Abstract
Since time immemorial human beings have constantly been fighting against viral infections. The ongoing and devastating COVID-19 pandemic represents one of the most severe and most significant public health emergencies in human history, highlighting an urgent need to develop broad-spectrum antiviral agents. Salicylamide (2-hydroxybenzamide) derivatives, represented by niclosamide and nitazoxanide, inhibit the replication of a broad range of RNA and DNA viruses such as flavivirus, influenza A virus and coronavirus. Moreover, nitazoxanide was effective in clinical trials against different viral infections including diarrhea caused by rotavirus and norovirus, uncomplicated influenza A and B, hepatitis B, and hepatitis C. In this review, we summarize the broad antiviral activities of salicylamide derivatives, the clinical progress, and the potential targets or mechanisms against different viral infections and highlight their therapeutic potential in combating the circulating and emerging viral infections in the future.
Keywords: salicylamide derivatives, infectious diseases, nitazoxanide, niclosamide, broad-spectrum antiviral agents
1. INTRODUCTION
There has been more than ten significant epidemics or pandemics of various viral diseases in the past two decades of the twenty-first century. This includes dengue, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), chikungunya, Zika fever, Ebola virus (EBOV) disease, H1N1 influenza, coronavirus disease 2019 (COVID-19), etc.1 The ongoing COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents one of the most challenging public health emergencies in human history2. Since SARS-CoV-2 was first reported in December 2019 in Wuhan, a series of public health measures such as business closings, staying-at-home orders, mask-wearing, and social distance were taken to control its spread3. However, due to its ability for asymptomatic transmission, SARS-CoV-2 quickly raged worldwide and was declared a global pandemic on March 11, 2020, by the World Health Organization (WHO), causing millions of associated fatalities and enormous economic losses4,5. Although it usually takes several years to develop new therapeutics, especially for novel emerging pathogens, the antiviral agent remdesivir was rapidly authorized emergence use by the FDA in May 2020 and finally approved in October 2020 for the treatment of COVID-196–8. Remdesivir was developed by Gilead initially to combat EBOV infection as an RNA-dependent RNA polymerase (RdRp) inhibitor and displayed broad-spectrum antiviral activities, highly effective against filoviruses, paramyxoviruses, respiratory syncytial virus (RSV), and pathogenic coronaviruses (CoVs)9–11. After the outbreak of COVID-19, remdesivir was quickly found to potently inhibit SARS-CoV-2 replication and shorten the recovery time in hospitalized patients with COVID-19, which promoted its clinical use to benefit numerous COVID-19 patients12,13. Molnupiravir, another broad-spectrum antiviral agent of an RdRp inhibitor developed by Emory University and Merck, showed robust anti-SARS-CoV-2 activity14. A phase 3, double-blind, randomized, placebo-controlled trial revealed that early treatment with molnupiravir could reduce the risk of hospitalization or in at-risk, unvaccinated COVID-19 patients, and it was authorized emergence use by the FDA on December 23, 2021 as an oral drug to treat mild-to-moderate COVID-19 in adults15,16. The rest of the three FDA-approved anti-SARS-CoV-2 drugs is nirmatrelvir which is an oral 3C-like protease (3CLpro) inhibitor developed by Pfizer. In a phase 2–3 double-blind, randomized, controlled trial, treatment with nirmatrelvir plus ritonavir resulted in a lower risk of progression to severe COVID-19 than that with placebo (relative risk reduction, 89.1%)17. The combination of nirmatrelvir and ritonavir (the brand name Paxlovid) was authorized as the first oral antiviral by FDA on December 22, 2021, for the treatment of mild-to-moderate COVID-19 in patients (12 years of age and older)18. As highlighted by the successful examples of remdesivir and molnupiravir, it is imperative to develop broad-spectrum small molecule antiviral agents to fight the highly heterogeneous unknown viral infections that may emerge in the future.
Salicylamide (2-hydroxybenzamide) derivatives are a class of compounds that possess broad-spectrum antiviral activities, exemplified by niclosamide (1) and nitazoxanide (2) (Figure 1)19–21. Niclosamide is an FDA-approved anthelminthic drug used to treat tapeworm infections in human for several decades. It works by suppressing oxidative phosphorylation and stimulating adenosine triphosphatase activity in the mitochondria. Niclosamide can modulate multiple signaling pathways and biological processes, including Wnt/β-catenin, STAT3, NF-κB, mTORC1, etc22. Nitazoxanide was initially developed as an oral antiparasitic agent and finally approved by FDA as an Orphan Drug to treat diarrhea caused by Cryptosporidium parvum and Giardia intestinalis in adults and children (≥ 12 months of age)21. Its mechanism of action against protozoa and anaerobic bacteria is related to inhibiting pyruvate:ferredoxin oxidoreductase (PFOR), an enzyme essential for anaerobic energy metabolism21. Nitazoxanide disrupts the membrane potential and pH homeostasis of Mycobacterium tuberculosis (Mtb), acting as an uncoupler23. Accumulated studies indicated that niclosamide, nitazoxanide, and its active circulating metabolite, tizoxanide (3), inhibit the replication of a broad range of RNA and DNA viruses such as flavivirus, influenza A virus, and coronavirus20,21,24. Moreover, nitazoxanide was effective in clinical trials against different viral infections, including diarrhea caused by rotavirus and norovirus, uncomplicated influenza A and B, hepatitis B, and hepatitis C21,24. In recent years, structure-activity relationship (SAR) optimization on salicylamide derivatives has obtained more effective and safer antiviral agents against different viruses24–31. This review summarizes the broad antiviral activities of salicylamide derivatives, the clinical progress, and the potential targets or mechanisms against different viral infections and highlights their therapeutic potential in combating the circulating and emerging viral infections in the future.
Figure 1.

The chemical structures of representative salicylamide derivatives niclosamide, nitazoxanide, and its metabolite tizoxanide.
2. ANTIVIRAL ACTIVITY OF SALICYLAMIDE DERIVATIVES
2.1. Anti-Coronavirus Activity
CoVs are a group of enveloped and positive-sense single-stranded RNA viruses which can be divided into four genera (α, β, γ, and δ) belonging to the family Coronaviridae within the order Nidovirales20. Currently, seven human coronaviruses (HCoVs) have been identified, including two α-CoVs, HCoV-NL63 and HCoV-229E, and five β-CoVs, HCoV-HKU1, HCoV-OC43, severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-29,32. Among them, four human strains, HCoV-229E, HCoV-OC43, HCoV-NL63, and HCoV-HKU1 usually produce mild symptoms like the common cold. In comparison, three strains, SARS-CoV, MERS-CoV, and SARS-CoV-2, can cause severe acute respiratory diseases (SARS, MERS, and COVID-19, respectively) with high morbidity and mortality9. The outbreak of SARS between November 2002 and July 2003 finally brought about 8098 confirmed cases and 774 associated deaths (case-fatality rate: 9.6%) in 29 countries and regions, while MERS-CoV, first reported in June 2012 in Saudi Arabia, has caused 2519 laboratory-confirmed cases in 27 countries, leading to 866 associated deaths (case-fatality rate: 34.3%) by the end of January 202033,34. SARS-CoV-2, as the third highly pathogenic HCoV, was first reported in Wuhan, China, in December 2019 and then fast spread worldwide due to its high transmissibility, leading to an ongoing COVID-19 pandemic3,9. As of June 20, 2021, SARS-CoV-2 accounted for more than 177 million cases of infection and more than 3,857,972 associated deaths globally5. Although remdesivir, molnupiravir, and nirmatrelvir as well as three vaccines (Pfizer-BioNTech, Moderna, and Janssen) have been approved by FDA for the treatment or prevention of COVID-19, there is still an urgent need to develop more effective and safer small molecule antiviral therapeutics to cope with the emergence of various SARS-CoV-2 variants with increased transmissibility8,35–39.
Niclosamide inhibits SARS-CoV and SARS-CoV-2 replication with EC50s of < 0.1 μM and 0.28 μM, respectively, and suppresses MERS-CoV infection by up to 1000-fold at 48 h post-infection (p.i.) at a concentration of 10 μM (Table 1)40–43. Niclosamide was reported to reduce MERS-CoV replication through S-phase kinase-associated protein 2 (SKP2) inhibition which increases the Benclin 1 (BECN1) level and enhances autophagy while it blocks SARS-CoV-2 spike-induced syncytia via inhibiting the activity of the calcium-activated TMEM16F (a.k.a. anoctamin 6), which acts as both a chloride channel and a lipid scramblase responsible for phosphatidylserine internalization on the cell surface42,44. When human volunteers received a single oral dose of 2000 mg of niclosamide, the maximum serum concentration of niclosamide reached 0.25–6.0 μg/mL (0.76–18.3 μM)45. To further improve the concentration of niclosamide in the lung, a significant site of SARS-CoV-2 infection, a new formulation for inhalation administration was developed and well-tolerated in healthy volunteers, warranting its further evaluation in COVID-19 patients46. Currently, several clinical trials have been conducted to test the efficacy of niclosamide in COVID-19 treatment. Broad anti-CoV activities were also observed for nitazoxanide and tizoxanide, displaying EC50 values of ~3 μM against MERS-CoV and SARS-CoV-213,47–49. In a phase 2b/3 clinical trial treating patients with nitazoxanide (600 mg controlled-release tablet) twice daily for 5 days, during the first 12 h after dosing on the morning of day 2, the plasma concentrations of tizoxanide ranged from 17.3 μM (4.6 μg/ml) to 3.0 μM (0.8 μg/ml), above the in vitro EC50 value against SARS-CoV-2, supporting the repurposing of nitazoxanide for the treatment of COVID-1950. Moreover, oral administration of one 500 mg nitazoxanide tablet with food could further increase the maximum plasma concentration of tizoxanide approximately to 37 μM (10 μg/mL)51,52. Nitazoxanide is currently undergoing a series of clinical trials to evaluate its efficacy as a monotherapy or in combination with other antivirals to treat COVID-19. Shamim et al. reported niclosamide derivatives 4-9 (Figure 2), which showed potent inhibitory activities against SARS-CoV-2 infection with sub-micromolar EC50 values, similar to niclosamide53. Especially, compound 9 with 5-bromo substitution in the salicylic acid region also displayed better drug-like properties, offering a good candidate for further evaluation in in vivo models of COVID-19. Recently, our group found that compound 10 with 3´-Cl-4´-NO2-aniline moiety was a potent allosteric inhibitor against SARS-CoV-2 3CLpro (IC50 = 3.9 μM) and exhibited anti-SARS-CoV-2 activity (EC50 = 1.75 μM) in A549-hACE2 cells54. In addition, Juan et al. also reported a series of niclosamide derivatives including compounds 10-12 as potent inhibitors against SARS-CoV-2 (EC50 = 0.057 μM, 0.39 μM, and 0.49μM, respectively). The phosphatidylserine externalization experiment through fluorescence microscopy demonstrated the inhibition of compound 10 towards TMEM16F, similar to niclosamide. Moreover, compounds 10-12 displayed higher stability in human plasma and liver S9 enzymes assay than niclosamide55. Blake et al. screened their salicylanilide compound library and found that compound 13 inhibited SARS-CoV-2 infection with an EC50 of 0.74 μM29. Compound 13 displayed a good bioavailability of 76% in mice. The treatment of compound 13 (when administrated orally at 5 mg/kg twice daily for 5 days) led to 10-fold lower viral titers, reduced the induction of key cytokines, and mitigated the associated weight loss in a mouse model of SARS-CoV-2 infection29,56. Considering its relatively high cytotoxicity in Vero cells (CC50 = 1.22 μM), the in vivo safety of compound 13 needs to be further investigated.
Table 1.
The activity of salicylamide derivatives against coronavirusa
| Compound | Activity against coronavirus | Ref. |
|---|---|---|
| Niclosamide (1) | SARS-CoV: EC50 < 0.1 μM, CC50 = 22.1 μM (Vero E6 cells) SARS-CoV-2: EC50 = 0.28 μM, CC50 > 50 μM (Vero cells) SARS-CoV-2: EC50 = 0.11 μM (Vero E6 cells/ USA_WA1/2020) Suppressed MERS-CoV infection by ~1000-fold at 48 h at 10 μM |
40,42,43,53 |
| Nitazoxanide (2) | Canine CoV S-378: EC50 = 3.3 μM (A72 cells) MERS-CoV: EC50 =3.0 μM (LLC-MK2 cells) SARS-CoV-2: EC50 = 2.12 μM, CC50 = 35.53 μM (Vero E6 cells) |
13,47,49 |
| Tizoxanide (3) | MERS-CoV: EC50 =3.1 μM (LLC-MK2 cells) SARS-CoV-2: EC50 = 3.16 μM (Vero E6 cells) |
47,48 |
| 4 | SARS-CoV-2: EC50 = 0.32 μM (Vero E6 cells/ USA_WA1/2020), cell viability: 61% at 30 μM in SNB-19 cells | 53 |
| 5 | SARS-CoV-2: EC50 = 0.38 μM (Vero E6 cells/ USA_WA1/2020), cell viability: 59% at 30 μM in SNB-19 cells | 53 |
| 6 | SARS-CoV-2: EC50 = 0.26 μM (Vero E6 cells/ USA_WA1/2020), cell viability: 63% at 30 μM in SNB-19 cells | 53 |
| 7 | SARS-CoV-2: EC50 = 0.13 μM (Vero E6 cells/ USA_WA1/2020), cell viability: 59% at 30 μM in SNB-19 cells; EC50 = 0.38 μM, CC50 = 1.18 μM (Vero E6 cells) | 53,55 |
| 8 | SARS-CoV-2: EC50 = 0.14 μM (Vero E6 cells/ USA_WA1/2020), cell viability: 55% at 30 μM in SNB-19 cells | 53 |
| 9 | SARS-CoV-2: EC50 = 0.10 μM (Vero E6 cells/ USA_WA1/2020), cell viability: 59% at 30 μM in SNB-19 cells | 53 |
| 10 | SARS-CoV-2: EC50 = 1.75 μM, CC50 = 4.75 μM (A549-hACE2 cells); EC50 = 0.057 μM, CC50 = 1.51 μM (Vero E6 cells); | 54,55 |
| 11 | SARS-CoV-2: EC50 = 0.39 μM, CC50 = 2.46 μM (Vero E6 cells) | 55 |
| 12 | SARS-CoV-2: EC50 = 0.49 μM, CC50 = 5.95 μM (Vero E6 cells) | 55 |
| 13 | SARS-CoV-2: EC50 = 0.74 μM, CC50 = 1.22 μM (Vero cells) | 29 |
SARS-CoV, severe acute respiratory syndrome coronavirus; MERS-CoV, Middle East respiratory syndrome coronavirus.
Figure 2.

The chemical structures of salicylamide derivatives 4-13
2.2. Anti-Flavivirus Activity
The flaviviruses (genus Flaviviruses) are a diverse group of over 50 species of arthropod-borne viruses (arboviruses) in the family Flaviviridae, many of which are human pathogens of global importance57. Among them, dengue virus four serotypes (DENV 1–4), Zika virus (ZIKV), yellow fever virus (YFV), Japanese encephalitis virus (JEV), tick-borne encephalitis virus (TBEV), and West Nile virus (WNV) are the best-known which can cause significant morbidity and mortality around the world58. DENV infection can cause life-threatening diseases called severe dengue, including dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS)59. Approximately 3.9 billion people in 128 countries are at risk of DNEV infection, and 390 million dengue infections are estimated to occur annually, leading to about 500,000 cases of severe dengue and 22,000 deaths worldwide60. Several significant ZIKV outbreaks have occurred worldwide in recent years, resulting in devastating diseases such as congenital microcephaly and Guillain-Barré syndrome31. Currently, effective vaccines are available for YFV, JEV, and TBEV but do not exist for WNV and ZIKV57; a DENV vaccine (Dengvaxia®) was recently approved in a few countries for people aged 9 to 45 years and further recommended to those with confirmed previous DENV infection by WHO59. However, there remains no clinically approved antiviral therapy to manage these viruses61.
Based on the previous finding that ZIKV infection of human neural progenitor cells (hNPCs) leads to an increased caspase-3 activation and subsequent cell death62, Xu et al. carried out a phenotypic screen via using the caspase-3 activity assay and identified niclosamide as an inhibitor of ZIKV infection that significantly suppressed production of infectious ZIKV particles with an IC50 value of 0.28 μM63. To improve the physicochemical properties, their group further conducted SAR studies focusing on the anilide and salicylic acid regions of niclosamide53. Introducing various substituents in these two regions generated compounds 4–9 and 14-17 (Figures 2 & 3), which displayed sub-micromolar to low micromolar inhibitory activities against ZIKV (Table 2). Modifications of 2´-H (4), 2´-OMe (5), and 5-Br (9) substitutions are advantageous with increased aqueous solubility and permeability, compared to niclosamide53. After screening 2816 approved and investigational drugs through a novel luciferase complementation (SLC)-based high-throughput screening (HTS) assay, our research team independently identified niclosamide and nitazoxanide as flavivirus N2B-NS3 interaction inhibitors which target the interface between viral protease NS3 and its cofactor NS2B and result in accumulation of nonfunctional viral polyprotein precursor64. Niclosamide inhibited the NS2B-NS3 protease activity with an IC50 value of 12.3 μM and showed similar in vitro efficacies for ZIKV, DENV-2, WNV, JEV, and YFV with EC50 values of 0.48–1.02 μM64. Nitazoxanide displayed an IC50 value of 15.9 μM against the NS2B-NS3 protease and EC50 values of 1.48 μM and 0.39 μM against ZIKV and JEV, respectively64,65. In vivo studies showed that nitazoxanide treatment can protect mice from the challenge of JEV65. Further screening a small library of niclosamide derivatives, we found compounds 18-20 maintained sub-micromolar potency against ZIKV and DENV-2, and 19 (JMX0207) with a 3-NO2 group also exhibited improved efficacy in inhibition of NS2B-NS3 interaction. Intriguingly, JMX0207 displayed significantly improved pharmacokinetic (PK) properties and the in vivo efficacy in the A129 ZIKV mouse model31. The docking studies indicated that JMX0207 and niclosamide could be well docked into the NS3 pocket, termed as 2B53 pocket, in a similar manner using the induced fit docking (IFD) protocol (Figure 4)31,64. JMX0207 and niclosamide were equally effective in reducing ZIKV titer even added at 24 h post-infection, suggesting that they act at a post-entry step31,64. Besides N2B-NS3 interaction inhibition, niclosamide was found to neutralize the low-pH intracellular compartments, which affect multiple steps of the DENV life cycle, including viral and host membrane fusion and uncoating, viral RNA replication, and maturation of virus particles rendering it non-infectious66,67.
Figure 3.

The chemical structures of salicylamide derivatives 14-20
Table 2.
The activity of salicylamide derivatives against flavivirusa
| Compound | Activity against flavivirus | Ref. |
|---|---|---|
| Niclosamide (1) | ZIKV: EC50 = 0.28 μM (SNB-19 cells); EC50 = 0.48 μM, CC50 = 4.8 μM (A549 cells/ PRV-ABC59) DENV-2: EC50 = 0.55 μM (A549 cells) IC50-pro = 12.3 μM (DENV-2 NS3-MBP fusion protein) WNV: EC50 = 0.54 μM (A549 cells) JEV: EC50 = 1.02 μM (A549 cells) YFV: EC50 = 0.84 μM (A549 cells) |
63,64 |
| Nitazoxanide (2) | ZIKV: EC50 = 1.48 μM, CC50 = 4.8 μM (A549 cells/ PRV-ABC59) IC50-pro = 15.9 μM (DENV-2 NS3-MBP fusion protein) JEV: EC50 = 0.39 μM, CC50 = 60.5 μM (BHK-21 cells) |
64,65 |
| Tizoxanide (3) | Completely abolished supernatant ZIKV proliferation at 10 μM | 225 |
| 4 | ZIKV: EC50 = 0.82 μM (SNB-19 cells/ viral titer reduction); EC50 = 2.5 μM (HEK293 cells/ NS1 assay), cell viability: 61% at 30 μM in SNB-19 cells | 53 |
| 5 | ZIKV: EC50 = 0.82 μM (SNB-19 cells/ viral titer reduction); EC50 = 2.5 μM (HEK293 cells/ NS1 assay), cell viability: 59% at 30 μM in SNB-19 cells | 53 |
| 6 | ZIKV: EC50 = 0.81 μM (SNB-19 cells/ viral titer reduction); EC50 = 1.7 μM (HEK293 cells/ NS1 assay), cell viability: 63% at 30 μM in SNB-19 cells | 53 |
| 7 | ZIKV: EC50 = 0.84 μM (SNB-19 cells/ viral titer reduction); EC50 = 0.8 μM (HEK293 cells/ NS1 assay), cell viability: 59% at 30 μM in SNB-19 cells | 53 |
| 8 | ZIKV: EC50 = 0.4 μM (SNB-19 cells/ viral titer reduction); EC50 = 1.6 μM (HEK293 cells/ NS1 assay), cell viability: 55% at 30 μM in SNB-19 cells | 53 |
| 9 | ZIKV: EC50 = 0.11 μM (SNB-19 cells/ viral titer reduction); EC50 = 0.84 μM (HEK293 cells/ NS1 assay), cell viability: 59% at 30 μM in SNB-19 cells | 53 |
| 14 | ZIKV: EC50 = 1.72 μM (HEK293 cells/ NS1 assay) | 53 |
| 15 | ZIKV: EC50 = 1.74 μM (HEK293 cells/ NS1 assay) | 53 |
| 16 | ZIKV: EC50 = 3.0 μM (HEK293 cells/ NS1 assay) | 53 |
| 17 | ZIKV: EC50 = 2.34 μM (HEK293 cells/ NS1 assay) | 53 |
| 18 | ZIKV: EC50 = 0.15 μM, CC50 = 50.2 μM (A549 cells/ PRV-ABC59) DENV-2: EC50 = 0.21 μM (A549 cells) IC50-pro = 44.5 μM (DENV-2 NS3-MBP fusion protein) |
31 |
| 19 | ZIKV: EC50 = 0.30 μM, CC50 = 31.9 μM (A549 cells/ PRV-ABC59) DENV-2: EC50 = 0.31 μM (A549 cells) IC50-pro = 8.2 μM (DENV-2 NS3-MBP fusion protein) |
31 |
| 20 | ZIKV: EC50 = 1.1 μM, CC50 = 241 μM (A549 cells/ PRV-ABC59) DENV-2: EC50 = 0.9 μM (A549 cells) IC50-pro = 20.3 μM (DENV-2 NS3-MBP fusion protein) |
31 |
DENV, dengue virus; ZIKV, Zika virus; YFV, yellow fever virus; JEV, Japanese encephalitis virus; TBEV; tick-borne encephalitis virus; WNV, West Nile virus.
Figure 4.

A) JMX0207 (magenta) docked into NS3pro of DENV2 (PDB Code: 2FOM). Binding site key interaction residues are highlighted in gray sticks. H-bonds are shown as purple dotted lines and π cation and π stacking are shown as cyan dotted lines. B) Niclosamide (green) docked into NS3pro of DENV2. H-bonds are shown as purple dotted lines. C) JMX0207 (magenta) and Niclosamide (green) superimposed at the predicted binding site in surface representation.
2.3. Anti-Hepatitis B Virus Activity
Hepatitis B virus (HBV) is a partially double-stranded DNA virus belonging to the genus Orthohepadnavirus of the family Hepadnaviridae, which can cause both acute (AHB) and chronic hepatitis B (CHB)68–70. HBV is primarily transmitted through contact with infected blood and body fluids including vertical transmission from mother to child during delivery or sexual contact70. HBV infection occurring in fancy or early childhood usually leads to lifelong chronic infection and carriage of the virus. In contrast, infection acquired later in life commonly develops into an acute, self-limiting illness with final successful control of the virus71. WHO estimated that in 2015, 257 million people had chronic HBV infection, 2.7 million people were coinfected with HBV and human immunodeficiency virus (HIV), and about 887,000 people died from long-term complications of HBV infection, mainly cirrhosis and hepatocellular carcinoma72,73. Vaccination remains the most effective measure to prevent HBV infection. HBV treatment is limited to either a few nucleoside/nucleotide drugs that inhibit reverse transcription or immunomodulatory agents such as conventional or pegylated type I interferons (IFNs) and is recommended for chronically infected patients with persistently elevated HBV DNA levels and serum alanine aminotransferase (a marker of liver damage)71,74. However, current anti-HBV drugs rarely attain complete viral elimination from patients (HBV cure), and a functional cure defined as the loss of detectable serum hepatitis B surface antigen (HBsAg) is a more practical goal for anti-HBV therapy, which is also not frequently achieved with currently available treatment74.
Nitazoxanide and tizoxanide were reported to inhibit both intracellular HBV replication (EC50 = 0.59 μM and 0.46 μM, respectively) and extracellular virus production (EC50 = 0.12 μM and 0.15 μM, respectively) with low cytotoxicity and high selectivity index (Table 3)51. Nitazoxanide could efficiently suppress the interaction between HBV regulatory protein X (HBx) and the host protein damage-specific DNA-binding protein 1 (DDB1), leading to the restoration of structural maintenance of chromosomes 5 (Smc5) protein levels and inhibition of viral transcription and viral protein production in the HBV minicircle system as well as in human primary hepatocytes naturally infected with HBV75. Two successful cases were reported that patients with CHB receiving nitazoxanide treatment or combination therapy of nitazoxanide with adefovir for two years developed undetectable HBV DNA as well as negative HBeAg and HBsAb76. In one pilot clinical trial involving 12 adults with CHB receiving nitazoxanide treatment (500 mg, twice daily for 12 months), 7 of 8 patients with hepatitis B e antigen (HBeAg) negative initially became HBV DNA negative, and two of them became HBsAg negative; 3 of 4 patients with HBeAg positive initially became HBeAg negative, two of them became HBV DNA negative, and one patient became HBsAg negative77. In another clinical trial, 8 of 9 patients with CHB displayed negative serum HBV DNA after nitazoxanide treatment for 4–20 weeks. Two patients with HBeAg positive initially became HBeAg negative, and 3 of 9 patients became HBsAg negative78. These data provide clinical evidence of the therapeutic potential of nitazoxanide in treating patients with CHB, and the clinical trial (NCT03905655) has been initiated to evaluate the effectiveness of the combined use of nitazoxanide with existing regions for the treatment of CHB.
Table 3.
Activities of salicylamides derivatives against HBV replicationa
| Compound | extracellular virion DNA | intracellular HBV RI | CC50 (μM) | SI | |||
|---|---|---|---|---|---|---|---|
| EC50 (μM) | EC90 (μM) | EC50 (μM) | EC90 (μM) | virion | RI | ||
| Nitazoxanide (2) | 0.12 | 0.83 | 0.59 | 2.10 | >100 | >121 | >48 |
| Tizoxanide (3) | 0.15 | 0.58 | 0.46 | 1.20 | >100 | >172 | >83 |
| 21 | 0.64 | 3.5 | 2.10 | 12.0 | 85 | 24 | 7.1 |
| 22 | 0.33 | 0.89 | 1.00 | 2.90 | 38 | 43 | 13 |
| 23 | 0.33 | 0.83 | 0.90 | 2.0 | 100 | >120 | >51 |
| 24 | 0.32 | 2.00 | 0.59 | 3.90 | 30 | 15 | 7.7 |
| 25 | 0.21 | 1.10 | 3.50 | 11.0 | 45.0 | 41 | 4.1 |
| 26 | 3.2 | 9.4 | 5.5 | 14.0 | >100 | >11 | >7.1 |
| 27 | 3.8 | 11.0 | 9.1 | 27.0 | >100 | >9.1 | >3.7 |
| 28 | 0.22 | 0.73 | 7.80 | 30.0 | >100 | >137 | 3.3 |
| 29 | 0.76 | 3.00 | 2.10 | 6.80 | >100 | >33 | >15 |
| 30 | 0.20 | 1.20 | 0.73 | 3.30 | >100 | >83 | >30 |
| 31 | 3.70 | 13.0 | >100 | ||||
| 32 | 0.28 | 0.73 | 0.66 | 1.7 | 89 | 122 | 52 |
| Niclosamide (1) | >10.0 | >10.0 | >100 | ||||
CC50: drug concentration at which a 2-fold lower level of neutral red dye uptake is observed relative to the average level in untreated cultures. RI: replication intermediate. EC50 or EC90: drug concentration required to reduce extracellular virion HBV DNA or intracellular HBV DNA by 50% or 90% relative to untreated cells. SI (virion) or SI (RI): CC50/EC90 for extracellular or intracellular HBV.
The SAR studies on nitazoxanide derivatives as antiviral agents were conducted by Stachulski et al., and thiazolides 21-28 (Figure 5) with different substitutions exhibited sub-micromolar to micromolar EC50 values against both intracellular HBV intermediates and extracellular HBV virions (Table 3)27. The nitro group at the C-5 position of the thiazole ring is not essential for anti-HBV activity. It can be substituted with other electron-withdrawing groups as exemplified by compounds 22-27, while substituting the phenyl ring displayed varied effects on activity. As the nitro group is often classified as a structural alert or a toxicophore due to its potential toxicity issues79, compound 22 with 5-Cl substitution excludes this liability and maintains the same level of potency against HBV, compared to nitazoxanide, thus selected for further development27. In general, salicylanilide derivatives were less active and showed variable anti-HBV activities, depending on the exact substituents on the phenyl ring. The monosubstituted analogs 29, 30, and trisubstituted 32 showed suitable activities against HBV, while disubstituted 31 displayed moderate potency. Due to the poor oral bioavailability and limited aqueous solubility presented by niclosamide, which was close in structure and inactive against HBV up to 10 μM, further optimization efforts on this class of derivatives as anti-HBV agents were not pursued by Stachulski et al27,80.
Figure 5.

The chemical structures of salicylamide derivatives 21–32
2.4. Anti-Hepatitis C Virus Activity
Hepatitis C virus (HCV) is a hepatotropic virus with a positive-sense single-stranded RNA genome which comprises at least 6 genotypes and more than 80 subtypes belonging to the genus Hepacivirus of the family Flaviviridae81–83. HCV is transmitted mainly through direct blood-to-blood contacts between humans and can cause both acute (AHC) and chronic hepatitis C (CHC); 30% of patients infected with HCV can spontaneously clear the infection within 6 months without any treatment, while the remaining 70% of patients will develop CHC, triggering a chronic inflammatory disease process which can lead to cirrhosis and hepatocellular carcinoma if left untreated84,85. WHO estimated that in 2015, 71 million persons were living with HCV infection globally, 2.3 million had HCV and HIV co-infection, and about 400,000 persons died from long-term complications of HCV infection, mainly cirrhosis and hepatocellular carcinoma like hepatitis B73. Due to the genetic diversity of HCV and limited models for testing vaccines, developing a vaccine for hepatitis C remains a challenge, and no effective vaccine is currently available to prevent HCV infection83. On the other hand, the emergence of direct-acting antivirals such as sofosbuvir led to a revolution in HCV therapy which is usually used in combination and can cure most people of their HCV infection, regardless of HCV genotype85–87.
Nitazoxanide and tizoxanide also exhibited sub-micromolar potency against HCV genotype 1a (EC50 = 0.33 μM and 0.25 μM, respectively) and genotype 1b (EC50 = 0.21 μM and 0.15 μM, respectively)51. Nitazoxanide can activate protein kinase activated by double-stranded RNA (PKR), a key mediator of the innate immune response, which phosphorylates eukaryotic initiation factor-2α (eIF2α), leading to the inhibition of translation initiation and thereby affecting viral protein synthesis88,89. In a study using bovine viral diarrhea virus (BVDV) as a surrogate model for HCV infection, nitazoxanide was shown to reduce both cytopathic and non-cytopathic BVDV replication, likely involving phosphorylation of PKR and eIF2α90. Besides, nitazoxanide was also found to delete ATP-sensitive intracellular Ca2+ stores, leading to mild endoplasmic reticulum (ER) stress and consequent perturbation of N-linked glycosylation of BVDV structural proteins90.
In a randomized clinical trial involving 47 adults with CHC genotype 4, 7 of 23 patients receiving nitazoxanide monotherapy achieved undetectable serum HCV RNA compared to 0 of 24 receiving placebo during therapy, and 4 of 6 virological responders, who were followed up for 24 weeks after treatment, suffered a sustained virological response (SVR)91. A pilot clinical trial (96 patients with CHC genotype 4) revealed that 36-week combination therapy of nitazoxanide plus PEG-IFN α−2a with (n = 28) or without (n = 28) ribavirin following a 12-week lead-in nitazoxanide treatment increased the percentages of patients with SVR (SVR rate = 79% and 61%, respectively), compared to that of patients (n = 40, SVR rate = 50%) treated with PEG-IFN plus ribavirin92. In another pilot, clinical trial enrolling 44 patients with CHC (40 with genotype 4; 3 with genotype 1; and 1 with genotype 2), an SVR rate of 80% was observed when patients received 36-week combination therapy of nitazoxanide plus PEG-IFN α−2a using a 4-week lead-in nitazoxanide monotherapy93. In addition, coadministration of nitazoxanide with PEG-IFN α−2a and ribavirin also increased the SVR rate in CHC genotype 1 patients, showing SVR rates of 44% for naïve patients (n = 73) and 7% for patients with nonresponse to combination therapy of PEG-IFN plus ribavirin (n = 42), while the control groups (n = 37 and 21, respectively) receiving placebo plus PEG-IFN α−2a and ribavirin achieved SVR rates of 32% and 0%, respectively, in two randomized, double-blind, placebo-controlled clinical trials94,95. Nevertheless, a conflicting result was obtained in a later clinical trial enrolling 100 patients with CHC genotype 4 that patients (n = 50) receiving combination therapy of nitazoxanide, PEG-IFN α−2a, and ribavirin showed no significant improvement in the virological or biochemical response rates as compared with those (n = 50) treated with PEG-IFN α−2a plus ribavirin95. Due to the development of direct-acting antivirals, no more clinical trials were further conducted to validate the clinical benefit of nitazoxanide in HCV treatment.
Stachulski et al. evaluated the salicylamide derivatives they developed against HCV, and compounds 21 and 33–41 (Figures 5 & 6) displayed sub-micromolar to micromolar potency against HCV genotype 1a and/or 1b (Table 4)28,51. Interestingly, the 5´-Cl analog 22 was basically inactive, in contrast to its sub-micromolar potency against HBV, while its O-acetate 33 displayed EC50 values of <1 μM against both HCV genotype 1a and 1b. Contrarily, the m-fluoro derivative 40 inhibited HCV genotype 1b replication with sub-micromolar EC50 value while its O-acetate 39 performed less well. Although niclosamide was inactive against HBV, it showed potent activity against HCV genotype 1b27,28. The SAR studies showed the anti-HCV activities of these derivatives are jointly affected by the substituents on both aromatic rings and electron-withdrawing groups are preferred at the C-5 and C-4 positions of the thiazole ring.
Figure 6.

The chemical structures of salicylamide derivatives 33–41
Table 4.
Activities of salicylamides derivatives against HCV replicationa
| Compound | Genotype 1b replicon (μM) | Genotype 1a replicon (μM) | ||||||
|---|---|---|---|---|---|---|---|---|
| EC50 | EC90 | CC50 | SI | EC50 | EC90 | CC50 | SI | |
| Nitazoxanide (2) | 0.21 | 0.93 | 38.0 | 181 | 0.33 | 1.10 | 49.0 | 149 |
| Tizoxanide (3) | 0.15 | 0.81 | 15.0 | 100 | 0.25 | 1.00 | 14.0 | 56 |
| 21 | 0.36 | 6.4 | 5.0 | 14 | 0.39 | 2.30 | 12.0 | 31 |
| 22 | 10.0 | 10.0 | 15.0 | |||||
| 33 | 0.23 | 1.10 | 4.3 | 19 | 0.40 | 1.90 | 5.7 | 14 |
| 34 | 2.0 | 5.9 | >100 | >50 | ||||
| 35 | 0.59 | 23.0 | 12.0 | 20 | 1.00 | 3.80 | 14.0 | 14 |
| 36 | 4.2 | 14.0 | 45.0 | 11 | ||||
| 37 | 3.5 | 10.0 | 26.0 | 7.4 | ||||
| 38 | 2.2 | 7.0 | 16.0 | 7.3 | ||||
| 39 | 3.8 | 11.0 | >100 | 26 | ||||
| 40 | 0.13 | 0.83 | 3.4 | 26 | ||||
| 41 | 0.06 | 0.46 | 4.4 | 76 | ||||
| Niclosamide (1) | 0.16 | 0.70 | 10.0 | 62 | ||||
Antiviral activity was assessed in a 3-day assay using the stably expressed HCV replicon cell lines AVA5 (for genotype 1B) and H/FL-Neo (for genotype 1A). CC50: drug concentration at which a 2-fold lower level of neutral red dye uptake is observed relative to the average level in untreated cultures. EC50 and EC90 were determined by measuring intracellular HCV RNA concentration in triplicate and with standard deviations of ±20% of the value quoted. SI: CC50/EC50
2.5. Anti-Influenza A Virus Activity
Influenza viruses are enveloped viruses of the family Orthomyxoviridae with a negative-sense, single-stranded, segmented RNA genome including four types termed influenza viruses A, B, C, and D (IAV, IBV, ICV, and IDV), which can infect various host species such as humans, birds, and pigs96,97. IAV and IBV circulate and cause annual epidemics of seasonal influenza, characterized by a sudden onset of fever, cough, sore throat, headache, a runny nose, muscle, and joint pain, etc., which results in millions of human infections and 291,243~645,832 associated deaths estimated per year globally along with significant health and financial burdens98–100. Among them, IAVs are especially important to public health due to their potential to cause flu pandemics99. IAVs are further subtyped according to the combinations of two surface glycoproteins, haemagglutinin (HA) and neuraminidase (NA), which facilitate viral entry and release, respectively, served as the main targets for the protective antibodies induced by the influenza virus and vaccination99. At present, there have been 16 antigenically different HA subtypes and 9 antigenically different NA subtypes identified in avian species, the main animal reservoir of influenza viruses101. IAV subtypes H1N1 and H3N2 are currently circulating in humans96. When most humans lack pre-existing immunity for HA or NA proteins of the new emerged IAV, which are usually transmitted from reservoirs such as birds and pigs to humans directly or originated through reassortment of an avian influenza virus with a human influenza virus, IAV pandemics can occur and typically cause higher morbidity and mortality among humans, e.g., the 1918 H1N1 Spanish influenza and 2009 H1N1 swine-origin pandemic102–104. Vaccines are the most effective way to prevent seasonal influenza and they need to be updated each year to match the expected circulating strains due to the constantly evolving state of influenza viruses105. Currently, the NA inhibitors (NAIs), oseltamivir and zanamivir, are usually used to treat influenza virus infection in most countries. Still, the drug resistance remains a continuing challenge, highlighting an urgent need to develop a novel generation of anti-influenza agents99,106.
Nitazoxanide and tizoxanide were reported to exhibit significant in vitro activities against a series of IAV and IBV strains, including oseltamivir-resistant and amantadine-resistant strains, with sub-micromolar to micromolar EC50 values (Table 5)107–110. Nitazoxanide affects the maturation of the viral hemagglutinin, impairs its intracellular transport and insertion into the host cell plasma membrane, and thus blocks the exit of mature virions from host cells109. Nitazoxanide functions at a post-translational level, different from the currently available anti-influenza drugs, the NA-inhibitors oseltamivir, and zanamivir, which prevent the release of virus from the infected cell, and the M2 inhibitors amantadine and rimantadine, which affect virus uncoating109. A synergistic effect was observed for the combination treatment of nitazoxanide with oseltamivir or zanamivir against H1N1 A/PR/8/34 (PR8) and H1N1 A/WSN/33 (WSN)108. In addition, in the flu-stimulated peripheral blood mononuclear cells (PBMCs), nitazoxanide was found to potentiate the production of type I IFN (α and β) produced by the host’s fibroblasts, but the importance of this activity is not fully elucidated111.
Table 5.
The activity of salicylamide derivatives against influenza viruses
| Compound | Activity against influenza viruses | Ref. |
|---|---|---|
| Niclosamide (1) | H1N1 A/PR/8/34: EC50 = 0.83 μM, CC50 = 20.2 μM (A549 cells) | 115 |
| Nitazoxanide (2) | H1N1 A/PR/8/34 (PR8): EC50 = 3.3 μM, CC50 > 163 μM (MDCK cells) H1N1 A/WSN/33 (WSN): EC50 = 1.6 μM (MDCK cells) H5N9 A/Ck/It/9097/97 (A/Ck): EC50 = 3.3 μM (MDCK cells) H1N1 A/California/7/2009 (CA09): EC50 = 3.2 μM (MDCK cells) H1N1 oseltamivir-resistant A/Parma/24/2009 (OST-R): EC50 = 1.9 μM (MDCK cells) H3N2 amantadine-resistant A/Parma/06/2007 (AMD-R): EC50 = 1.0 μM (MDCK cells) H1N1 A/goose/Italy/296246/2003 (A/Tk): EC50 = 3.2 μM (MDCK cells) H7N1 A/turkey/Italy/RA5563/1999: EC50 = 1.6 μM (MDCK cells) H3N2v A/Ohio/83/2012: EC90 = 0.86 μM (MDCK cells) H3N2v A/Ohio/88/2012: EC90 = 1.09 μM (MDCK cells) |
107–109 |
| Tizoxanide (3) | H1N1 A/PR/8/34 (PR8): EC50 = 3.8 μM, CC50 > 189 μM (MDCK cells) H1N1 A/WSN/33 (WSN): EC50 = 1.9 μM (MDCK cells) H5N9 A/Ck/It/9097/97 (A/Ck): EC50 = 1.9 μM (MDCK cells) H1N1pdm09 (54 strains): EC50 = 0.1–1.23 μM (MDCK-SIAT cells) H3N2 (53 strains): EC50 = 0.22–1.14 μM (MDCK-SIAT cells) IBV Yamagata lineage (56 strains): EC50 = 0.34–0.88 μM (MDCK-SIAT cells) IBV Victoria lineage (47 strains): EC50 = 0.25–0.92 μM (MDCK-SIAT cells) NAI-resistant IAVs (10 strains): EC50 = 0.31–1.00 μM (MDCK-SIAT cells) NAI-resistant IBVs (4 strains): EC50 = 0.79–0.92 μM (MDCK-SIAT cells) |
109,110 |
| 21 | H1N1 A/PR/8/34 (PR8): EC50 = 7.2 μM, CC50 = 129 μM (MDCK cells) | 30 |
| 26 | H1N1 A/PR/8/34 (PR8): EC50 = 4.1 μM, CC50 > 204 μM (MDCK cells) | 30 |
| 33 | H1N1 A/PR/8/34 (PR8): EC50 = 3.4 μM, CC50 = 67 μM (MDCK cells) | 30 |
| 42 | H1N1 A/PR/8/34 (PR8): EC50 = 0.4 μM, CC50 > 189 μM (MDCK cells) | 30 |
| 43 | H1N1 A/PR/8/34 (PR8): EC50 = 0.29 μM, CC50 > 168 μM (MDCK cells) | 30 |
| 44 | H1N1 A/PR/8/34 (PR8): EC50 = 0.14 μM, CC50 > 141 μM (MDCK cells) | 30 |
| 45 | H1N1 A/WSN/33 (WSN): EC50 = 3.0–8.9 μM, CC50 > 297 μM (MDBK cells) | 116 |
| 46 | H1N1 A/WSN/33 (WSN): EC50 = 0.66–1.0 μM, CC50 > 332 μM (MDBK cells) | 116 |
| 47 | H1N1 A/WSN/33 (WSN): EC50 = 0.25 μM, CC50 = 110 μM (MDBK cells) | 117 |
| 48 | H1N1 A/WSN/33 (WSN): EC50 = 0.57 μM (MDBK cells) | 118 |
| 49 | H1N1 A/WSN/33 (WSN): EC50 = 0.18 μM, CC50 = 246 μM (MDCK cells) | 119,120 |
In a double-blind, randomized, placebo-controlled, phase 2b/3 trial involving 624 patients aged 12–65 years with influenza-like illness (ILI) within 48 h of symptom onset, influenza-confirmed patients receiving nitazoxanide 600 mg (n = 79) or 300 mg (n = 89) twice daily for 5 days showed shorter times to alleviation of symptoms of 95.5 h (p = 0.0084) and 109.1 h (p = 0.52, not statistically significant), respectively, than those receiving placebo (116.7 h, n = 89)50. Moreover, a significant decrease (p = 0.0006) in TCID50 viral titers was shown by the nitazoxanide 600 mg group during treatment compared to the placebo group50. Similar results were also observed for all treated participants that the nitazoxanide 600 mg group (n = 211) experienced a significantly shorter duration of symptoms (94.9 h, p = 0.0052) as compared with the control group (108.2 h, n = 212)50. Besides, in two clinical trials, compared to the placebo group, nitazoxanide treatment reduced the time of symptom resolution by >3 and 3 days in children (1–11 years of age) and adults and adolescents (≥ 12 years of age) with a viral respiratory infection, respectively112,113. However, another double-blind, placebo-controlled trial involving 257 patients with severe ILI revealed that there was no significant difference in duration of hospitalization between nitazoxanide treatment and placebo treatment in all anticipants (p = 0.56), children (p = 0.29), adults (p = 0.62), and influenza-confirmed patients (p = 0.32)114. Thus, more clinical trials are needed to evaluate the efficacy of nitazoxanide as monotherapy or in combination with NAIs, such as oseltamivir or zanamivir, for the treatment of influenza infection in otherwise healthy patients as well as those with severe illness or at risk of influenza complications.
Stachulski et al. designed and synthesized the second-generation thiazolides as effective IAV inhibitors and, among these derivatives, 42 with para hydroxyl, 43 and 44 with 4´-sulfonyl displayed outstanding potency against IAV with low sub-micromolar EC50 values, low cytotoxicity, and very high selectivity index (Figure 7 & Table 5)30. The insertion of alkylsulfone groups at the C4 position led to a significant improvement in anti-IAV activity. Niclosamide was found to inhibit influenza H1N1 A/PR/8/34 virus with an EC50 of 0.83 μM115. Researchers at Bristol Myers Squibb reported a series of N-alkyl substituted salicylamide derivatives as potent inhibitors against the H1 and H2 subtypes of IAV, represented by compounds 45-49116–120. Compound 45 (BMY-27709) was found to be active against the H1 subtype influenza viruses A/WSN/33 (EC50 = 3.0–8.9 μM) and A/PR/8/34 as well as against the H2 subtype influenza virus A/Japan/307/57, but it was inactive against H3 subtype viruses (A/HK/8/68 and A/Udorn/307/57) and influenza B/Lee/40 virus, partly limiting the significant utility of this series of compounds as anti-influenza agents121. Genetic analysis revealed that the mutation from Phe to Ser at position 110 of the HA2 subunit conferred resistance to BMY-27709 inhibition, indicating that this class of compounds likely target the hemagglutinin protein. BMY-27709 blocks viral replication at an early stage of the IAV life cycle and inhibits the virus-induced red blood cell hemolysis in a dose-dependent manner121. Further mechanistic studies suggested that BMY-27709 inhibits the HA-mediated endosomal fusion of virus and cell membrane through suppressing the essential low-pH-induced conformational change of the HA protein118,122. Removing the 4-NH2 substitution on the phenyl ring of BMY-27709 led to a significant increase in activity, as shown by compound 46116. Compounds 47–49 with diversified cycloalkyl or heterocycloalkyl groups all showed potent inhibitory activity against H1N1 A/WSN/33 virus with sub-micromolar EC50 values117–120.
Figure 7.

The chemical structures of salicylamide derivatives 42–49
2.6. Anti-Human Adenovirus Activity
Human adenoviruses (HAdVs) are non-enveloped viruses with a linear and double-stranded DNA genome of approximately 34–36 kb in size, which includes more than 100 different HAdV types classified into seven species (HAdV A-G)123–127. HAdV infections are widespread in humans, primarily occurring in young children, which can cause respiratory disease, gastroenteritis, hepatitis, conjunctivitis, myocarditis, etc. In healthy individuals, HAdV infections are usually self-limiting with mild symptoms, providing long-lasting immunity to that infected type after recovery128,129. However, in immunocompromised patients, such as solid-organ transplant (SOT) and allogenic hematopoietic stem cell transplant (allo-HSCT) recipients, who cannot easily clear the virus, HAdV infection can disseminate to multiple organ systems and cause life-threatening diseases128,130. Currently, there is no antiviral specifically approved for treating HAdV infections, and the off-label use of intravenous cidofovir remains the major therapeutic choice to treat these infections, with unsatisfactory clinical responses and significant toxicity issues125,131.
Our research team first found that niclosamide, oxyclozanide (50), and rafoxanide (51) exhibited significant anti-HAdV activity with low micromolar IC50 values (Figure 8 & Table 6)132. The mechanistic studies indicated that niclosamide and rafoxanide inhibit the accessibility of the HAdV genome to the nucleus while oxyclozanide impacts the transcription of HAdV immediate early gene E1A132. To develop highly effective and safe anti-HAdV agents, we further conducted SAR optimization studies using niclosamide as a lead compound and discovered a series of salicylamide derivatives which showed equal or improved anti-HAdV activity with nanomolar to submicromolar IC50 values and significantly increased selectivity index (SI > 100), as exemplified by compounds 52-5725,133,134. Our mechanistic assays suggested different mechanisms of HAdV inhibition for these derivatives; compounds 52, 54, and 56 inhibit later steps after DNA replication, 53 likely targets the HAdV DNA replication, and 57 stabilizes the viral capsid to prevent the uncoating and blocks HAdV particle escape from the endosome25,134. Moreover, compounds 54-56 displayed very low in vivo toxicity in the immunosuppressed Syrian hamster model, presenting maximum tolerated dose (MTD) values of 12.5 mg/kg, 150 mg/kg, and 50 mg/kg, respectively, as compared with niclosamide (MTD = 1 mg/kg), and thus were selected for further in vivo PK and efficacy evaluation for the treatment of HAdV infection133.
Figure 8.

The chemical structures of salicylamide derivatives 50–57
Table 6.
The activity of salicylamide derivatives against human adenovirusa
| Compound | Activity against human adenovirus | Ref. |
|---|---|---|
| Niclosamide (1) | HAdV-5: IC50 = 0.6 ± 0.05 μM (293β5 cells); CC50 = 22.9 ± 9.8 μM (A549 cells); SI = 38.2 HAdV-16: IC50 = 0.45 ± 0.1 μM (293β5 cells) Virus yield reduction (HAdV-5 WT): 82 ± 35 folds at 5 μM Virus yield reduction (HAdV-16 WT): 21 ± 0 folds at 5 μM Maximum tolerated dose (MTD) = 1 mg/kg |
132 |
| Oxyclozanide (50) | HAdV-5: IC50 = 2.3 ± 0.7 μM (293β5 cells); CC50 = 76.1 ± 14.4 μM (A549 cells); SI = 33.0 HAdV-16: IC50 = 1.26 ± 0.0 μM (293β5 cells) Virus yield reduction (HAdV-5 WT): 10 ± 2 folds at 25 μM Virus yield reduction (HAdV-16 WT): 26 ± 7 folds at 25 μM |
132 |
| Rafoxanide (51) | HAdV-5: IC50 = 1.3 ± 0.1 μM (293β5 cells); CC50 = 80.6 ± 34.7 μM (A549 cells); SI = 62.0 HAdV-16: IC50 = 1.38 ± 0.2 μM (293β5 cells) Virus yield reduction (HAdV-5 WT): 186 ± 58 folds at 25 μM Virus yield reduction (HAdV-16 WT): 42 ± 6 folds at 25 μM |
132 |
| 52 | HAdV-5: IC50 = 0.05 ± 0.01 μM (293β5 cells); CC50 = 10.9 ± 0.5 μM (A549 cells); SI = 218.2 Virus yield reduction (HAdV-5 WT): 1.8 ± 0.3 folds at 2 μM |
25 |
| 53 | HAdV-5: IC50 = 0.08 ± 0.00 μM (293β5 cells); CC50 = 35.0 ± 3.7 μM (A549 cells); SI = 437.5 Virus yield reduction (HAdV-5 WT): 175 ± 33 folds at 10 μM |
25 |
| 54 | HAdV-5: IC50 = 0.18 ± 0.1 μM (293β5 cells); CC50 = 120.0 ± 33.6 μM (A549 cells); SI = 666.7 Virus yield reduction (HAdV-5 WT): 989 ± 361 folds at 50 μM MTD = 12.5 mg/kg |
25 |
| 55 | HAdV-5: IC50 = 0.27 ± 0.16 μM (293β5 cells); CC50 = 156.8 ± 11.0 μM (A549 cells); SI = 580.7 Virus yield reduction (HAdV-5 WT): 52.6 ± 6.2 folds at 2.7 μM MTD = 150 mg/kg |
133 |
| 56 | HAdV-5: IC50 = 0.45 ± 0.06 μM (293β5 cells); CC50 = 200.0 ± 1.9 μM (A549 cells); SI = 444.4 Virus yield reduction (HAdV-5 WT): 213 ± 78 folds at 50 μM MTD = 50 mg/kg |
25 |
| 57 | HAdV-5: IC50 = 0.78 ± 0.01 μM (293β5 cells); CC50 = 91.2 ± 18.4 μM (A549 cells); SI = 116.9 Virus yield reduction (HAdV-5 WT): 208 ± 108 folds at 7.8 μM |
134 |
HAdV, human adenovirus; MTD, maximum tolerated dose; SI: CC50/EC50.
2.7. Anti-Respiratory Syncytial Virus Activity
RSV is an enveloped virus with a non-segmented, negative-sense, single-stranded RNA genome which consists of two major subgroups A and B, belonging to the Orthopneumovirus genus of the Pneumoviridae family135. RSV is the most common cause of acute lower respiratory infection (ALRI) in infants, resulting in airway inflammation, bronchiolitis, pneumonia, and even respiratory failure136. Virtually all children get an RSV infection by the age of two, and reinfection may occur in later life, especially during the first few years of life137,138. It was estimated that globally in 2015, 33.1 million episodes of RSV-ALRI led to about 3.2 million hospital admissions and 59,600 in-hospital deaths in children younger than 5 years; about 45% of RSV-associated hospital admissions and in-hospital deaths occurred in children younger than 6 months139. In addition, RSV infection can also be dangerous for the elderly, immunocompromised individuals, and patients with chronic medical conditions, with an increased chance of morbidity and mortality140–142. Ribavirin is only approved antiviral as an inhaled formulation for RSV infection but is associated with limited efficacy and significant safety concerns143. Palivizumab, a monoclonal antibody (mAb) licensed in 1998 for prophylactic use in high-risk infants, is not very cost-effective144,145. Therefore, an urgent need remains to develop effective, safe, and specific antiviral therapies for RSV infection.
Niclosamide inhibited RSV infection in time- and dose-dependent manners and 6 h-pretreatment of niclosamide at a sub-micromolar concentration presented the highest anti-RSV activity, displaying an EC50 value of 0.022 μM (Table 7)146. Niclosamide significantly suppressed laboratory and clinical strains of RSV-A and -B in the human bronchial epithelial BEAS-2B cell at a concentration of 0.25 μM. Niclosamide was suggested to inhibit RSV by inactivating protein kinase B (AKT), which led to an increase of apoptotic protein markers (cleaved-caspase-3 and cleaved-PARP) and early apoptosis signal (AKT-apoptosis pathway), independent from pH and mTORC1 inhibition146. Tizoxanide and thiazolide (22) were found to inhibit RSV with low micromolar EC50 values49. Our group identified a series of substituted N-(4-amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide analogs as potent RSV inhibitors26. In our assay, niclosamide exhibited a 1.47 log reduction in RSV titer at 10 μM. Compounds 58-63 (Figure 9) effectively inhibited RSV replication with reduction in virus titer ranging from 1.74 to 2.59 log, while displaying relatively low cytotoxicity. Moreover, these derivatives suppressed RSV-induced interferon regulatory factor 3 (IRF3) and NF-κB activation and decreased replication-independent and -dependent cytokines/chemokines production, highlighting their great potential to ease RSV-associated symptoms as anti-inflammatory agents26.
Table 7.
The activity of salicylamide derivatives against respiratory syncytial virus (RSV)
| Compound | Activity against respiratory syncytial virus | Ref. |
|---|---|---|
| Niclosamide (1) | EC50 = 0.022 μM, CC50 = 0.25 μM (Hep-2 cells) Virus titer reduction: 1.47 ± 0.04 log at 10 μM (A549 cells) |
26,146 |
| Tizoxanide (3) | EC50 = 1.9 μM, CC50 = 18.9 μM (Hela-ATCC) | 49 |
| 22 | EC50 = 1.6 μM, CC50 = 20.9 μM (Hela-ATCC) | 49 |
| 58 | Virus titer reduction: 1.88 ± 0.43 log at 10 μM; CC50 = 82.7 ± 1.6 μM (A549 cells) | 26 |
| 59 | Virus titer reduction: 1.82 ± 0.14 log at 10 μM; CC50 = 86.5 ± 3.7 μM (A549 cells) | 26 |
| 60 | Virus titer reduction: 2.59 ± 0.15 log at 10 μM; CC50 = 22.9 ± 0.3 μM (A549 cells) | 26 |
| 61 | Virus titer reduction: 2.13 ± 0.23 log at 10 μM; CC50 = 76.3 ± 2.3 μM (A549 cells) | 26 |
| 62 | Virus titer reduction: 1.91 ± 0.33 log at 10 μM; CC50 = 322.6 ± 45.8 μM (A549 cells) | 26 |
| 63 | Virus titer reduction: 1.74 ± 0.33 log at 10 μM; CC50 = 89.9 ± 4.7 μM (A549 cells) | 26 |
Figure 9.

The chemical structures of salicylamide derivatives 58–63
2.8. Activity Against Viral Gastroenteritis
Viral gastroenteritis, a.k.a. stomach flu, is an inflammation of the stomach and intestine with symptoms characterized by watery diarrhea, abdominal pain, nausea or vomiting, and sometimes fever147. Viral gastroenteritis can be caused by several viruses, including rotaviruses, noroviruses, sapoviruses, astroviruses, enteric adenoviruses, etc147,148. This illness is commonly transmitted through close contact with an infected person or ingesting contaminated food or water. It can affect people of all ages, causing potentially severe complications in young children and elderly adults who are susceptible to dehydration148–150. Viral gastroenteritis is typically self-limited within 2–5 days in immunocompetent individuals. However, in immunocompromised patients, this illness can last several weeks to months, even years. Currently, there is no specific antiviral therapy for viral gastroenteritis, and the primary treatment is maintaining adequate hydration148.
2.8.1. Anti-rotavirus activity
Rotavirus is a genus of non-enveloped, segmented, double-stranded RNA viruses, which includes nine species (rotavirus A-I) belonging to the family Reoviridae148,151. Rotavirus is the leading cause of severe diarrhea disease in infants and young children (≤ 5 years of age) worldwide, especially in developing countries, accounting for about 40% of diarrhea-related hospitalizations and half a million associated deaths per year148,152–155. In 2006, two new rotavirus vaccines, which were demonstrated to be safe and effective in children, were licensed and recommended by WHO as routine vaccines included in all national immunization programs, significantly decreasing the incidence of severe rotavirus gastroenteritis in many countries156–158. However, due to inherent limitations of vaccines such as availability and insufficient protection, there remains a need to develop safe and effective antiviral therapy to treat rotavirus infections.
Nitazoxanide and tizoxanide inhibit simian A/SA11-G3P and human WA-G1P rotavirus replication in a dose-dependent manner in MA-104 cells, with EC50 values of 1.9–6.5 μM (Table 8)159,160. Mechanistic studies indicated that tizoxanide alters rotavirus viroplasm formation by preventing the NSP5/NSP2 interaction, leading to a decrease of rotavirus double-stranded RNA (dsRNA) formation, not affecting viral entry and viral protein synthesis159. In a double-blind placebo-controlled trial in 50 children with severe rotavirus diarrhea, nitazoxanide treatment (7.5 mg/kg twice for 3 days) showed a short median time of illness resolution of 31 h as compared with 75 h for the control group160. In another two randomized control trials conducted in hospitalized children with acute rotavirus diarrhea, nitazoxanide treatment was reported to significantly reduce the median duration of illness and hospitalization compared to the standard treatment of diarrhea161,162. In addition, a randomized, double-blind placebo-controlled clinical trial revealed that nitazoxanide treatment significantly reduced the median time of symptom resolution to 1.5 days in out-patients (≥ 12 years of age) with rotavirus or norovirus related gastroenteritis, compared to 2.5 days for the placebo group163.
Table 8.
The activity of salicylamide derivatives against other virusesa
| Virus/strain | Cell line | IC50 or EC50 (μM) | Ref. | ||
|---|---|---|---|---|---|
| Nitazoxanide | Tizoxanide | Niclosamide | |||
| Rotavirus/simian A/SA11-G3P | MA-104 | 3.3 | 1.9 | 159,160 | |
| Rotavirus/ human WA-G1P | MA-104 | 6.5 | 3.8 | 159 | |
| Human norovirus (HuNV) | HG23 | Nitazoxanide and tizoxanide significantly inhibit HuNV replication at 1 μg/mL | 171 | ||
| Human astrovirus strain 1 (HAstV-1) | Caco-2 | 1.47 | 180 | ||
| HIV-1 | PBMCs/TZM-bl/ SupT1 | 87.8% at 10 μg/mL | 0.119 (TZM-bl)/0.102 (SupT1) | 183,188 | |
| EBOV | A549/ Vero 76 | Virus titer reduction: 2.5 log at 40 μM | 1.5 | 199,200 | |
| CHIKV | BHK-21 | 2.96 | 0.95 | 206 | |
| HRV/ 1A, 2, 14 and 16 | Hela | >162 | 0.84–1.38 | 49,115 | |
| EBV | P3HR-1 | 0.13 (RI)/ 0.092 (virion) | 213 | ||
| KSHV | BCBL1 | 0.14 (RI)/ 0.17 (virion) | 213 | ||
| HCMV/AD169, TB40, VR1814, 759rD100, PFArD100 | HFF | 3.0 – 3.7 | 214 | ||
CHIKV, chikungunya virus; EBOV, Ebola virus; HIV, human immunodeficiency virus; HRV, human rhinovirus; EBV, Epstein–Barr virus; KSHV, Kaposi’s sarcoma-associated herpesvirus; HCMV, human cytomegalovirus. RI, intracellular replication intermediate.
2.8.2. Anti-norovirus activity
Noroviruses are a genetically diverse group of nonenveloped viruses with a linear, non-segmented, single-stranded, positive-sense RNA genome which can be divided into at least seven different genogroups with numerous genotypes within each genogroup belonging to the family Caliciviridae164–166. Noroviruses affect people in all age groups and are the most common cause of acute viral gastroenteritis. It was estimated that norovirus resulted in about 699 million illnesses and 219,000 deaths per year worldwide, and children younger than 5 years are most often affected, accounting for about 43% of deaths167. In addition, due to the suppressed immune system which facilitates viral infections, noroviruses gastroenteritis can result in long-term diarrhea and other complications in immunocompromised populations, causing considerable economic burden168–170. At present, no vaccine or specific antiviral therapy is available for the treatment of norovirus infection.
Nitazoxanide and tizoxanide were reported to significantly suppress human norovirus (HuNV) replication at 1 μg/mL, in a dose-dependent manner171. These thiazolides inhibit HuNV replication, likely through activating cellular antiviral response by inducing the expression of a series of interferon-stimulated genes, especially IRF-1, independent of the JAK-STAT pathway171. Long-term treatment of nitazoxanide in combination with ribavirin showed a synergistic inhibitory effect against HuNV infection and completely cleared the replicons from host cells, supporting the potential of the combined use of nitazoxanide with ribavirin to treat norovirus gastroenteritis171.
Cases were reported that nitazoxanide successfully treated norovirus gastroenteritis in a pediatric kidney transplant recipient and a 43-year-old patient with relapsed refractory acute myelogenous leukemia (AML), respectively172,173. Several retrospective reports revealed that nitazoxanide treatment was effective to cure norovirus gastroenteritis in immunocompromised patients after receiving chemotherapy and/or hematopoietic stem cell transplantation174–176.
2.8.3. Anti-human astrovirus activity
Human astroviruses (HAstVs) are characterized by a non-segmented, positive-sense, single-stranded RNA genome within a non-enveloped icosahedral capsid which includes 8 classic serotypes in genotype MAstV 1 and other serotypes in genotypes MAstV 6, MAstV 8, and MAstV 9, belonging to the genera Mamastrovirus of the family Astroviridae177. HAstVs are a leading cause of diarrhea, especially in children, the elderly, and immunocompromised individuals, and responsible for 2 to 9% of all acute nonbacterial gastroenteritis in children worldwide177–179. So far, no vaccine or antiviral treatment exists for HAstV infections.
Nitazoxanide suppresses HAstV-1 replication in a dose-dependent manner, with an EC50 value of approximately 1.47 μM180. Nitazoxanide can significantly decrease dsRNA production and completely inhibit HAstV-1 replication when administered up to 8 h post-infection, indicating that it may act at an early stage of the viral life cycle after entry and uncoating but before structural protein synthesis180. Further studies suggested this inhibition is dependent on IFN induction. Nitazoxanide was also effective against multiple classical HAstV serotypes and clinical isolates at 2.5 μM, displaying broad anti-HAstV activity. Moreover, nitazoxanide could reduce viral shedding and clinical disease in the symptomatic turkey poult model, suggesting its potential as a clinical therapy for astrovirus disease180.
2.9. Activity Against Other Viruses
2.9.1. Anti-HIV activity
Human immunodeficiency virus (HIV) is a retrovirus that attacks the human immune system and causes acquired immunodeficiency syndrome (AIDS) if not treated, continuing to be a major global public health threat. It was estimated that 37.7 million people were living with HIV at the end of 2020, and about 680,000 people died from HIV-associated causes in 2020181. HIV infection can be managed by antiretroviral therapy (ART), which is usually composed of a combination of three or more antiretroviral drugs but cannot be cured181.
Nitazoxanide was shown to reduce HIV-1 replication in monocyte-derived macrophages (MDMs), acting at early and late stages of the HIV life cycle associated with downregulation of HIV-1 receptors CD4 and CCR5, and upregulation of two known type I IFN-induced anti-HIV host factors APOBEC 3A/3G and tetherin, which affect vial particle infectivity and release, respectively182. Nitazoxanide was also reported to inhibit HIV-1 replication in PBMCs with a percentage of 87.8% at 10 μg/mL, related to the activation of multifaceted innate immune responses and upregulation of several interferon-stimulated genes (ISGs), including those involved in cholesterol pathways, especially the cholesterol-25 hydroxylase (CH25H), an enzyme which converts cholesterol to 25-hydroxycholesterol (25HC) that broadly inhibits the growth of enveloped viruses by blocking virus-cell membrane fusion183,184. Besides, nitazoxanide is highly synergized with glucocorticoids against HIV by targeting different steps of the HIV life cycle; nitazoxanide indirectly inhibits reverse transcription while glucocorticoids affect HIV-long terminal repeat (LTR)-driven transcription185–187. Niclosamide inhibits HIV-1 replication in dose- and time-dependent manners, and pretreatment with niclosamide at 0.625 μM for 6 h displayed the most efficient inhibitory activity against HIV-1 infection, showing EC50 values of 0.119 μM in TZM-bl cells and 0.102 μM in SupT1 cells188. Niclosamide treatment downregulates mTORC1 via AMPK activation and reduces the synthesis of HIV-2 p24 protein188. Our group found that niclosamide exhibits both antiviral and anti-mycobacterial activity in an in vitro model of HIV and MtB Beijing strain co-infection using human primary monocyte-derived macrophages, supporting the further exploration of niclosamide and its derivatives as dual-acting agents in HIV/TB co-infection189.
2.9.2. Anti-Ebola virus activity
EBOV, a.k.a. Zaire ebolavirus, is a negative-sense single-stranded RNA virus that is one of six known species within the genus Ebolavirus of the family Filoviridae190–192. EBOV, and three other known ebolaviruses, can cause severe hemorrhagic fever, a.k.a. Ebola virus disease (EVD), with a high case-fatality rate varying from 25 to 90% in the past outbreaks193–195. Currently, the vaccine Ervebo and two monoclonal antibodies, Inmazeb and Ebanga, have been approved by FDA to prevent or treat Zaire ebolavirus (EBOV) infections196–198.
Nitazoxanide completely suppresses EBOV replication in A549 cells at a concentration of 20 μM. Mechanistic studies suggested that nitazoxanide could counteract EBOV VP35 protein’s ability to escape from retinoic-acid-inducible protein 1 (RIG-1) and PKR sensing of EBOV and broadly amplify the host innate immune response, including increasing RIG-1-like receptor (RLR) activation and enhancing mitochondrial antiviral signaling protein (MAVS), interferon regulatory factor 3 (IRF3), and IFN activities199. Through an HTS campaign of FDA-approved drugs, niclosamide was found to potently inhibit EBOV infection with an EC50 value of 1.5 μM200.
2.9.3. Anti-chikungunya virus activity
Chikungunya virus (CHIKV) is an arbovirus with a positive-sense single-stranded RNA genome of about 11.6 kb, which belongs to the genus Alphavirus, and the family Togaviridae201,202. The most common symptoms of CHIKV infections are fever and severe joint pain, while other symptoms may include muscle pain, headache, joint swelling, nausea, fatigue, and rash203,204. Sporadic outbreaks of chikungunya disease have been recorded in Africa, America, and Asia for decades203,205. Currently, no vaccine or specific drug is available against CHIKV.
Through an HTS using a CHIKV 26S mediated insect cell fusion inhibition assay, niclosamide, and nitazoxanide were found to possess anti-CHIKV activities with EC50 values of 0.95 μM and 2.96 μM, respectively206. Niclosamide and nitazoxanide were able to block CHIKV entry, disrupt the release of CHIKV virions, and significantly suppress cell-to-cell transmission of CHIKV infection206.
2.9.4. Anti-human rhinovirus activity
Human rhinoviruses (HRVs) are positive-sense, single-stranded RNA viruses which comprise over 100 different HRV serotypes divided into three species (HRV A−C), belonging to the genus Enterovirus in the family Picornaviridae207. HRV is the primary cause of the common cold and is associated with asthma exacerbations, severe bronchiolitis in infants and children, and fatal pneumonia in elderly and immunocompromised individuals207,208. At present, no antiviral agents were approved to treat HRV infection.
Niclosamide was reported to protect Hela cells from pH-dependent HRV serotypes 1A, 2, 14, and 16 infections with EC50 values of 0.84–1.38 μM115. Niclosamide inhibits HRV entry through neutralizing acidic membrane-bounded compartments acting as a protonophore. However, thiazolides (nitazoxanide and 19) showed no inhibitory activity against HRV serotype 2 up to 50 μg/mL49.
2.9.5. Anti-herpesvirus activity
Herpesviruses are a large group of enveloped double-stranded DNA viruses that cause a range of diseases in animals and humans. So far there are more than 130 identified herpesvirus types classified into three subfamilies, Alphaherpesvirinae, Betaherpesvirinae, and Gammaherpesvirinae209. Among them, nine herpesviruses are known to infect humans, including herpes simplex virus 1 and 2 (HSV-1 and HSV-2, also known as HHV-1 and HHV-2), varicella-zoster virus (VZV or HHV-3), Epstein-Barr virus (EBV or HHV-4), human cytomegalovirus (HCMV or HHV-5), human herpesviruses 6A (HHV-6A), HHV6B, HHV-7, and Kaposi’s sarcoma-associated herpesvirus (KSHV or HHV-8)209,210.
EBV and KSHV are two oncogenic γ-herpesviruses that can cause several types of severe malignances211,212. Niclosamide was found to inhibit viral DNA lytic replication (EC50 = 0.13 μM and 0.14 μM, respectively) and virion production (EC50 = 0.092 μM and 0.17 μM, respectively) of EBV and KSHV in lymphoma cells, through disrupting mTOR activation213. HMCV is an β-herpesvirus that is extremely widespread among humans and can cause life-threatening diseases in immunocompromised individuals such as organ transplant recipients, HIV-infected patients, and newborn infants214. Nitazoxanide inhibits the replication of different strains of HCMV, including strains resistant to the licensed anti-HCMV drugs ganciclovir or foscarnet, which target the viral DNA polymerase, displaying EC50 values of 3.0 – 3.7 μM. Nitazoxanide prevents viral DNA synthesis and early and late gene expression through inhibiting the viral transcription factor Immediate-Early 2 (IE2)-dependent transactivating activity214.
3. CONCLUSIONS AND FUTURE DIRECTIONS
Despite various potential targets or mechanisms of salicylamide derivatives against different viral infections were suggested by previous studies (Table 9), their exact mechanisms of broad antiviral activity remain to be elucidated. Given the diversity of viruses that salicylamide derivatives are effective against, the broad-spectrum antiviral activities likely attribute to targeting some commonality in the viral life cycle rather than specific viral proteins. From viral entry and replication to virion assembly and release, viruses almost require host cell factors for every step of their life cycle. Identifying the most common host cell factors and pathways used by different viruses may be a feasible direction to figure out the exact mechanism of action of salicylamide derivatives, offering the opportunity for developing broad-spectrum antivirals. Host-based antivirals usually have a high barrier to resistance compared to direct-acting antivirals but may raise a concern of potential on-target toxicity. Actually, a number of salicylamide derivatives summarized in this review were found to exhibit relatively high cytotoxicity. The combinational use of host-based antivirals and direct-acting antivirals that target different processes involved in the viral life cycle may slow the development of drug resistance and reduce the potential side effects due to the decreased effective concentration of individual drugs. Nevertheless, for those chronic viral infections such as hepatitis B and hepatitis C, the potential toxicity caused by long-term antiviral treatment may make host-based antivirals less attractive. Although newly developed salicylamides were only assessed on one or several selected viruses, varied antiviral effects were observed for different chemical modifications; salicylanilides 4–9 potently suppress both SARS-CoV-2 and ZIKV infections, same as niclosamide, while thiazolide 22, unlike nitazoxanide, only displays good inhibitory activity against HBV, basically inactive against HCV. Thus, to get meaningful SAR and develop broad-spectrum antivirals of salicylamides derivatives, it is essential to build robust salicylamide libraries and conduct systematic screening against various viruses.
Table 9.
Summary of potential targets or mechanisms of salicylamide derivatives against different viral infectionsa
| Viral mode | Summary of potential targets or mechanisms of salicylamide derivatives against different viral infections | Ref. |
|---|---|---|
| MERS-CoV | Niclosamide inhibits SKP2, increases the BECN1 level, and enhances autophagy. | 42 |
| SARS-CoV-2 | Niclosamide blocks SARS-CoV-2 spike-induced syncytia via inhibiting the activity of the calcium-activated TMEM16F. | 44 |
| ZIKV | Inhibits NS2B-NS3 interaction and results in accumulation of nonfunctional viral polyprotein precursor. | 31,64 |
| DENV | Niclosamide affects viral and host membrane fusion and uncoating, viral RNA replication, and maturation of virus particles rendering it non-infectious. | 66,67 |
| HBV | Nitazoxanide inhibits HBx-DDB1 interaction, restores Smc5 protein levels, and suppresses viral transcription and viral protein production. | 75 |
| HCV | Nitazoxanide increases PKR and eIF2α phosphorylation; it depletes ATP-sensitive intracellular Ca2+ stores and induces mild ER stress. | 89,90 |
| IAV | Nitazoxanide affects the maturation of the viral HA, impairs its intracellular trafficking and insertion into the host plasma membrane, and thus blocks the exit of mature virions from host cells. Nitazoxanide potentiates the production of type I IFN (α and β) in PBMCs. BMY-27709 inhibits the HA-mediated endosomal fusion of virus and cell membrane through suppressing the essential low-pH-induced conformational change of the HA. |
109,111,118,121,122 |
| RSV | Niclosamide reduces AKT pro-survival protein, activates cleaved-caspase-3 and cleaved-PARP, and induces early apoptosis. | 146 |
| Rotavirus | Tizoxanide alters rotavirus viroplasm formation through preventing the NSP5/NSP2 interaction and decreases rotavirus dsRNA formation. | 159 |
| Norovirus | Nitazoxanide activates cellular antiviral response by inducing the expression of a series of IFN-stimulated genes, especially IRF-1, independent of the JAK-STAT pathway. | 171 |
| HIV | Nitazoxanide downregulates HIV-1 receptors CD4 and CCR5 and upregulates anti-HIV host factors APOBEC 3A/3G and tetherin. Nitazoxanide activates multifaceted innate immune responses and upregulates several ISGs, especially CH25H. Niclosamide downregulates mTORC1 via AMPK activation and reduces the synthesis of HIV-2 p24 protein. |
182,183,188 |
| EBOV | Nitazoxanide counteracts EBOV VP35 protein’s ability to escape from RIG-1 and PKR sensing of EBOV and broadly amplify the host innate immune response, including increasing RLR activation and enhancing MAVS, IRF3, and IFN activities. | 199 |
| CHIKV | Niclosamide and nitazoxanide block viral entry, disrupt virion release, and significantly suppress cell-to-cell transmission of CHIKV infection. | 206 |
| HRV | Niclosamide inhibits HRV entry through neutralizing acidic membrane-bounded compartments acting as a protonophore. | 115 |
| EBV | Niclosamide disrupts mTOR activation. | 213 |
| HCMV | Nitazoxanide prevents viral DNA synthesis and early and late gene expression through inhibiting the viral transcription factor IE2-dependent transactivating activity. | 214 |
MERS-CoV, Middle East respiratory syndrome coronavirus; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; ZIKV, Zika virus; DENV, dengue virus; HBV, Hepatitis B virus; HCV, Hepatitis C virus; IAV, influenza A virus; RSV, respiratory syncytial virus; HIV, human immunodeficiency virus; EBOV, Ebola virus; CHIKV, chikungunya virus; HRV, human rhinovirus; EBV, Epstein–Barr virus; HCMV, human cytomegalovirus; SKP2, S-phase kinase-associated protein 2; BECN1, Benclin 1; TMEM16F, anoctamin 6; HBx, HBV regulatory protein X; DDB1, the host protein damage-specific DNA-binding protein 1; Smc5, structural maintenance of chromosomes 5; PKR, protein kinase activated by double-stranded RNA; eIF2α, eukaryotic initiation factor-2α; ER, endoplasmic reticulum; HA, haemagglutinin; IFN, interferon; PBMC, peripheral blood mononuclear cell; AKT, protein kinase B; IRF, interferon regulatory factor; JAK, Janus kinase, STAT, signal transducer and activator of transcription protein; ISG, interferon-stimulated gene; CH25H, cholesterol-25 hydroxylase; mTORC1, the mammalian target of rapamycin complex 1; AMPK, 5’ AMP-activated protein kinase; RIG-1, retinoic-acid-inducible protein 1; RLR, RIG-1-like receptor; MAVS, mitochondrial antiviral signaling protein; mTOR, the mammalian target of rapamycin; IE2, Immediate-Early 2.
To develop salicylamide derivatives as broad-spectrum antiviral agents, the PK properties and cytotoxicity of these agents are some critical features that need special attention and may be improved. The thiazolides, nitazoxanide and 33 (RM5038) are only absorbed from the gastrointestinal tract and quickly hydrolyzed into the active circulating metabolite, tizoxanide and 22 (RM4848), which are subsequently excreted from the body primarily as the O-glucuronides 64 and 65 (Figure 10)27,215,216. Besides UGT1A1-mediated glucuronidation, niclosamide also undergoes efficient CYP1A2-medaited hydroxylation, and these two metabolic reactions generate the O-glucuronide 66 and the hydroxylated metabolite 67, respectively217. Compared to niclosamide that has limited aqueous solubility and relatively low oral bioavailability (F = 10%), compound 19 (JMX0207) with a 3-NO2 group instead of the 5-Cl group displayed significantly improved PK properties31; compound 13 exhibited a very high bioavailability of F = 76% in mice and equal anti-SARS-CoV-2 activity, in contrast to niclosamide, providing significant optimization directions for salicylamide derivatives to improve their PK properties29,56. The ester or amino-acid ester prodrugs of salicylamide derivatives (e.g., 68–70) also effectively increase systemic drug exposure and extend the duration of exposure218,219. However, it should be noted that such amino-acid esters of salicylanilide derivatives could rearrange to form pseudopeptide derivatives, structurally related to compound 56220,221. In addition, it may be a fast and valuable approach to develop nano-based formulations of salicylamide derivatives to improve their PK properties and maximize their therapeutic potential, e.g., the inhaled niclosamide for COVID-19222–224. As the nitro group may induce severe toxicity and is always classified as a metabolic alert, to remove this potential liability, replacing the nitro group of niclosamide or nitazoxanide with other substituents yielded derivatives 22, 43, 54, and 55, which also displays similar or improved antiviral activities and low cytotoxicity24,79,133. Learning from the COVID-19 pandemic is vital to develop more effective, safer, and further drug-like salicylamide derivatives as broad-spectrum antiviral agents to ultimately fight the circulating viruses as well as the highly heterogeneous unknown zoonotic ones that may emerge in the future.
Figure 10.

The chemical structures of the metabolites (64–67) and prodrugs (68–70) of salicylamide derivatives
ACKNOWLEDGMENTS
This work was supported by grants AI131669, AI140726, and AI141178 from the National Institute of Allergy and Infectious Diseases (NIAID), the National Institutes of Health (Hongmin Li and Jia Zhou). Additionally, Jia Zhou is partly supported by John D. Stobo, the M.D. Distinguished Chair Endowment Fund at UTMB. Hongmin Li is additionally supported by NIH grants AI133219, AI134568, AI140406, and AI140491.
Abbreviations:
- AHB
acute hepatitis B
- AHC
acute hepatitis C
- AIDS
acquired immunodeficiency syndrome
- AKT
protein kinase B
- allo-HSCT
allogenic hematopoietic stem cell transplant
- ALRI
acute lower respiratory infection
- AML
acute myelogenous leukemia
- AMPK
5’ AMP-activated protein kinase
- ART
antiretroviral therapy
- BECN1
Benclin 1
- BVDV
bovine viral diarrhoea virus
- CH25H
cholesterol-25 hydroxylase
- CHB
chronic hepatitis B
- CHC
chronic hepatitis C
- CHIKV
chikungunya virus
- 3CLpro
3C-like protease
- CoV
coronavirus
- COVID-19
coronavirus disease 2019
- DDB1
the host protein damage-specific DNA-binding protein 1
- DENV
dengue virus
- DHF
dengue hemorrhagic fever
- DSS
dengue shock syndrome
- dsRNA
double-stranded RNA
- EBOV
Ebola virus
- EBV
Epstein–Barr virus
- eIF2α
eukaryotic initiation factor-2α
- ER
endoplasmic reticulum
- EVD
Ebola virus disease
- HA
haemagglutinin
- HAdV
human adenovirus
- HAstV
human astrovirus
- HBeAg
hepatitis B e antigen
- HBsAg
hepatitis B surface antigen
- HBV
Hepatitis B virus
- HBx
HBV regulatory protein X
- HCMV
human cytomegalovirus
- HCoV
human coronaviruses
- HCV
Hepatitis C virus
- HHV
human herpesviruses
- HIV
human immunodeficiency virus
- hNPCs
human neural progenitor cells
- HRV
human rhinovirus
- HSV
herpes simplex virus
- HTS
high-throughput screening
- HuNV
human norovirus
- IAV
influenza A virus
- IE2
Immediate-Early 2
- IFN
interferon
- ILI
influenza-like illness
- IRF
interferon regulatory factor
- ISG
interferon-stimulated gene
- JAK
Janus kinase
- JEV
Japanese encephalitis virus
- KSHV
Kaposi’s sarcoma-associated herpesvirus
- mTORC1
the mammalian target of rapamycin complex 1
- mAb
monoclonal antibody
- MAVS
mitochondrial antiviral signaling protein
- MDM
monocyte-derived macrophage
- MERS
Middle East respiratory syndrome
- MERS-CoV
Middle East respiratory syndrome coronavirus
- Mtb
Mycobacterium tuberculosis
- MTD
maximum tolerated dose
- mTOR
the mammalian target of rapamycin
- NA
neuraminidase
- NAIs
the neuraminidase inhibitor
- PBMC
peripheral blood mononuclear cell
- PFOR
pyruvate:ferredoxin oxidoreductase
- p.i.
post-infection
- PKR
protein kinase activated by double-stranded RNA
- RdRp
RNA-dependent RNA polymerase
- RIG-1
retinoic-acid-inducible protein 1
- RLR
RIG-1-like receptor
- RSV
respiratory syncytial virus
- SAR
structure-activity relationship
- SARS
severe acute respiratory syndrome
- SARS-CoV-2
severe acute respiratory syndrome coronavirus 2
- SKP2
S-phase kinase-associated protein 2
- Smc5
structural maintenance of chromosomes 5
- SOT
solid-organ transplant
- STAT
signal transducer and activator of transcription protein
- SVR
sustained virological response
- TBEV
tick-borne encephalitis virus
- TMEM16F
anoctamin 6
- VZV
varicella–zoster virus
- WHO
World Health Organization
- WNV
West Nile virus
- YFV
yellow fever virus
- ZIKV
Zika virus
- 25HC
25-hydroxycholesterol
Biographies
Jimin Xu received his B.Sc. degree in Basic Pharmacy from China Pharmaceutical University in 2009 and his Ph.D. degree from Shanghai Institute of Materia Medica, Chinese Academy of Sciences in 2014 under the supervision of Professor Fajun Nan. He worked as a Research Scientist in pharmaceutical companies for two years. Dr. Xu has pursued his postdoctoral training and is continuing his drug discovery efforts as a Research Scientist in Professor Jia Zhou’s Chemical Biology Program at the University of Texas Medical Branch (UTMB). His research interests include the rational design and chemical synthesis of small molecules as novel pharmacological probes and therapeutics for infectious diseases and human cancers.
Yu Xue obtained his Ph.D. in Medicinal Chemistry from China Pharmaceutical University (CPU) in 2018 under the supervision of Professor Liping Sun at CPU and Professor Ao Zhang at Shanghai Institute of Materia Medica, Chinese Academy of Sciences. He is currently pursuing his postdoctoral training under the supervision of Professor Jia Zhou at the Chemical Biology Program, Department of Pharmacology and Toxicology at UTMB. His research interests focus on design and synthesis of novel small molecules as chemical probes and drug candidates for infectious diseases, cancer, and other human diseases.
Andrew A. Bolinger was born and raised in beautiful Colorado. He did his undergraduate research under the supervision of Dr. Joseph L. Richards and completed his BS degree in 2013 at Colorado Mesa University. He then joined Dr. Don Coltart’s group in 2014 where he studied organic methodologies related to umpolung alkylation via azo compounds at the University of Houston. He later completed his Ph.D. with a rotation in Dr. Jeremy May’s lab. After graduating in 2019 he did a postdoc at Rutgers - Newark in the Freundlich lab before joining Dr. Jia Zhou’s group at UTMB. His current research is focused on the rational design and synthesis of novel small molecules that target GPCR receptors related to substance use disorders and other human diseases.
Jun Li received his B.A. in Pharmacy from Ocean University of China in 2012. Afterwards, he stayed in the same university, and graduated in 2015 with a M.S. in Medicinal Chemistry. He then moved to the United States and completed his Ph.D. in 2021 in Organic Chemistry at University of Houston, under the supervision of Prof. Dr. Scott Gilbertson. After his graduation, he joined Prof. Dr. Jia Zhou’s research group at the University of Texas Medical Branch as a postdoctoral research fellow. His research interests include rational drug design, synthesis, and biological evaluation of a variety of bioactive small molecules as potential pharmacological tools and novel therapeutics.
Mingxiang Zhou received his Ph.D. in organic chemistry from Texas Tech University in 2021 under the supervision of Professors Andrew M. Harned and David M. Birney. After the graduation, he joined UTMB as a postdoctoral fellow under the supervision of Professor Jia Zhou. His research interests include design and synthesis of novel small molecules as chemical probes and drug candidates for infectious diseases, cancer, and other human diseases.
Haiying Chen obtained her BSc degree in Engineering from Tianjin University (Branch) in 1995. She then worked in Tianjin Research Institute of Construction Machinery as an engineer focusing on designing and programming computer testing systems. Since 2014, she has been conducting drug discovery and development research in Dr. Jia Zhou’s Chemical Biology Program as a Senior Research Associate by utilizing the computer-assisted rational drug design of novel target-based small molecules and the understanding of key interactions and binding modes of ligands with various drug targets for the treatment of human cancers, CNS disorders, and infectious diseases.
Hongmin Li obtained his Ph.D. in Molecular Biology from Institute of Biophysics, Chinese Academy of Sciences in 1995. Then he joined as a postdoctoral affiliate in Dr. Roy Mariuzza’s lab at the Center for Advanced Research in Biotechnology, University of Maryland Biotechnology Institutes. In 2000, Dr. Li became a faculty member in the Wadsworth Center, New York State Department of Health, where he held a Research Scientist 6 position (professor-equivalent) at the Wadsworth Center at Albany. Dr. Li is currently R. Ken and Donna Coit Endowed Chair Professor at the Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona. Dr. Li authored more than 100 scientific research articles and book chapters, as well as several patents.
Jia Zhou obtained his Ph.D. in organic chemistry from Nankai University in China in 1997. He then joined the chemistry faculty with the promotion to associate professor at the same university. He pursued his postdoctoral research training in organic and bioorganic chemistry with Dr. Sidney M. Hecht at the University of Virginia in 1999. After additional postdoctoral training in medicinal chemistry and drug discovery with Dr. Alan P. Kozikowski at Georgetown University Medical Center, he conducted research in the U.S. pharmaceuticals as a Senior Principal Scientist for seven years. He is currently a tenured full professor at UTMB, leading drug discovery research programs for the treatment of CNS disorders, human cancers, inflammatory and infectious diseases. He is an author of over 230 peer-reviewed articles and seven book chapters as well as an inventor of 32 patents. Dr. Zhou is a National Academy of Inventors (NAI) Fellow, and the Editor-in-Chief of Current Topics in Medicinal Chemistry.
Footnotes
CONFLICT OF INTEREST
The authors declare no competing financial interest.
DATA AVAILABILITY STATEMENT
No experimental data were generated for this review article.
REFERENCES
- 1.Meganck RM, Baric RS. Developing therapeutic approaches for twenty-first-century emerging infectious viral diseases. Nat Med. 2021;27(3):401–410. [DOI] [PubMed] [Google Scholar]
- 2.Roychoudhury S, Das A, Sengupta P, et al. Viral pandemics of the last four decades: Pathophysiology, health impacts and perspectives. Int J Environ Res Public Health. 2020;17(24):9411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zhou P, Yang X-L, Wang X-G, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579(7798):270–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hoehl S, Rabenau H, Berger A, et al. Evidence of SARS-CoV-2 infection in returning travelers from Wuhan, China. N Engl J Med. 2020;382(13):1278–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.World Health Organization. Coronavirus disease (COVID-2019) situation reports. Weekly Epidemiological and Operational updates June 2021. https://www.who.int/emergencies/diseases/novel-coronavirus-2019/situation-reports/. Accessed June 24, 2021. [Google Scholar]
- 6.Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discovery. 2004;3(8):673–683. [DOI] [PubMed] [Google Scholar]
- 7.U.S. Food and Drug Administration (FDA). Coronavirus (COVID-19) Update: FDA Issues Emergency Use Authorization for Potential COVID-19 Treatment. May 1, 2020; https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-issues-emergency-use-authorization-potential-covid-19-treatment. Accessed June 23, 2021.
- 8.U.S. Food and Drug Administration (FDA). FDA’s approval of Veklury (remdesivir) for the treatment of COVID-19—The Science of Safety and Effectiveness. October 22, 2020 https://www.fda.gov/drugs/drug-safety-and-availability/fdas-approval-veklury-remdesivir-treatment-covid-19-science-safety-and-effectiveness. Accessed June 23, 2021.
- 9.Xu J, Xue Y, Zhou R, Shi P-Y, Li H, Zhou J. Drug repurposing approach to combating coronavirus: Potential drugs and drug targets. Med Res Rev. 2021;41(3):1375–1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sheahan TP, Sims AC, Graham RL, et al. Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci Transl Med. 2017;9(396):eaal3653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lo MK, Jordan R, Arvey A, et al. GS-5734 and its parent nucleoside analog inhibit Filo-, Pneumo-, and Paramyxoviruses. Sci Rep. 2017;7:43395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of Covid-19 — final report. N Engl J Med. 2020;383(19):1813–1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30(3):269–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sheahan TP, Sims AC, Zhou S, et al. An orally bioavailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice. Sci Transl Med. 2020;12(541):eabb5883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jayk Bernal A, Gomes da Silva MM, Musungaie DB, et al. Molnupiravir for oral treatment of Covid-19 in nonhospitalized patients. N Engl J Med. 2021;386(6):509–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.U.S. Food and Drug Administration (FDA). Coronavirus (COVID-19) Update: FDA Authorizes Additional Oral Antiviral for Treatment of COVID-19 in Certain Adults. December 23, 2021; https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-additional-oral-antiviral-treatment-covid-19-certain. Accessed August 7, 2022.
- 17.Hammond J, Leister-Tebbe H, Gardner A, et al. Oral nirmatrelvir for high-risk, nonhospitalized adults with Covid-19. N Engl J Med. 2022;386(15):1397–1408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.U.S. Food and Drug Administration (FDA). Coronavirus (COVID-19) Update: FDA Authorizes First Oral Antiviral for Treatment of COVID-19. December 22, 2021; https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-additional-oral-antiviral-treatment-covid-19-certain. Accessed August 8, 2022.
- 19.Krátký M, Vinšová J. Antiviral activity of substituted salicylanilides-a review. Mini Rev Med Chem. 2011;11(11):956–967. [DOI] [PubMed] [Google Scholar]
- 20.Xu J, Shi P-Y, Li H, Zhou J. Broad spectrum antiviral agent niclosamide and its therapeutic potential. ACS Infect Dis. 2020;6(5):909–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Rossignol JF. Nitazoxanide: a first-in-class broad-spectrum antiviral agent. Antiviral Res. 2014;110:94–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chen W, Mook RA, Premont RT, Wang J. Niclosamide: Beyond an antihelminthic drug. Cell Signal. 2018;41:89–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.de Carvalho LPS, Darby CM, Rhee KY, Nathan C. Nitazoxanide disrupts membrane potential and intrabacterial pH homeostasis of Mycobacterium tuberculosis. ACS Med Chem Lett. 2011;2(11):849–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Stachulski AV, Taujanskas J, Pate SL, et al. Therapeutic potential of nitazoxanide: An appropriate choice for repurposing versus SARS-CoV-2? ACS Infect Dis. 2021;7(6):1317–1331. [DOI] [PubMed] [Google Scholar]
- 25.Xu J, Berastegui-Cabrera J, Chen H, Pachón J, Zhou J, Sánchez-Céspedes J. Structure–activity relationship studies on diversified salicylamide derivatives as potent inhibitors of human adenovirus infection. J Med Chem. 2020;63(6):3142–3160. [DOI] [PubMed] [Google Scholar]
- 26.Xu J, Wu W, Chen H, Xue Y, Bao X, Zhou J. Substituted N-(4-amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide analogues potently inhibit respiratory syncytial virus (RSV) replication and RSV infection-associated inflammatory responses. Bioorg Med Chem. 2021;39:116157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Stachulski AV, Pidathala C, Row EC, et al. Thiazolides as novel antiviral agents. 1. Inhibition of hepatitis B virus replication. J Med Chem. 2011;54(12):4119–4132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Stachulski AV, Pidathala C, Row EC, et al. Thiazolides as novel antiviral agents. 2. Inhibition of hepatitis C virus replication. J Med Chem. 2011;54(24):8670–8680. [DOI] [PubMed] [Google Scholar]
- 29.Blake S, Shaabani N, Eubanks LM, et al. Salicylanilides reduce SARS-CoV-2 replication and suppress induction of inflammatory cytokines in a rodent model. ACS Infect Dis. 2021;7(8):2229–2237. [DOI] [PubMed] [Google Scholar]
- 30.Stachulski AV, Santoro MG, Piacentini S, et al. Second-generation nitazoxanide derivatives: thiazolides are effective inhibitors of the influenza A virus. Future Med Chem. 2018;10(8):851–862. [DOI] [PubMed] [Google Scholar]
- 31.Li Z, Xu J, Lang Y, et al. JMX0207, a niclosamide derivative with improved pharmacokinetics, suppresses Zika virus infection both in vitro and in vivo. ACS Infect Dis. 2020;6(10):2616–2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zumla A, Chan JFW, Azhar EI, Hui DSC, Yuen K-Y. Coronaviruses - drug discovery and therapeutic options. Nat Rev Drug Discov. 2016;15(5):327–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.World Health Organization. Summary of probable SARS cases with onset of illness from 1 November 2002 to 31 July 2003. December 31, 2003. https://www.who.int/csr/sars/country/table2004_04_21/en/. Accessed June 22, 2021.
- 34.World Health Organization. Middle East respiratory syndrome coronavirus (MERS-CoV). January, 2020. https://www.who.int/emergencies/mers-cov/en/. Accessed June 22, 2021.
- 35.Harvey WT, Carabelli AM, Jackson B, et al. SARS-CoV-2 variants, spike mutations and immune escape. Nat Rev Microbiol. 2021;19(7):409–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Abdool Karim SS, de Oliveira T. New SARS-CoV-2 variants — clinical, public health, and vaccine implications. N Engl J Med. 2021;384(19):1866–1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.U.S. Food and Drug Administration (FDA). Pfizer-BioNTech COVID-19 Vaccine. December 11, 2020; https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/pfizer-biontech-covid-19-vaccine. Accessed June 23, 2021.
- 38.U.S. Food and Drug Administration (FDA). Moderna COVID-19 Vaccine. December 18, 2020; https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/moderna-covid-19-vaccine. Accessed June 23, 2021.
- 39.U.S. Food and Drug Administration (FDA). Janssen COVID-19 Vaccine. February 27, 2021; https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/janssen-covid-19-vaccine. Accessed June 23, 2021.
- 40.Wen C-C, Kuo Y-H, Jan J-T, et al. Specific plant terpenoids and lignoids possess potent antiviral activities against severe acute respiratory syndrome coronavirus. J Med Chem. 2007;50(17):4087–4095. [DOI] [PubMed] [Google Scholar]
- 41.Wu C-J, Jan J-T, Chen C-M, et al. Inhibition of severe acute respiratory syndrome coronavirus replication by niclosamide. Antimicrob Agents Chemother. 2004;48(7):2693–2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gassen NC, Niemeyer D, Muth D, et al. SKP2 attenuates autophagy through Beclin1-ubiquitination and its inhibition reduces MERS-Coronavirus infection. Nat Commun. 2019;10(1):5770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jeon S, Ko M, Lee J, et al. Identification of antiviral drug candidates against SARS-CoV-2 from FDA-approved drugs. Antimicrob Agents Chemother. 2020;64(7):e00819–00820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Braga L, Ali H, Secco I, et al. Drugs that inhibit TMEM16 proteins block SARS-CoV-2 spike-induced syncytia. Nature. 2021;594(7861):88–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Andrews P, Thyssen J, Lorke D. The biology and toxicology of molluscicides, bayluscide. Pharmacol Ther. 1982;19(2):245–295. [DOI] [PubMed] [Google Scholar]
- 46.Backer V, Sjöbring U, Sonne J, et al. A randomized, double-blind, placebo-controlled phase 1 trial of inhaled and intranasal niclosamide: A broad spectrum antiviral candidate for treatment of COVID-19. Lancet Reg Health Eur. 2021;4:100084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rossignol J-F. Nitazoxanide, a new drug candidate for the treatment of Middle East respiratory syndrome coronavirus. J Infect Public Health. 2016;9(3):227–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. https://opendata.ncats.nih.gov/covid19/databrowser.
- 49.Rossignol J-F, Santoro MG. Activity of Thiazolides Against Other Respiratory Viruses than Influenza. Paper presented at: Programme & Abstract Book, Influenza and Other Respiratory Virus Infections: Advances in Clinical Management. Third isirv-Antiviral Group Conference. Abstract P54, 81; June 4 - June 6, 2014; Keio Plaza Hotel, Tokyo, Japan. [Google Scholar]
- 50.Haffizulla J, Hartman A, Hoppers M, et al. Effect of nitazoxanide in adults and adolescents with acute uncomplicated influenza: a double-blind, randomised, placebo-controlled, phase 2b/3 trial. Lancet Infect Dis. 2014;14(7):609–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Korba BE, Montero AB, Farrar K, et al. Nitazoxanide, tizoxanide and other thiazolides are potent inhibitors of hepatitis B virus and hepatitis C virus replication. Antiviral Res. 2008;77(1):56–63. [DOI] [PubMed] [Google Scholar]
- 52.Stockis A, De Bruyn S, Gengler C, Rosillon D. Nitazoxanide pharmacokinetics and tolerability in man during 7 days dosing with 0.5 g and 1 g b.i.d. Int J Clin Pharmacol Ther. 2002;40(5):221–227. [DOI] [PubMed] [Google Scholar]
- 53.Shamim K, Xu M, Hu X, et al. Application of niclosamide and analogs as small molecule inhibitors of Zika virus and SARS-CoV-2 infection. Bioorg Med Chem Lett. 2021;40:127906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Samrat SK, Xu J, Xie X, et al. Allosteric inhibitors of the main protease of SARS-CoV-2. Antivir Res. 2022;205:105381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Juang Y-P, Chou Y-T, Lin R-X, et al. Design, synthesis and biological evaluations of niclosamide analogues against SARS-CoV-2. Eur J Med Chem. 2022;235:114295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Blake S, Thanissery R, Rivera AJ, et al. Salicylanilide analog minimizes relapse of clostridioides difficile infection in mice. J Med Chem. 2020;63(13):6898–6908. [DOI] [PubMed] [Google Scholar]
- 57.Barrows NJ, Campos RK, Liao KC, et al. Biochemistry and molecular biology of flaviviruses. Chem Rev. 2018;118(8):4448–4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Molecular Virology and Control of Flaviviruses. Caister Academic Press; January 2012. [Google Scholar]
- 59.Xu J, Xie X, Ye N, et al. Design, synthesis, and biological evaluation of substituted 4,6-dihydrospiro[[1,2,3]triazolo[4,5-b]pyridine-7,3’-indoline]-2’,5(3H)-dione analogues as potent NS4B inhibitors for the treatment of dengue virus infection. J Med Chem. 2019;62(17):7941–7960. [DOI] [PubMed] [Google Scholar]
- 60.Xu J, Xie X, Chen H, et al. Design, synthesis and biological evaluation of spiropyrazolopyridone derivatives as potent dengue virus inhibitors. Bioorg Med Chem Lett. 2020;30(11):127162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Boldescu V, Behnam MAM, Vasilakis N, Klein CD. Broad-spectrum agents for flaviviral infections: dengue, Zika and beyond. Nat Rev Drug Discovery. 2017;16(8):565–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Tang H, Hammack C, Ogden SC, et al. Zika virus infects human cortical neural progenitors and attenuates their growth. Cell Stem Cell. 2016;18(5):587–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Xu M, Lee EM, Wen Z, et al. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat Med. 2016;22(10):1101–1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li Z, Brecher M, Deng Y-Q, et al. Existing drugs as broad-spectrum and potent inhibitors for Zika virus by targeting NS2B-NS3 interaction. Cell Res. 2017;27(8):1046–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shi Z, Wei J, Deng X, et al. Nitazoxanide inhibits the replication of Japanese encephalitis virus in cultured cells and in a mouse model. Virol J. 2014;11(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jung E, Nam S, Oh H, et al. Neutralization of acidic intracellular vesicles by niclosamide inhibits multiple steps of the dengue virus life cycle in vitro. Sci Rep. 2019;9(1):8682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kao JC, HuangFu WC, Tsai TT, et al. The antiparasitic drug niclosamide inhibits dengue virus infection by interfering with endosomal acidification independent of mTOR. PLoS Negl Trop Dis. 2018;12(8):e0006715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wachtman L, Mansfield K. Chapter 1 - Viral Diseases of Nonhuman Primates. In: Abee CR, Mansfield K, Tardif S, Morris T, eds. Nonhuman Primates in Biomedical Research (Second Edition). Boston: Academic Press; 2012:1–104. [Google Scholar]
- 69.Lamontagne RJ, Bagga S, Bouchard MJ. Hepatitis B virus molecular biology and pathogenesis. Hepatoma Research. 2016;2:163–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yuen M-F, Chen D-S, Dusheiko GM, et al. Hepatitis B virus infection. Nat Rev Dis Primers. 2018;4(1):18035. [DOI] [PubMed] [Google Scholar]
- 71.Iannacone M, Guidotti LG. Immunobiology and pathogenesis of hepatitis B virus infection. Nat Rev Immunol. 2021. [DOI] [PubMed] [Google Scholar]
- 72.World Health Organization. Hepatitis B. July 27, 2020; https://www.who.int/news-room/fact-sheets/detail/hepatitis-b. Accessed July 20, 2021.
- 73.Global hepatitis report 2017. World Health Organization, Geneva; 2017. [Google Scholar]
- 74.Fanning GC, Zoulim F, Hou J, Bertoletti A. Therapeutic strategies for hepatitis B virus infection: towards a cure. Nat Rev Drug Discovery. 2019;18(11):827–844. [DOI] [PubMed] [Google Scholar]
- 75.Sekiba K, Otsuka M, Ohno M, et al. Inhibition of HBV transcription from cccDNA with nitazoxanide by targeting the HBx-DDB1 interaction. Cell Mol Gastroenterol Hepatol. 2019;7(2):297–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kolozsi W, El-Gohary Y, Keeffe E, Rossignol J-F. Treatment of chronic hepatitis B (CHB) with nitazoxanide (Ntz) Alone or Ntz plus adefovir (ADV) for Two years with loss of hepatitis B e antigen (HBeAg) and hepatitis B surface antigen (HbsAg): Report of two cases: 390. Am J Gastroenterol. 2008;103:S150–S151. [Google Scholar]
- 77.Rossignol J-F, Keeffe EB. Thiazolides: a new class of drugs for the treatment of chronic hepatitis B and C. Future Microbiol. 2008;3(5):539–545. [DOI] [PubMed] [Google Scholar]
- 78.Rossignol J-F, Bréchot C A pilot clinical trial of nitazoxanide in the treatment of chronic hepatitis B. Hepatol Commun. 2019;3(6):744–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Nepali K, Lee H-Y, Liou J-P. Nitro-group-containing drugs. J Med Chem. 2019;62(6):2851–2893. [DOI] [PubMed] [Google Scholar]
- 80.Chang Y-W, Yeh T-K, Lin K-T, Chen W-C, Yao H-T. Pharmacokinetics of anti-SARS-CoV agent niclosamide and its analogs in rats. J Food Drug Anal. 2006;14(4):329–333. [Google Scholar]
- 81.Ohno O, Mizokami M, Wu RR, et al. New hepatitis C virus (HCV) genotyping system that allows for identification of HCV genotypes 1a, 1b, 2a, 2b, 3a, 3b, 4, 5a, and 6a. J Clin Microbiol. 1997;35(1):201–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Chevaliez S, Pawlotsky J-M. HCV Genome and Life Cycle. In: Tan SL, ed. Hepatitis C Viruses: Genomes and Molecular Biology. Norfolk (UK): Horizon Bioscience; 2006:5–47. [PubMed] [Google Scholar]
- 83.Bailey JR, Barnes E, Cox AL. Approaches, progress, and challenges to hepatitis C vaccine development. Gastroenterology. 2019;156(2):418–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.World Health Organization. Hepatitis C. July 27, 2020; https://www.who.int/news-room/fact-sheets/detail/hepatitis-c. Accessed July 27, 2021.
- 85.Manns MP, Buti M, Gane E, et al. Hepatitis C virus infection. Nat Rev Dis Primers. 2017;3:17006. [DOI] [PubMed] [Google Scholar]
- 86.Lawitz E, Mangia A, Wyles D, et al. Sofosbuvir for previously untreated chronic hepatitis C infection. N Engl J Med. 2013;368(20):1878–1887. [DOI] [PubMed] [Google Scholar]
- 87.Bourlière M, Gordon SC, Flamm SL, et al. Sofosbuvir, velpatasvir, and voxilaprevir for previously treated HCV infection. N Engl J Med. 2017;376(22):2134–2146. [DOI] [PubMed] [Google Scholar]
- 88.Prostko CR, Brostrom MA, Malara EM, Brostrom CO. Phosphorylation of eukaryotic initiation factor (eIF) 2 alpha and inhibition of eIF-2B in GH3 pituitary cells by perturbants of early protein processing that induce GRP78. J Biol Chem. 1992;267(24):16751–16754. [PubMed] [Google Scholar]
- 89.Elazar M, Liu M, McKenna SA, et al. The anti-hepatitis C agent nitazoxanide induces phosphorylation of eukaryotic initiation factor 2α via protein kinase activated by double-stranded RNA activation. Gastroenterology. 2009;137(5):1827–1835. [DOI] [PubMed] [Google Scholar]
- 90.Ashiru O, Howe JD, Butters TD. Nitazoxanide, an antiviral thiazolide, depletes ATP-sensitive intracellular Ca2+ stores. Virology. 2014;462–463:135–148. [DOI] [PubMed] [Google Scholar]
- 91.ROSSIGNOL JF, KABIL SM, EL-GOHARY Y, ELFERT A, KEEFFE EB. Clinical trial: randomized, double-blind, placebo-controlled study of nitazoxanide monotherapy for the treatment of patients with chronic hepatitis C genotype 4. Aliment Pharmacol Ther. 2008;28(5):574–580. [DOI] [PubMed] [Google Scholar]
- 92.Rossignol JF, Elfert A, El–Gohary Y, Keeffe EB. Improved Virologic response in chronic hepatitis C genotype 4 treated with nitazoxanide, peginterferon, and ribavirin. Gastroenterology. 2009;136(3):856–862. [DOI] [PubMed] [Google Scholar]
- 93.Rossignol JF, Elfert A, Keeffe EB. Treatment of chronic hepatitis C using a 4-week lead-in with nitazoxanide before peginterferon plus nitazoxanide. J Clin Gastroenterol. 2010;44(7):504–509. [DOI] [PubMed] [Google Scholar]
- 94.Shiffman ML, Ahmed A, Jacobson IM, Pruitt RE, Keeffe EB. 1188 phase 2 randomized, double-blind, placebo-controlled study of nitazoxanide with peginterferon alfa-2a and ribavirin in nonresponders (NR) with chronic hepatitis c genotype 1: Final report. J Hepatol. 2010;52:S461. [Google Scholar]
- 95.Bacon BR, Shiffman ML, Lim JK, Berman A, Rustgi VK, Keeffe EB. A phase II, randomized, double-blind, placebo-controlled study of nitazoxanide plus peginterferon and ribavirin in naïve patients with chronic hepatitis C genotype 1 infection: Final report. Gastroenterology. 2010;139(1):e18. [Google Scholar]
- 96.Medina RA, García-Sastre A. Influenza A viruses: new research developments. Nat Rev Microbiol. 2011;9(8):590–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Iwasaki A, Pillai PS. Innate immunity to influenza virus infection. Nat Rev Immunol. 2014;14(5):315–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Iuliano AD, Roguski KM, Chang HH, et al. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. Lancet. 2018;391(10127):1285–1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Krammer F, Smith GJD, Fouchier RAM, et al. Influenza. Nat Rev Dis Primers. 2018;4(1):3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Krammer F. The human antibody response to influenza A virus infection and vaccination. Nat Rev Immunol. 2019;19(6):383–397. [DOI] [PubMed] [Google Scholar]
- 101.Flerlage T, Boyd DF, Meliopoulos V, Thomas PG, Schultz-Cherry S. Influenza virus and SARS-CoV-2: pathogenesis and host responses in the respiratory tract. Nat Rev Microbiol. 2021;19(7):425–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.De Clercq E. Antiviral agents active against influenza A viruses. Nat Rev Drug Discov. 2006;5(12):1015–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Dunning J, Thwaites RS, Openshaw PJM. Seasonal and pandemic influenza: 100 years of progress, still much to learn. Mucosal Immunol. 2020;13(4):566–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Eisfeld AJ, Neumann G, Kawaoka Y. At the centre: influenza A virus ribonucleoproteins. Nat Rev Microbiol. 2015;13(1):28–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gerdil C. The annual production cycle for influenza vaccine. Vaccine. 2003;21(16):1776–1779. [DOI] [PubMed] [Google Scholar]
- 106.Gaisina IN, Peet NP, Cheng H, et al. Optimization of 4-aminopiperidines as inhibitors of influenza A viral entry that are synergistic with oseltamivir. J Med Chem. 2020;63(6):3120–3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sleeman K, Mishin VP, Guo Z, et al. Antiviral susceptibility of variant influenza A(H3N2)v viruses isolated in the United States from 2011 to 2013. Antimicrob Agents Chemother. 2014;58(4):2045–2051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Belardo G, Cenciarelli O, La Frazia S, Rossignol JF, Santoro MG. Synergistic effect of nitazoxanide with neuraminidase inhibitors against influenza A viruses in vitro. Antimicrob Agents Chemother. 2015;59(2):1061–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Rossignol JF, La Frazia S, Chiappa L, Ciucci A, Santoro MG. Thiazolides, a new class of anti-influenza molecules targeting viral hemagglutinin at the post-translational level. J Biol Chem. 2009;284(43):29798–29808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Tilmanis D, van Baalen C, Oh DY, Rossignol JF, Hurt AC. The susceptibility of circulating human influenza viruses to tizoxanide, the active metabolite of nitazoxanide. Antiviral Res. 2017;147:142–148. [DOI] [PubMed] [Google Scholar]
- 111.Clerici M, Trabattoni D, Pacei M, Biasin M, Rossignol J-F. The anti-infective Nitazoxanide shows strong immumodulating effects. J Immunol. 2011;186(1 Supplement):155.121. [Google Scholar]
- 112.Lopez-Chegne N, Julcamoro L, Rossignol J, Carrion M, Bardin M. Randomized, double blind, pilot study of nitazoxanide (NTZ) versus placebo (PCB) for the treatment of symptoms associated with viral respiratory infection (VRI) in adults and adolescents. Oral abstract 142. Paper presented at: Program and abstracts of the 49th Annual Meeting of the Infectious Diseases Society of America, Boston, MA 20–23 October 2011. [Google Scholar]
- 113.Lopez-Chegne N, Julcamoro L, Rossignol J, Carrion M, Bardin M. Randomized, double blind, pilot study of nitazoxanide (NTZ) versus placebo (PCB) for the treatment of symptoms associated with viral respiratory infection (VRI) in children. Oral abstract 1341. Paper presented at: Program and abstracts of the 49th Annual Meeting of the Infectious Diseases Society of America, Boston, MA 20–23 October 2011. [Google Scholar]
- 114.Gamiño-Arroyo AE, Guerrero ML, McCarthy S, et al. Efficacy and Safety of Nitazoxanide in Addition to Standard of Care for the Treatment of Severe Acute Respiratory Illness. Clin Infect Dis. 2019;69(11):1903–1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Jurgeit A, McDowell R, Moese S, Meldrum E, Schwendener R, Greber UF. Niclosamide is a proton carrier and targets acidic endosomes with broad antiviral effects. PLoS Pathog. 2012;8(10):e1002976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Yu K-L, Ruediger E, Luo G, et al. Novel quinolizidine salicylamide influenza fusion inhibitors. Bioorg Med Chem Lett. 1999;9(15):2177–2180. [DOI] [PubMed] [Google Scholar]
- 117.Combrink KD, Gulgeze HB, Yu K-L, et al. Salicylamide inhibitors of influenza virus fusion. Bioorg Med Chem Lett. 2000;10(15):1649–1652. [DOI] [PubMed] [Google Scholar]
- 118.Cianci C, Yu KL, Dischino DD, et al. pH-dependent changes in photoaffinity labeling patterns of the H1 influenza virus hemagglutinin by using an inhibitor of viral fusion. J Virol. 1999;73(3):1785–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Deshpande MS, Wei J, Luo G, et al. An approach to the identification of potent inhibitors of influenza virus fusion using parallel synthesis methodology. Bioorg Med Chem Lett. 2001;11(17):2393–2396. [DOI] [PubMed] [Google Scholar]
- 120.Yu K-L, Torri AF, Luo G, et al. Structure–activity relationships for a series of thiobenzamide influenza fusion inhibitors derived from 1,3,3-Trimethyl-5-hydroxy-cyclohexylmethylamine. Bioorg Med Chem Lett. 2002;12(23):3379–3382. [DOI] [PubMed] [Google Scholar]
- 121.Luo G, Colonno R, Krystal M. Characterization of a hemagglutinin-specific inhibitor of influenza A virus. Virology. 1996;226(1):66–76. [DOI] [PubMed] [Google Scholar]
- 122.Luo G, Torri A, Harte WE, et al. Molecular mechanism underlying the action of a novel fusion inhibitor of influenza A virus. J Virol. 1997;71(5):4062–4070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Robinson CM, Singh G, Lee JY, et al. Molecular evolution of human adenoviruses. Sci Rep. 2013;3:1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Hage E, Gerd Liebert U, Bergs S, Ganzenmueller T, Heim A. Human mastadenovirus type 70: a novel, multiple recombinant species D mastadenovirus isolated from diarrhoeal faeces of a haematopoietic stem cell transplantation recipient. J Gen Virol. 2015;96(9):2734–2742. [DOI] [PubMed] [Google Scholar]
- 125.Wold WSM, Tollefson AE, Ying B, Spencer JF, Toth K. Drug development against human adenoviruses and its advancement by Syrian hamster models. FEMS Microbiol Rev. 2019;43(4):380–388. [DOI] [PubMed] [Google Scholar]
- 126.Wold WS, Ison MG. Adenovirus. In: Fields Virology 6th Edition. Lippincott, Williams & Wilkins; 2013:1732–1767. [Google Scholar]
- 127.HAdV Working Group. Human Adenovirus Working Group. http://hadvwg.gmu.edu/. Accessed August 20, 2021.
- 128.Lion T. Adenovirus infections in immunocompetent and immunocompromised patients. Clin Microbiol Rev. 2014;27(3):441–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Echavarria M. Adenoviruses in immunocompromised hosts. Clin Microbiol Rev. 2008;21(4):704–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Martínez-Aguado P, Serna-Gallego A, Marrugal-Lorenzo JA, Gómez-Marín I, Sánchez-Céspedes J. Antiadenovirus drug discovery: potential targets and evaluation methodologies. Drug Discov Today. 2015;20(10):1235–1242. [DOI] [PubMed] [Google Scholar]
- 131.Lenaerts L, De Clercq E, Naesens L. Clinical features and treatment of adenovirus infections. Rev Med Virol. 2008;18(6):357–374. [DOI] [PubMed] [Google Scholar]
- 132.Marrugal-Lorenzo JA, Serna-Gallego A, Berastegui-Cabrera J, Pachón J, Sánchez-Céspedes J. Repositioning salicylanilide anthelmintic drugs to treat adenovirus infections. Sci Rep. 2019;9(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Xu J, Berastegui-Cabrera J, Ye N, et al. Discovery of novel substituted N-(4-Amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide analogues as potent human adenovirus inhibitors. J Med Chem. 2020;63(21):12830–12852. [DOI] [PubMed] [Google Scholar]
- 134.Xu J, Berastegui-Cabrera J, Carretero-Ledesma M, et al. Discovery of a small molecule inhibitor of human adenovirus capable of preventing escape from the endosome. Int J Mol Sci. 2021;22(4):1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Afonso CL, Amarasinghe GK, Bányai K, et al. Taxonomy of the order Mononegavirales: update 2016. Arch Virol. 2016;161(8):2351–2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Cockerill GS, Good JAD, Mathews N. State of the art in respiratory syncytial virus drug discovery and development. J Med Chem. 2019;62(7):3206–3227. [DOI] [PubMed] [Google Scholar]
- 137.Henderson FW, Collier AM, Clyde WA Jr., Denny FW. Respiratory-syncytial-virus infections, reinfections and immunity. A prospective, longitudinal study in young children. N Engl J Med. 1979;300(10):530–534. [DOI] [PubMed] [Google Scholar]
- 138.Glezen WP, Taber LH, Frank AL, Kasel JA. Risk of primary infection and reinfection with respiratory syncytial virus. Am J Dis Child. 1986;140(6):543–546. [DOI] [PubMed] [Google Scholar]
- 139.Shi T, McAllister DA, O’Brien KL, et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: a systematic review and modelling study. Lancet. 2017;390(10098):946–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Jensen TO, Stelzer-Braid S, Willenborg C, et al. Outbreak of respiratory syncytial virus (RSV) infection in immunocompromised adults on a hematology ward. J Med Virol. 2016;88(10):1827–1831. [DOI] [PubMed] [Google Scholar]
- 141.Falsey AR. Respiratory syncytial virus infection in adults. Semin Respir Crit Care Med. 2007;28(2):171–181. [DOI] [PubMed] [Google Scholar]
- 142.Falsey AR, Hennessey PA, Formica MA, Cox C, Walsh EE. Respiratory syncytial virus infection in elderly and high-risk adults. N Engl J Med. 2005;352(17):1749–1759. [DOI] [PubMed] [Google Scholar]
- 143.Ventre K, Randolph AG. Ribavirin for respiratory syncytial virus infection of the lower respiratory tract in infants and young children. Cochrane Database Syst Rev. 2007(1):CD000181. [DOI] [PubMed] [Google Scholar]
- 144.Resch B. Product review on the monoclonal antibody palivizumab for prevention of respiratory syncytial virus infection. Hum Vaccines Immunother. 2017;13(9):2138–2149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Committee On Infectious Diseases and Bronchiolitis Guidelines Committee. Updated guidance for palivizumab prophylaxis among infants and young children at increased risk of hospitalization for respiratory syncytial virus infection. Pediatrics. 2014;134(2):e620. [DOI] [PubMed] [Google Scholar]
- 146.Niyomdecha N, Suptawiwat O, Boonarkart C, Thitithanyanont A, Auewarakul P. Repurposing of antiparasitic niclosamide to inhibit respiratory syncytial virus (RSV) replication. Virus Res. 2021;295:198277. [DOI] [PubMed] [Google Scholar]
- 147.Blacklow NR, Greenberg HB. Viral Gastroenteritis. N Engl J Med. 1991;325(4):252–264. [DOI] [PubMed] [Google Scholar]
- 148.Bányai K, Estes MK, Martella V, Parashar UD. Viral gastroenteritis. Lancet. 2018;392(10142):175–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Desselberger U. Viral gastroenteritis. Medicine (Abingdon). 2017;45(11):690–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Fong TT, Lipp EK. Enteric viruses of humans and animals in aquatic environments: health risks, detection, and potential water quality assessment tools. Microbiol Mol Biol Rev. 2005;69(2):357–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Suzuki H. Rotavirus replication: Gaps of knowledge on virus entry and morphogenesis. Tohoku J Exp Med. 2019;248(4):285–296. [DOI] [PubMed] [Google Scholar]
- 152.Dennehy PH. Rotavirus infection: A disease of the past? Infect Dis Clin North Am. 2015;29(4):617–635. [DOI] [PubMed] [Google Scholar]
- 153.Tate JE, Burton AH, Boschi-Pinto C, Steele AD, Duque J, Parashar UD. 2008 estimate of worldwide rotavirus-associated mortality in children younger than 5 years before the introduction of universal rotavirus vaccination programmes: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12(2):136–141. [DOI] [PubMed] [Google Scholar]
- 154.Rheingans R, Heylen J, Giaquinto C. Economics of rotavirus gastroenteritis and vaccination in europe: What makes sense? Pediatr Infect Dis J. 2006;25:S48–S55. [DOI] [PubMed] [Google Scholar]
- 155.Lanata CF, Fischer-Walker CL, Olascoaga AC, Torres CX, Aryee MJ, Black RE. Global causes of diarrheal disease mortality in children <5 years of age: a systematic review. PLoS ONE. 2013;8(9):e72788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Dennehy PH. Rotavirus vaccines: an overview. Clin Microbiol Rev. 2008;21(1):198–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Diggle L. Rotavirus diarrhoea and future prospects for prevention. Br J Nurs. 2007;16(16):970–974. [DOI] [PubMed] [Google Scholar]
- 158.Jiang V, Jiang B, Tate J, Parashar UD, Patel MM. Performance of rotavirus vaccines in developed and developing countries. Hum Vaccin. 2010;6(7):532–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.La Frazia S, Ciucci A, Arnoldi F, et al. Thiazolides, a new class of antiviral agents effective against rotavirus infection, target viral morphogenesis, inhibiting viroplasm formation. J Virol. 2013;87(20):11096–11106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Rossignol J-F, Abu-Zekry M, Hussein A, Santoro MG. Effect of nitazoxanide for treatment of severe rotavirus diarrhoea: randomised double-blind placebo-controlled trial. Lancet. 2006;368(9530):124–129. [DOI] [PubMed] [Google Scholar]
- 161.Teran CG, Teran-Escalera CN, Villarroel P. Nitazoxanide vs. probiotics for the treatment of acute rotavirus diarrhea in children: a randomized, single-blind, controlled trial in Bolivian children. Int J Infect Dis. 2009;13(4):518–523. [DOI] [PubMed] [Google Scholar]
- 162.Mahapatro S, Mahilary N, Satapathy AK, Das RR. Nitazoxanide in acute rotavirus diarrhea: A randomized control trial from a developing country. J Trop Med. 2017;2017:7942515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Rossignol JF, El-Gohary YM. Nitazoxanide in the treatment of viral gastroenteritis: a randomized double-blind placebo-controlled clinical trial. Aliment Pharmacol Ther. 2006;24(10):1423–1430. [DOI] [PubMed] [Google Scholar]
- 164.Thorne LG, Goodfellow IG. Norovirus gene expression and replication. J Gen Virol. 2014;95(2):278–291. [DOI] [PubMed] [Google Scholar]
- 165.Hoa Tran TN, Trainor E, Nakagomi T, Cunliffe NA, Nakagomi O. Molecular epidemiology of noroviruses associated with acute sporadic gastroenteritis in children: Global distribution of genogroups, genotypes and GII.4 variants. J Clin Virol. 2013;56(3):269–277. [DOI] [PubMed] [Google Scholar]
- 166.Atmar RL, Baehner F, Cramer JP, et al. Persistence of antibodies to 2 virus-like Particle norovirus vaccine candidate formulations in healthy adults: 1-year follow-up with memory probe vaccination. J Infect Dis. 2019;220(4):603–614. [DOI] [PubMed] [Google Scholar]
- 167.Bartsch SM, Lopman BA, Ozawa S, Hall AJ, Lee BY. Global economic burden of norovirus gastroenteritis. PLoS ONE. 2016;11(4):e0151219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Ye X, Van JN, Munoz FM, et al. Noroviruses as a cause of diarrhea in immunocompromised pediatric hematopoietic stem cell and solid organ transplant recipients. Am J Transplant. 2015;15(7):1874–1881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Kaufman SS, Chatterjee NK, Fuschino ME, et al. Characteristics of human calicivirus enteritis in intestinal transplant recipients. J Pediatr Gastroenterol Nutr. 2005;40(3):328–333. [DOI] [PubMed] [Google Scholar]
- 170.Schwartz S, Vergoulidou M, Schreier E, et al. Norovirus gastroenteritis causes severe and lethal complications after chemotherapy and hematopoietic stem cell transplantation. Blood. 2011;117(22):5850–5856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Dang W, Xu L, Ma B, et al. Nitazoxanide inhibits human norovirus replication and synergizes with ribavirin by activation of cellular antiviral response. Antimicrob Agents Chemother. 2018;62(11):e00707–00718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Haubrich K, Gantt S, Blydt-Hansen T. Successful treatment of chronic norovirus gastroenteritis with nitazoxanide in a pediatric kidney transplant recipient. Pediatr Transplant. 2018;22(4):e13186. [DOI] [PubMed] [Google Scholar]
- 173.Siddiq DM, Koo HL, Adachi JA, Viola GM. Norovirus gastroenteritis successfully treated with nitazoxanide. J Infect. 2011;63(5):394–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Morris J, Brown W, Morris CL. Nitazoxanide is effective therapy for norovirus gastroenteritis after chemotherapy and hematopoietic stem cell transplantation (HSCT). Blood. 2013;122(21):4581–4581. [Google Scholar]
- 175.Morris J, Morris C. Nitazoxanide is effective therapy for norovirus gastroenteritis after chemotherapy and hematopoietic stem cell transplantation (HSCT). Biol Blood Marrow Transplant. 2015;21(2):S255–S256. [Google Scholar]
- 176.Gorgeis J, Sizemore C, Bashey A, et al. Nitazoxanide is effective therapy for norovirus gastroenteritis after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2017;23(3):S197–S198. [Google Scholar]
- 177.Bosch A, Pintó RM, Guix S. Human astroviruses. Clin Microbiol Rev. 2014;27(4):1048–1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Bosch Albert, Guis Susana, Pintó RM. Astrovirus Research: Essential Ideas, Everyday Impacts, Future Directions. In: Epidemiology of human astroviruses. New York, NY: Springer; 2013:1–18. [Google Scholar]
- 179.Tseng WC, Wu FT, Hsiung CA, et al. Astrovirus gastroenteritis in hospitalized children of less than 5 years of age in Taiwan, 2009. J Microbiol Immunol Infect. 2012;45(4):311–317. [DOI] [PubMed] [Google Scholar]
- 180.Hargest V, Sharp B, Livingston B, Cortez V, Schultz-Cherry S. Astrovirus replication is inhibited by nitazoxanide in vitro and in vivo. J Virol. 2020;94(5):e01706–01719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.World Health Organization. HIV/AIDS. July 14, 2021; https://www.who.int/news-room/fact-sheets/detail/hiv-aids. Accessed September 8, 2021.
- 182.Gekonge B, Bardin MC, Montaner LJ. Short communication: Nitazoxanide inhibits HIV viral replication in monocyte-derived macrophages. AIDS Res Hum Retroviruses. 2015;31(2):237–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Trabattoni D, Gnudi F, Ibba SV, et al. Thiazolides elicit anti-viral innate immunity and reduce HIV replication. Sci Rep. 2016;6:27148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Liu S-Y, Aliyari R, Chikere K, et al. Interferon-inducible cholesterol-25-hydroxylase broadly inhibits viral entry by production of 25-hydroxycholesterol. Immunity. 2013;38(1):92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Tan X, Hu L, Luquette LJ 3rd, et al. Systematic identification of synergistic drug pairs targeting HIV. Nat Biotechnol. 2012;30(11):1125–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Russo FO, Patel PC, Ventura AM, Pereira CA. HIV-1 long terminal repeat modulation by glucocorticoids in monocytic and lymphocytic cell lines. Virus Res. 1999;64(1):87–94. [DOI] [PubMed] [Google Scholar]
- 187.Hanley TM, Viglianti GA. Nuclear receptor signaling inhibits HIV-1 replication in macrophages through multiple trans-repression mechanisms. J Virol. 2011;85(20):10834–10850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Niyomdecha N, Suptawiwat O, Boonarkart C, Jitobaom K, Auewarakul P. Inhibition of human immunodeficiency virus type 1 by niclosamide through mTORC1 inhibition. Heliyon. 2020;6(6):e04050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Fan X, Xu J, Files M, et al. Dual activity of niclosamide to suppress replication of integrated HIV-1 and Mycobacterium tuberculosis (Beijing). Tuberculosis (Edinb). 2019;116s:S28–S33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Kuhn JH, Becker S, Ebihara H, et al. Proposal for a revised taxonomy of the family Filoviridae: classification, names of taxa and viruses, and virus abbreviations. Arch Virol. 2010;155(12):2083–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Nanbo A, Watanabe S, Halfmann P, Kawaoka Y. The spatio-temporal distribution dynamics of Ebola virus proteins and RNA in infected cells. Sci Rep. 2013;3:1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Jacob ST, Crozier I, Fischer WA, et al. Ebola virus disease. Nat Rev Dis Primers. 2020;6(1):13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.World Health Organization. Ebola virus disease. February 23, 2021; https://www.who.int/en/news-room/fact-sheets/detail/ebola-virus-disease. Accessed September 22, 2021.
- 194.Holmes EC, Dudas G, Rambaut A, Andersen KG. The evolution of Ebola virus: Insights from the 2013–2016 epidemic. Nature. 2016;538(7624):193–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Ilunga Kalenga O, Moeti M, Sparrow A, Nguyen V-K, Lucey D, Ghebreyesus TA. The ongoing Ebola epidemic in the Democratic Republic of Congo, 2018–2019. N Engl J Med. 2019;381(4):373–383. [DOI] [PubMed] [Google Scholar]
- 196.U.S. Food and Drug Administration (FDA). First FDA-approved vaccine for the prevention of Ebola virus disease, marking a critical milestone in public health preparedness and response. December 19, 2019; https://www.fda.gov/news-events/press-announcements/first-fda-approved-vaccine-prevention-ebola-virus-disease-marking-critical-milestone-public-health. Accessed September 22, 2021.
- 197.U.S. Food and Drug Administration (FDA). FDA Approves Treatment for Ebola Virus. December 21, 2020; https://www.fda.gov/drugs/drug-safety-and-availability/fda-approves-treatment-ebola-virus. Accessed September 22, 2021.
- 198.U.S. Food and Drug Administration (FDA). FDA Approves First Treatment for Ebola Virus. October 15, 2020; https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-ebola-virus. Accessed September 22, 2021.
- 199.Jasenosky LD, Cadena C, Mire CE, et al. The FDA-approved oral drug nitazoxanide amplifies host antiviral responses and inhibits Ebola virus. iScience. 2019;19:1279–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Madrid PB, Panchal RG, Warren TK, et al. Evaluation of Ebola virus inhibitors for drug repurposing. ACS Infect Dis. 2015;1(7):317–326. [DOI] [PubMed] [Google Scholar]
- 201.Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines. 2012;11(9):1087–1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Burt FJ, Rolph MS, Rulli NE, Mahalingam S, Heise MT. Chikungunya: a re-emerging virus. Lancet. 2012;379(9816):662–671. [DOI] [PubMed] [Google Scholar]
- 203.World Health Organization. Chikungunya. September 15, 2020; https://www.who.int/news-room/fact-sheets/detail/chikungunya. Accessed September 23, 2021.
- 204.Burt FJ, Chen W, Miner JJ, et al. Chikungunya virus: an update on the biology and pathogenesis of this emerging pathogen. Lancet Infect Dis. 2017;17(4):e107–117. [DOI] [PubMed] [Google Scholar]
- 205.Schwartz O, Albert ML. Biology and pathogenesis of chikungunya virus. Nat Rev Microbiol. 2010;8(7):491–500. [DOI] [PubMed] [Google Scholar]
- 206.Wang YM, Lu JW, Lin CC, et al. Antiviral activities of niclosamide and nitazoxanide against chikungunya virus entry and transmission. Antiviral Res. 2016;135:81–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Jacobs SE, Lamson DM, George KS, Walsh TJ. Human rhinoviruses. Clin Microbiol Rev. 2013;26(1):135–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Turner RB. Rhinovirus: More than just a common cold virus. J Infect Dis. 2007;195(6):765–766. [DOI] [PubMed] [Google Scholar]
- 209.Damania B. Oncogenic γ-herpesviruses: comparison of viral proteins involved in tumorigenesis. Nat Rev Microbiol. 2004;2(8):656–668. [DOI] [PubMed] [Google Scholar]
- 210.Connolly SA, Jardetzky TS, Longnecker R. The structural basis of herpesvirus entry. Nat Rev Microbiol. 2021;19(2):110–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Ganem D. KSHV and the pathogenesis of Kaposi sarcoma: listening to human biology and medicine. J Clin Investig. 2010;120(4):939–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Kutok JL, Wang F. Spectrum of Epstein-Barr virus–associated diseases. Annu Rev Pathol: Mech Dis. 2006;1(1):375–404. [DOI] [PubMed] [Google Scholar]
- 213.Huang L, Yang M, Yuan Y, Li X, Kuang E. Niclosamide inhibits lytic replication of Epstein-Barr virus by disrupting mTOR activation. Antivir Res. 2017;138:68–78. [DOI] [PubMed] [Google Scholar]
- 214.Mercorelli B, Luganini A, Nannetti G, et al. Drug Repurposing Approach Identifies Inhibitors of the Prototypic Viral Transcription Factor IE2 that Block Human Cytomegalovirus Replication. Cell Chem Biol. 2016;23(3):340–351. [DOI] [PubMed] [Google Scholar]
- 215.Broekhuysen J, Stockis A, Lins RL, De Graeve J, Rossignol JF. Nitazoxanide: pharmacokinetics and metabolism in man. Int J Clin Pharmacol Ther. 2000;38(8):387–394. [DOI] [PubMed] [Google Scholar]
- 216.Stachulski AV, Rossignol JF, Pate S, et al. Synthesis, antiviral activity, preliminary pharmacokinetics and structural parameters of thiazolide amine salts. Future Med Chem. 2021;13(20):1731–1741. [DOI] [PubMed] [Google Scholar]
- 217.Lu D, Ma Z, Zhang T, Zhang X, Wu B. Metabolism of the anthelmintic drug niclosamide by cytochrome P450 enzymes and UDP-glucuronosyltransferases: metabolite elucidation and main contributions from CYP1A2 and UGT1A1. Xenobiotica. 2016;46(1):1–13. [DOI] [PubMed] [Google Scholar]
- 218.Stachulski AV, Swift K, Cooper M, et al. Synthesis and pre-clinical studies of new amino-acid ester thiazolide prodrugs. Eur J Med Chem. 2017;126:154–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Mook RA, Wang J, Ren X-R, et al. Structure–activity studies of Wnt/β-catenin inhibition in the Niclosamide chemotype: Identification of derivatives with improved drug exposure. Bioorg Med Chem. 2015;23(17):5829–5838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Vinšová J, Imramovský A, Krátký M, et al. An unprecedented rearrangement of salicylanilide derivatives: imidazolinone intermediate formation. Tetrahedron Lett. 2010;51(1):23–26. [Google Scholar]
- 221.Pauk K, Zadražilová I, Imramovský A, et al. New derivatives of salicylamides: Preparation and antimicrobial activity against various bacterial species. Bioorg Med Chem. 2013;21(21):6574–6581. [DOI] [PubMed] [Google Scholar]
- 222.Brunaugh AD, Seo H, Warnken Z, Ding L, Seo SH, Smyth HDC. Development and evaluation of inhalable composite niclosamide-lysozyme particles: A broad-spectrum, patient-adaptable treatment for coronavirus infections and sequalae. PLoS ONE. 2021;16(2):e0246803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Jara MO, Warnken ZN, Sahakijpijarn S, et al. Niclosamide inhalation powder made by thin-film freezing: Multi-dose tolerability and exposure in rats and pharmacokinetics in hamsters. Int J Pharm. 2021;603:120701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Kunzelmann K. Getting hands on a drug for COVID-19: Inhaled and intranasal niclosamide. Lancet Reg Health Eur. 2021;4:100094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Cao R-Y, Xu Y-F, Zhang T-H, et al. Pediatric drug nitazoxanide: A potential choice for control of Zika. Open Forum Infect Dis. 2017;4(1):ofx009. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No experimental data were generated for this review article.
