour Kasgari H, Moradi S, Shabani AM, Babamahmoodi F, Davoudi Badabi AR, Davoudi L, et al. Evaluation of the efficacy of sofosbuvir plus daclatasvir in combination with ribavirin for hospitalized COVID-19 patients with moderate disease compared with standard care: a single-centre, randomized controlled trial. Journal of Antimicrobial Chemotherapy. 2020; 75: 3373–3378.
[59]
Izzi A, Messina V, Rinaldi L, Maggi P. Editorial - Sofosbuvir/Velpatasvir as a combination with strong potential activity against SARS-CoV2 (COVID-19) infection: how to use direct-acting antivirals as broad-spectrum antiviral agents. European Review for Medical and Pharmacological Sciences. 2020; 24: 5193–5194.
[60]
Sayad B, Khodarahmi R, Najafi F, Miladi R, Mohseni Afshar Z, Mansouri F, et al. Efficacy and safety of sofosbuvir/velpatasvir versus the standard of care in adults hospitalized with COVID-19: a single-centre, randomized controlled trial. Journal of Antimicrobial Chemotherapy. 2021; 76: 2158–2167.
[61]
Khalil MI, Salih MA, Mustafa AA. Broad beans (Vicia faba) and the potential to protect from COVID-19 coronavirus infection. Sudanese Journal of Paediatrics. 2020; 20: 10–12.
[62]
Toelzer C, Gupta K, Yadav SKN, Borucu U, Davidson AD, Kavanagh Williamson M, et al. Free fatty acid binding pocket in the locked structure of SARS-CoV-2 spike protein. Science. 2020; 370: 725–730.
[63]
Li J, Xu D, Wang L, Zhang M, Zhang G, Li E, et al. Glycyrrhizic Acid Inhibits SARS-CoV-2 Infection by Blocking Spike Protein-Mediated Cell Attachment. Molecules. 2021; 26: 6090.
[64]
Caliebe RH, Scior T, Ammon HPT. Binding of boswellic acids to functional proteins of the SARS‐CoV‐2 virus: Bioinformatic studies. Archiv Der Pharmazie. 2021; 354: e2100160.
[65]
Gomaa AA, Mohamed HS, Abd-ellatief RB, Gomaa MA, Hammam DS. Advancing combination treatment with glycyrrhizin and boswellic acids for hospitalized patients with moderate COVID-19 infection: a randomized clinical trial. Inflammopharmacology. 2022; 30: 477–486.
[66]
Ohashi H, Watashi K, Saso W, Shionoya K, Iwanami S, Hirokawa T, et al. Potential anti-COVID-19 agents, cepharanthine and nelfinavir, and their usage for combination treatment. IScience. 2021; 24: 102367.
[67]
Wang R, Chan JF, Wang S, Li H, Zhao J, Ip TK, et al. Orally administered bismuth drug together with N-acetyl cysteine as a broad-spectrum anti-coronavirus cocktail therapy. Chemical Science. 2021; 13: 2238–2248.
[68]
Mahase E. Covid-19: UK becomes first country to authorise antiviral molnupiravir. British Medical Journal. 2021; 375: n2697.
[69]
Singh AK, Singh A, Singh R, Misra A. Molnupiravir in COVID-19: A systematic review of literature. Diabetology & Metabolic Syndrome. 2021; 15: 102329.
[70]
Hahn F, Wangen C, Häge S, Peter AS, Dobler G, Hurst B, et al. IMU-838, a Developmental DHODH Inhibitor in Phase II for Autoimmune Disease, Shows Anti-SARS-CoV-2 and Broad-Spectrum Antiviral Efficacy In Vitro. Viruses. 2020; 12: 1394.
[71]
Biering SB, Van Dis E, Wehri E, Yamashiro LH, Nguyenla X, Dugast-Darzacq C, et al. Screening a Library of FDA-Approved and Bioactive Compounds for Antiviral Activity against SARS-CoV-2. ACS Infectious Diseases. 2021; 7: 2337–2351.
[72]
Choy K, Wong AY, Kaewpreedee P, Sia SF, Chen D, Hui KPY, et al. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antiviral Research. 2020; 178: 104786.
[73]
Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, et al. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. New England Journal of Medicine. 2021; 384: 795–807.
[74]
Mesa RA. Tipifarnib: farnesyl transferase inhibition at a crossroads. Expert Review of Anticancer Therapy. 2006; 6: 313–319.
[75]
Munster P, Aggarwal R, Hong D, Schellens JHM, van der Noll R, Specht J, et al. First-in-Human Phase I Study of GSK2126458, an Oral Pan-Class i Phosphatidylinositol-3-Kinase Inhibitor, in Patients with Advanced Solid Tumor Malignancies. Clinical Cancer Research. 2016; 22: 1932–1939.
[76]
Kongsomros S, Suksatu A, Kanjanasirirat P, Manopwisedjaroen S, Prasongtanakij S, Jearawuttanakul K, et al. Anti-SARS-CoV-2 Activity of Extracellular Vesicle Inhibitors: Screening, Validation, and Combination with Remdesivir. Biomedicines. 2021; 9: 1230.
[77]
Pohl MO, Busnadiego I, Marrafino F, Wiedmer L, Hunziker A, Fernbach S, et al. Combined computational and cellular screening identifies synergistic inhibition of SARS-CoV-2 by lenvatinib and remdesivir. Journal of General Virology. 2021; 102: 001625.
[78]
Deng Q, Rasool RU, Russell RM, Natesan R, Asangani IA. Targeting androgen regulation of TMPRSS2 and ACE2 as a therapeutic strategy to combat COVID-19. iScience. 2021; 24: 102254.
[79]
Zhang Q, Chen CZ, Swaroop M, Xu M, Wang L, Lee J, et al. Heparan sulfate assists SARS-CoV-2 in cell entry and can be targeted by approved drugs in vitro. Cell Discovery. 2020; 6: 80.
[80]
Brunotte L, Zheng S, Mecate-Zambrano A, Tang J, Ludwig S, Rescher U, Schloer S. Combination Therapy with Fluoxetine and the Nucleoside Analog GS-441524 Exerts Synergistic Antiviral Effects against Different SARS-CoV-2 Variants In Vitro. Pharmaceutics. 2021; 13: 1400.
[81]
Gautret P, Lagier J, Parola P, Hoang VT, Meddeb L, Mailhe M, et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. International Journal of Antimicrobial Agents. 2020; 56: 105949.
[82]
Lagier J, Million M, Gautret P, Colson P, Cortaredona S, Giraud-Gatineau A, et al. Outcomes of 3,737 COVID-19 patients treated with hydroxychloroquine/azithromycin and other regimens in Marseille, France: a retrospective analysis. Travel Medicine and Infectious Disease. 2020; 36: 101791.
[83]
Molina JM, Delaugerre C, Le Goff J, Mela-Lima B, Ponscarme D, Goldwirt L, et al. No evidence of rapid antiviral clearance or clinical benefit with the combination of hydroxychloroquine and azithromycin in patients with severe COVID-19 infection. MéDecine Et Maladies Infectieuses. 2020; 50: 384.
[84]
Ramireddy A, Chugh H, Reinier K, Ebinger J, Park E, Thompson M, et al. Experience with Hydroxychloroquine and Azithromycin in the Coronavirus Disease 2019 Pandemic: Implications for QT Interval Monitoring. Journal of the American Heart Association. 2020; 9: e017144.
[85]
Kang CK, Seong M, Choi S, Kim TS, Choe PG, Song SH, et al. In vitro activity of lopinavir/ritonavir and hydroxychloroquine against severe acute respiratory syndrome coronavirus 2 at concentrations achievable by usual doses. The Korean Journal of Internal Medicine. 2020; 35: 782–787.
[86]
Abd-Elsalam S, Soliman S, Esmail ES, Khalaf M, Mostafa EF, Medhat MA, et al. Do Zinc Supplements Enhance the Clinical Efficacy of Hydroxychloroquine?: a Randomized, Multicenter Trial. Biological Trace Element Research. 2021; 199: 3642–3646.
[87]
Calistri A, Luganini A, Mognetti B, Elder E, Sibille G, Conciatori V, et al. The New Generation hDHODH Inhibitor MEDS433 Hinders the In Vitro Replication of SARS-CoV-2 and Other Human Coronaviruses. Microorganisms. 2021; 9: 1731.
[88]
Kawase M, Shirato K, van der Hoek L, Taguchi F, Matsuyama S. Simultaneous Treatment of Human Bronchial Epithelial Cells with Serine and Cysteine Protease Inhibitors Prevents Severe Acute Respiratory Syndrome Coronavirus Entry. Journal of Virology. 2012; 86: 6537–6545.
[89]
Raschi E, Caraceni P, Poluzzi E, De Ponti F. Baricitinib, JAK inhibitors and liver injury: a cause for concern in COVID-19? Expert Opinion on Drug Safety. 2020; 19: 1367–1369.
[90]
Tsuchiya K, Fujisawa T, Mochizuka Y, Takuma S, Oishi K, Endo Y, et al. Transient leukocytopenia following combination therapy for COVID-19. Respiratory Investigation. 2022; 60: 158–161.
[91]
Wang J, Pendurthi UR, Yi G, Rao LVM. SARS-CoV-2 infection induces the activation of tissue factor-mediated coagulation via activation of acid sphingomyelinase. Blood. 2021; 138: 344–349.
[92]
Carpinteiro A, Edwards MJ, Hoffmann M, Kochs G, Gripp B, Weigang S, et al. Pharmacological Inhibition of Acid Sphingomyelinase Prevents Uptake of SARS-CoV-2 by Epithelial Cells. Cell Reports Medicine. 2020; 1: 100142.
[93]
Facente SN, Reiersen AM, Lenze EJ, Boulware DR, Klausner JD. Fluvoxamine for the Early Treatment of SARS-CoV-2 Infection: a Review of Current Evidence. Drugs. 2021; 81: 2081–2089.
[94]
Reis G, dos Santos Moreira-Silva EA, Silva DCM, Thabane L, Milagres AC, Ferreira TS, et al. Effect of early treatment with fluvoxamine on risk of emergency care and hospitalisation among patients with COVID-19: the together randomised, platform clinical trial. The Lancet Global Health. 2022; 10: e42–e51.
[95]
Glebov OO. Low-Dose Fluvoxamine Modulates Endocytic Trafficking of SARS-CoV-2 Spike Protein: A Potential Mechanism for Anti-COVID-19 Protection by Antidepressants. Frontiers in Pharmacology. 2021; 12: 787261.
[96]
Kamat S, Kumari M. Repurposing Chloroquine Against Multiple Diseases With Special Attention to SARS-CoV-2 and Associated Toxicity. Frontiers in Pharmacology. 2021; 12: 576093.
[97]
Okesli A, Khosla C, Bassik MC. Human pyrimidine nucleotide biosynthesis as a target for antiviral chemotherapy. Current Opinion in Biotechnology. 2017; 48: 127–134.
[98]
Mahase E. Covid-19: Pfizer’s paxlovid is 89% effective in patients at risk of serious illness, company reports. British Medical Journal. 2021; 375: n2713.
[99]
Shytaj IL, Fares M, Gallucci L, Lucic B, Tolba MM, Zimmermann L, et al. The FDA-Approved Drug Cobicistat Synergizes with Remdesivir To Inhibit SARS-CoV-2 Replication In Vitro and Decreases Viral Titers and Disease Progression in Syrian Hamsters. mBio. 2022; 13: e0370521.
[100]
Chen J, Xia L, Liu L, Xu Q, Ling Y, Huang D, et al. Antiviral Activity and Safety of Darunavir/Cobicistat for the Treatment of COVID-19. Open Forum Infectious Diseases. 2020; 7: ofaa241.
[101]
Colunga Biancatelli RML, Berrill M, Catravas JD, Marik PE. Quercetin and Vitamin C: An Experimental, Synergistic Therapy for the Prevention and Treatment of SARS-CoV-2 Related Disease (COVID-19). Frontiers in Immunology. 2020; 11: 1451.
[102]
Papapanou M, Papoutsi E, Giannakas T, Katsaounou P. Plitidepsin: Mechanisms and Clinical Profile of a Promising Antiviral Agent against COVID-19. Journal of Personalized Medicine. 2021; 11: 668.
[103]
Benfield T, Bodilsen J, Brieghel C, Harboe ZB, Helleberg M, Holm C, et al. Improved Survival among Hospitalized Patients with Coronavirus Disease 2019 (COVID-19) Treated with Remdesivir and Dexamethasone. A Nationwide Population-Based Cohort Study. Clinical Infectious Diseases. 2021; 73: 2031–2036.
[104]
Ye Z, Yuan S, Chan JF, Zhang AJ, Yu C, Ong CP, et al. Beneficial effect of combinational methylprednisolone and remdesivir in hamster model of SARS-CoV-2 infection. Emerging Microbes & Infections. 2021; 10: 291–304.
[105]
D’Ascanio L, Vitelli F, Cingolani C, Maranzano M, Brenner MJ, Di Stadio A. Randomized clinical trial ”olfactory dysfunction after COVID-19: olfactory rehabilitation therapy vs. intervention treatment with Palmitoylethanolamide and Luteolin”: preliminary results. European Review for Medical and Pharmacological Sciences. 2021; 25: 4156–4162.
[106]
Robaina Cabrera CL, Keir-Rudman S, Horniman N, Clarkson N, Page C. The anti-inflammatory effects of cannabidiol and cannabigerol alone, and in combination. Pulmonary Pharmacology & Therapeutics. 2021; 69: 102047.
[107]
Santos S, Barata P, Charmier A, Lehmann I, Rodrigues S, Melosini MM, et al. Cannabidiol and Terpene Formulation Reducing SARS-CoV-2 Infectivity Tackling a Therapeutic Strategy. Frontiers in Immunology. 2022; 13: 841459.
[108]
López-Iranzo FJ, López-Rodas AM, Franco L, López-Rodas G. Pentoxifylline and Oxypurinol: Potential Drugs to Prevent the “Cytokine Release (Storm) Syndrome” Caused by SARS-CoV-2? Current Pharmaceutical Design. 2020; 26: 4515–4521.
[109]
Caprio M, Mammi C, Rosano GM. Vitamin D: a novel player in endothelial function and dysfunction. Archives of Medical Science. 2012; 8: 4–5.
[110]
Aini K, Fukuda D, Tanaka K, Higashikuni Y, Hirata Y, Yagi S, et al. Vildagliptin, a DPP-4 Inhibitor, Attenuates Endothelial Dysfunction and Atherogenesis in Nondiabetic Apolipoprotein E-Deficient Mice. International Heart Journal. 2019; 60: 1421–1429.
[111]
Pinheiro MM, Fabbri A, Infante M. Cytokine storm modulation in COVID-19: a proposed role for vitamin D and DPP-4 inhibitor combination therapy (VIDPP-4i). Immunotherapy. 2021; 13: 753–765.
[112]
Farouk A, Salman S. Dapsone and doxycycline could be potential treatment modalities for COVID-19. Medical Hypotheses. 2020; 140: 109768.
[113]
Jin Y, Ji W, Yang H, Chen S, Zhang W, Duan G. Endothelial activation and dysfunction in COVID-19: from basic mechanisms to potential therapeutic approaches. Signal Transduction and Targeted Therapy. 2020; 5: 293.
[114]
Debol SM, Herron MJ, Nelson RD. Anti-inflammatory action of dapsone: inhibition of neutrophil adherence is associated with inhibition of chemoattractant-induced signal transduction. Journal of Leukocyte Biology. 1997; 62: 827–836.
[115]
Sharma A, Sharma C, Raina S, Singh B, Dadhwal DS, Dogra V, et al. A randomized open-label trial to evaluate the efficacy and safety of triple therapy with aspirin, atorvastatin, and nicorandil in hospitalised patients with SARS Cov-2 infection: a structured summary of a study protocol for a randomized controlled trial. Trials. 2021; 22: 451.
[116]
Procter BC, Ross C, Pickard V, Smith E, Hanson C, McCullough PA. Clinical outcomes after early ambulatory multidrug therapy for high-risk SARS-CoV-2 (COVID-19) infection. Reviews in Cardiovascular Medicine. 2020; 21: 611–614.
[117]
Procter, MD BC, APRN, FNP-C CRM, PA-C, MPAS VP, PA-C, MPAS ES, PA-C, MPAS CH, McCullough, MD, MPH PA. Early Ambulatory Multidrug Therapy Reduces Hospitalization and Death in High-Risk Patients with SARS-CoV-2 (COVID-19). International Journal of Innovative Research in Medical Science. 2021; 6: 219–221.
[118]
Derwand R, Scholz M, Zelenko V. COVID-19 outpatients: early risk-stratified treatment with zinc plus low-dose hydroxychloroquine and azithromycin: a retrospective case series study. International Journal of Antimicrobial Agents. 2020; 56: 106214.
[119]
Boriskin Y, Leneva I, Pecheur E-, Polyak S. Arbidol: a Broad-Spectrum Antiviral Compound that Blocks Viral Fusion. Current Medicinal Chemistry. 2008; 15: 997–1005.
[120]
Deng L, Li C, Zeng Q, Liu X, Li X, Zhang H, et al. Arbidol combined with LPV/r versus LPV/r alone against Corona Virus Disease 2019: a retrospective cohort study. Journal of Infection. 2020; 81: e1–e5.
[121]
Chen J, Lin S, Niu C, Xiao Q. Clinical evaluation of Shufeng Jiedu Capsules combined with umifenovir (Arbidol) in the treatment of common-type COVID-19: a retrospective study. Expert Review of Respiratory Medicine. 2021; 15: 257–265.
[122]
Yuan S, Yin X, Meng X, Chan JF, Ye Z, Riva L, et al. Clofazimine broadly inhibits coronaviruses including SARS-CoV-2. Nature. 2021; 593: 418–423.
[123]
Gendrot M, Duflot I, Boxberger M, Delandre O, Jardot P, Le Bideau M, et al. Antimalarial artemisinin-based combination therapies (ACT) and COVID-19 in Africa: in vitro inhibition of SARS-CoV-2 replication by mefloquine-artesunate. International Journal of Infectious Diseases. 2020; 99: 437–440.
[124]
Shionoya K, Yamasaki M, Iwanami S, Ito Y, Fukushi S, Ohashi H, et al. Mefloquine, a Potent Anti-severe Acute Respiratory Syndrome-Related Coronavirus 2 (SARS-CoV-2) Drug as an Entry Inhibitor in vitro. Frontiers in Microbiology. 2021; 12: 651403.
[125]
Fintelman-Rodrigues N, Sacramento CQ, Ribeiro Lima C, Souza da Silva F, Ferreira AC, Mattos M, et al. Atazanavir, alone or in Combination with Ritonavir, Inhibits SARS-CoV-2 Replication and Proinflammatory Cytokine Production. Antimicrobial Agents and Chemotherapy. 2020; 64: e00825-20.
[126]
Purwati, Miatmoko A, Nasronudin, Hendrianto E, Karsari D, Dinaryanti A, et al. An in vitro study of dual drug combinations of anti-viral agents, antibiotics, and/or hydroxychloroquine against the SARS-CoV-2 virus isolated from hospitalized patients in Surabaya, Indonesia. PLoS ONE. 2021; 16: e0252302.
[127]
Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G, et al. A Trial of Lopinavir–Ritonavir in Adults Hospitalized with Severe Covid-19. New England Journal of Medicine. 2020; 382: 1787–1799.
[128]
Ahmad I, Alam M, Saadi R, Mahmud S, Saadi E. Doxycycline and Hydroxychloroquine as Treatment for High-Risk COVID-19 Patients: Experience from Case Series of 54 Patients in Long-Term Care Facilities. medRxiv. 2020. (in press)
[129]
Mahmud R, Rahman MM, Alam I, Ahmed KGU, Kabir AKMH, Sayeed SKJB, et al. Ivermectin in combination with doxycycline for treating COVID-19 symptoms: a randomized trial. Journal of International Medical Research. 2021; 49: 030006052110135.
[130]
Szolnoky G. Further aspects of doxycycline therapy in COVID-19. Dermatologic Therapy. 2020; 33: e13810.
[131]
Gautam SS, Gautam CS, Garg VK, Singh H. Combining hydroxychloroquine and minocycline: potential role in moderate to severe COVID-19 infection. Expert Review of Clinical Pharmacology. 2020; 13: 1183–1190.
[132]
Schloer S, Brunotte L, Mecate‐Zambrano A, Zheng S, Tang J, Ludwig S, et al. Drug synergy of combinatory treatment with remdesivir and the repurposed drugs fluoxetine and itraconazole effectively impairs SARS‐CoV‐2 infection in vitro. British Journal of Pharmacology. 2021; 178: 2339–2350.
[133]
XIA L, SHI Y, SU J, Friedemann T, TAO Z, Lu Y, et al. Shufeng Jiedu, a promising herbal therapy for moderate COVID-19:Antiviral and anti-inflammatory properties, pathways of bioactive compounds, and a clinical real-world pragmatic study. Phytomedicine. 2021; 85: 153390.
[134]
Gopal M, Padayatchi N, Metcalfe JZ, O’Donnell MR. Systematic review of clofazimine for the treatment of drug-resistant tuberculosis. The International Journal of Tuberculosis and Lung Disease. 2013; 17: 1001–1007.
[135]
Schloer S, Goretzko J, Rescher U. Repurposing Antifungals for Host-Directed Antiviral Therapy? Pharmaceuticals. 2022; 15: 212.
[136]
Van Damme E, De Meyer S, Bojkova D, Ciesek S, Cinatl J, De Jonghe S, et al. In vitro activity of itraconazole against SARS‐CoV‐2. Journal of Medical Virology. 2021; 93: 4454–4460.
[137]
Liesenborghs L, Spriet I, Jochmans D, Belmans A, Gyselinck I, Teuwen L, et al. Itraconazole for COVID-19: preclinical studies and a proof-of-concept randomized clinical trial. EBioMedicine. 2021; 66: 103288.
[138]
Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Research. 2020; 30: 269–271.
[139]
Moyle G, Back D. Principles and practice of HIV-protease inhibitor pharmacoenhancement. HIV Medicine. 2001; 2: 105–113.
[140]
Arabi YM, Alothman A, Balkhy HH, Al-Dawood A, AlJohani S, Al Harbi S, et al. Treatment of Middle East Respiratory Syndrome with a combination of lopinavir-ritonavir and interferon-β1b (MIRACLE trial): study protocol for a randomized controlled trial. Trials. 2018; 19: 81.
[141]
RECOVERY Collaborative Group. Lopinavir-ritonavir in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2020; 396: 1345–1352.
[142]
Sankar J, Dhochak N, Kabra SK, Lodha R. COVID-19 in Children: Clinical Approach and Management. The Indian Journal of Pediatrics. 2020; 87: 433–442.
[143]
Singh H, Kakkar AK, Chauhan P. Repurposing minocycline for COVID-19 management: mechanisms, opportunities, and challenges. Expert Review of Anti-Infective Therapy. 2020; 18: 997–1003.
[144]
Sodhi M, Etminan M. Therapeutic Potential for Tetracyclines in the Treatment of COVID-19. Pharmacotherapy. 2020; 40: 487–488.
[145]
Jacobs L, Johnson KP. A Brief History of the Use of Interferons as Treatment of Multiple Sclerosis. Archives of Neurology. 1994; 51: 1245–1252.
[146]
Lu C, Chen M, Lee W, Chang Y. Potential therapeutic agents against COVID-19: what we know so far. Journal of the Chinese Medical Association. 2020; 83: 534–536.
[147]
Dastan F, Nadji SA, Saffaei A, Marjani M, Moniri A, Jamaati H, et al. Subcutaneous administration of interferon beta-1a for COVID-19: a non-controlled prospective trial. International Immunopharmacology. 2020; 85: 106688.
[148]
Baghaei P, Dastan F, Marjani M, Moniri A, Abtahian Z, Ghadimi S, Valizadeh M, Heshmatnia J, Sadat Mirenayat M, Abedini A, Kiani A, Eslaminejad A, MohammadReza Hashemian S, Jamaati H, Zali A, Akbar Velayati A, Tabarsi P. Combination therapy of IFNβ1 with lopinavir-ritonavir, increases oxygenation, survival and discharging of sever COVID-19 infected inpatients. International Immunopharmacology. 2021; 92: 107329.
[149]
Ader F, Peiffer-Smadja N, Poissy J, Bouscambert-Duchamp M, Belhadi D, Diallo A, et al. An open-label randomized controlled trial of the effect of lopinavir/ritonavir, lopinavir/ritonavir plus IFN-β-1a and hydroxychloroquine in hospitalized patients with COVID-19. Clinical Microbiology and Infection. 2021; 27: 1826–1837.
[150]
Zhou Q, Chen V, Shannon CP, Wei XS, Xiang X, Wang X, et al. Interferon-α2b Treatment for COVID-19. Frontiers in Immunology. 2020; 11: 1061.
[151]
Wang B, Li D, Liu T, Wang H, Luo F, Liu Y. Subcutaneous injection of IFN alpha-2b for COVID-19: an observational study. BMC Infectious Diseases. 2020; 20: 723.
[152]
Hung IF, Lung KC, Tso EY, Liu R, Chung TW, Chu MY, et al. Triple combination of interferon beta-1b, lopinavir-ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, phase 2 trial. Lancet. 2020; 395: 1695–1704.
[153]
Ianevski A, Yao R, Lysvand H, Grødeland G, Legrand N, Oksenych V, et al. Nafamostat-Interferon-α Combination Suppresses SARS-CoV-2 Infection In Vitro and In Vivo by Cooperatively Targeting Host TMPRSS2. Viruses. 2021; 13: 1768.
[154]
Ianevski A, Yao R, Zusinaite E, Lello LS, Wang S, Jo E, et al. Synergistic Interferon-Alpha-Based Combinations for Treatment of SARS-CoV-2 and Other Viral Infections. Viruses. 2021; 13: 2489.
[155]
Ali S, Khalid S, Afridi M, Akhtar S, Khader YS, Akhtar H. Notes From the Field: The Combined Effects of Tocilizumab and Remdesivir in a Patient With Severe COVID-19 and Cytokine Release Syndrome. JMIR Public Health and Surveillance. 2021; 7: e27609.
[156]
Brown LK, Moran E, Goodman A, Baxendale H, Bermingham W, Buckland M, et al. Treatment of chronic or relapsing COVID-19 in immunodeficiency. Journal of Allergy and Clinical Immunology. 2022; 149: 557–561.e1.
[157]
Jassem J, Marek-Trzonkowska NM, Smiatacz T, Arcimowicz Ł, Papak I, Jassem E, Zaucha JM. Successful Treatment of Persistent SARS-CoV-2 Infection in a B-Cell Depleted Patient with Activated Cytotoxic T and NK Cells: A Case Report. International Journal of Molecular Sciences. 2021; 22: 10934.
[158]
Malsy J, Veletzky L, Heide J, Hennigs A, Gil-Ibanez I, Stein A, et al. Sustained Response after Remdesivir and Convalescent Plasma Therapy in a B-Cell–Depleted Patient with Protracted Coronavirus Disease 2019 (COVID-19). Clinical Infectious Diseases. 2021; 73: e4020–e4024.
[159]
Helleberg M, Niemann CU, Moestrup KS, Kirk O, Lebech A, Lane C, et al. Persistent COVID-19 in an Immunocompromised Patient Temporarily Responsive to Two Courses of Remdesivir Therapy. The Journal of Infectious Diseases. 2020; 222: 1103–1107.
[160]
Iaboni A, Wong N, Betschel SD. A Patient with X-Linked Agammaglobulinemia and COVID-19 Infection Treated with Remdesivir and Convalescent Plasma. Journal of Clinical Immunology. 2021; 41: 923–925.
[161]
Giudice V, Pagliano P, Vatrella A, Masullo A, Poto S, Polverino BM, et al. Combination of Ruxolitinib and Eculizumab for Treatment of Severe SARS-CoV-2-Related Acute Respiratory Distress Syndrome: A Controlled Study. Frontiers in Pharmacology. 2020; 11: 857.
[162]
Rosa-Guerrero P, Trujillo-Aguilera A, Molina J, Navas A, López-Martín C, Jurado A, et al. Case Report: Successful Response to Intravenous Immunoglobulin and Steroid Pulses in a Renal Transplant Recipient With Severe Covid-19 Disease and Associated Acute Allograft Failure. Frontiers in Immunology. 2021; 12: 671013.
[163]
Sancho-López A, Caballero-Bermejo AF, Ruiz-Antorán B, Múñez Rubio E, García Gasalla M, Buades J, et al. Efficacy and Safety of Sarilumab in patients with COVID19 Pneumonia: a Randomized, Phase III Clinical Trial (SARTRE Study). Infectious Diseases and Therapy. 2021; 10: 2735–2748.
[164]
Wyler E, Adler JM, Eschke K, Teixeira Alves G, Peidli S, Pott F, et al. Key benefits of dexamethasone and antibody treatment in COVID-19 hamster models revealed by single-cell transcriptomics. Molecular Therapy. 2022; 30: 1952–1965.
[165]
Cai Y, Xu W, Gu C, Cai X, Qu D, Lu L, et al. Griffithsin with a Broad-Spectrum Antiviral Activity by Binding Glycans in Viral Glycoprotein Exhibits Strong Synergistic Effect in Combination with a Pan-Coronavirus Fusion Inhibitor Targeting SARS-CoV-2 Spike S2 Subunit. Virologica Sinica. 2020; 35: 857–860.
[166]
Akhter J, Quéromès G, Pillai K, Kepenekian V, Badar S, Mekkawy AH, et al. The Combination of Bromelain and Acetylcysteine (BromAc) Synergistically Inactivates SARS-CoV-2. Viruses. 2021; 13: 425.
[167]
Coelho Dos Reis JGA, Ferreira GM, Lourenço AA, Ribeiro ÁL, da Mata CPDSM, de Melo Oliveira P, et al. Ex-vivo mucolytic and anti-inflammatory activity of BromAc in tracheal aspirates from COVID-19. Biomedicine & Pharmacotherapy. 2022; 148: 112753.
[168]
Kritis P, Karampela I, Kokoris S, Dalamaga M. The combination of bromelain and curcumin as an immune-boosting nutraceutical in the prevention of severe COVID-19. Metabolism Open. 2020; 8: 100066.
[169]
Lusvarghi S, Bewley CA. Griffithsin: An Antiviral Lectin with Outstanding Therapeutic Potential. Viruses. 2016; 8: 296.
[170]
Soni VK, Mehta A, Ratre YK, Tiwari AK, Amit A, Singh RP, et al. Curcumin, a traditional spice component, can hold the promise against COVID-19? European Journal of Pharmacology. 2020; 886: 173551.
[171]
Sagar S, Rathinavel AK, Lutz WE, Struble LR, Khurana S, Schnaubelt AT, et al. Bromelain inhibits SARS-CoV-2 infection in VeroE6 cells. bioRxiv. 2020. (in press)
[172]
Troyano-Hernáez P, Reinosa R, Holguín Á. Evolution of SARS-CoV-2 Envelope, Membrane, Nucleocapsid, and Spike Structural Proteins from the Beginning of the Pandemic to September 2020: A Global and Regional Approach by Epidemiological Week. Viruses. 2021; 13: 243.
[173]
Greaney AJ, Starr TN, Gilchuk P, Zost SJ, Binshtein E, Loes AN, et al. Complete Mapping of Mutations to the SARS-CoV-2 Spike Receptor-Binding Domain that Escape Antibody Recognition. Cell Host & Microbe. 2021; 29: 44–57.e9.
Share
Back to top