Next Article in Journal
Construction of Porous Starch-Based Hydrogel via Regulating the Ratio of Amylopectin/Amylose for Enhanced Water-Retention
Previous Article in Journal
Promising Antimicrobial Properties of Bioactive Compounds from Different Honeybee Products
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Niclosamide Is Active In Vitro against Mycetoma Pathogens

by
Abdelhalim B. Mahmoud
1,2,3,†,
Shereen Abd Algaffar
4,†,
Wendy van de Sande
5,
Sami Khalid
3,4,
Marcel Kaiser
1,2 and
Pascal Mäser
1,2,*
1
Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
2
Faculty of Science, University of Basel, 4001 Basel, Switzerland
3
Faculty of Pharmacy, University of Science and Technology, Omdurman 14411, Sudan
4
Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan
5
Erasmus Medical Center, Department of Medical Microbiology and Infectious Diseases, 3000 Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2021, 26(13), 4005; https://doi.org/10.3390/molecules26134005
Submission received: 30 May 2021 / Revised: 21 June 2021 / Accepted: 24 June 2021 / Published: 30 June 2021

Abstract

:
Redox-active drugs are the mainstay of parasite chemotherapy. To assess their repurposing potential for eumycetoma, we have tested a set of nitroheterocycles and peroxides in vitro against two isolates of Madurella mycetomatis, the main causative agent of eumycetoma in Sudan. All the tested compounds were inactive except for niclosamide, which had minimal inhibitory concentrations of around 1 µg/mL. Further tests with niclosamide and niclosamide ethanolamine demonstrated in vitro activity not only against M. mycetomatis but also against Actinomadura spp., causative agents of actinomycetoma, with minimal inhibitory concentrations below 1 µg/mL. The experimental compound MMV665807, a related salicylanilide without a nitro group, was as active as niclosamide, indicating that the antimycetomal action of niclosamide is independent of its redox chemistry (which is in agreement with the complete lack of activity in all other nitroheterocyclic drugs tested). Based on these results, we propose to further evaluate the salicylanilides, niclosamide in particular, as drug repurposing candidates for mycetoma.

1. Introduction

First described from Madurai as Madura foot, the disease eumycetoma is endemic in the ‘mycetoma belt’ from India to the Middle East and across the Sahel [1,2]. The highest prevalence today is in Sudan [1,2]. Eumycetoma is a chronic subcutaneous mycosis that slowly spreads, starting from an initial lesion at the site of inoculation into the skin and deeper tissues, ultimately destroying muscles, tendons, and bones. The leg and foot are most often affected, likely due to inoculation via thorn pricks. While eumycetoma can be caused by various fungi of the orders Sordariales and Pleosporales [3], the majority of cases in Sudan are due to Madurella mycetomatis [2]. Eumycetoma is a debilitating, disfiguring, and stigmatizing disease. It is also an enigmatic disease in the light of the many open questions regarding its epidemiology, pathogenesis, and the biology of the causative agents [2,4].
In contrast to actinomycetoma, which is caused by filamentous bacteria and can be treated with antibiotics, there is no satisfactory treatment for eumycetoma. The current therapy consists of a combination of surgery and long-term chemotherapy with antifungal azoles, such as itraconazole [2]. However, the cure rates are low, and amputation of the affected limb may be the only measure to stop the flesh-eating fungus [5]. Given the urgent need for better drugs and the fact that eumycetoma is a neglected disease affecting neglected patients, drug repurposing suggests itself as a fast and cost-effective way towards new antimycetomal agents [6,7].
In this article, we pursued this strategy by testing a small set of redox-active parasiticides and antibiotics for their in vitro activity against M. mycetomatis. Redox-active molecules are the mainstay of current parasite chemotherapy [8]. The artemisinins for malaria, benznidazole and nifurtimox for Chagas’ disease, fexinidazole for sleeping sickness, and metronidazole for intestinal protozoa: all of these are prodrugs that will produce cytotoxic radicals once they are activated by the parasite’s metabolism. The activation occurs by chemical reduction, i.e., the acquisition of an electron [9]. Since saprophytic fungi dwell in hypoxic environments and may possess reducing agents of low redox potential, we speculated that Madurella, too, might be susceptible to prodrugs that are activated by electron transfer.

2. Results

A selection of redox-active drugs and experimental compounds was evaluated for their in vitro activity against two different isolates of M. mycetomatis, SO1 and CBS131320. It included six nitroimidazoles (fexinidazole, metronidazole, secnidazole, plus three experimental compounds [10,11]), three nitrofurans (nifurtimox, nifuroxacide, nitrofurantoine), a salicylanilide (niclosamide), and five peroxides (artemisinin, dihydroartemisinin, artesunate, artemether, and the experimental ozonide OZ78). Itraconazole was included as a reference. All molecules were tested in serial dilution against M. mycetomatis cultures [12]. The minimal inhibitory concentration (MIC) was defined as the lowest concentration that, after 7 days of incubation, had inhibited the growth by at least 80% compared to untreated cultures. Itraconazole had a MIC of 0.13 µg/mL and 0.25 µg/mL against SO1 and CBS131320, respectively (Table 1). The tested peroxides were inactive, which is in agreement with the reported lack of activity of artemisinin [13]. Moreover, all the nitroimidazoles and nitrofurans were inactive. The one notable exception was niclosamide (Figure 1), which had a MIC around 1 µg/mL (Table 1).
This interesting finding was followed up by testing the three compounds niclosamide, niclosamide-ethanolamine (NEN), and MMV665807 against the two isolates of M. mycetomatis and two species of Actinomadura, A. madurae and A. syzygii, causative agents of actinomycetoma. NEN is the ethanolamine salt of niclosamide [14]. MMV665807 (Figure 1) is a salicylanilide from the Medicines for Malaria Venture’s malaria box [15] that has shown antibacterial [16], antiprotozoal [17,18], and anticestodal [19,20] activity. All three compounds exhibited good activity against Madurella as well as Actinomadura, with MIC values somewhat higher than the reference drug itraconazole for M. mycetomatis and considerably lower than the reference drug cotrimoxazole for Actinomadura (Table 2).

3. Discussion

In this small drug repurposing study, we have evaluated redox-active parasiticides and antibiotics against M. mycetomatis based on the rationale that the metabolism of the fungus would be able to reduce, and thereby activate, the prodrugs. This hypothesis turned out to be wrong, as all tested molecules were inactive—except for niclosamide. Given the lack of activity of the tested nitrofurans and nitroimidazoles, the observed activity of niclosamide is likely not due to its nitro group, but rather due to the salicylanilide moiety. This is supported by the good activity of MMV665807 against M. mycetomatis, a salicylanilide that lacks a nitro group (Figure 1). Thus, the discovery of niclosamide as a hit for mycetoma pathogens was serendipitous. The nitro group of niclosamide was found to be dispensable also for its inhibitory action on Wnt signaling, an activity that has raised interest in niclosamide as an anticancer agent [21]; another such activity is mitochondrial uncoupling [22,23].
Niclosamide is an old drug of many uses [24,25]. It was developed by Bayer (Bayer 2353) in the 1950s as a molluscicide for schistosomiasis control. Since 1982, when it was approved by the FDA for human use, its primary indication has been as a broad-spectrum anthelmintic for tapeworms (Taenia spp., Diphyllobothrium latum) and intestinal fluke (Fasciolopsis buski) [26]. Niclosamide was shown to have promising activity against bacteria [27,28], fungi [29], and even coronavirus [30,31]. What restricted its use to intestinal pathogens was the poor oral bioavailability, i.e., the fact that niclosamide is not significantly absorbed from the gastrointestinal tract [32,33]. Different carriers or formulations have been employed to overcome this issue, for example, [34,35,36]. The ethanolamine salt of niclosamide (NEN, also called niclosamide olamine or clonitralide) has a better water-solubility and bioavailability [33], and it is being considered for different (re)purposes [22,37,38,39,40]. In our in vitro assays, NEN was as active as niclosamide against M. mycetomatis and Actinomadura spp.
The finding that a drug like niclosamide, which is on the WHO’s list of Essential Medicines, exhibited in vitro activity against both Madurella mycetomatis and Actinomadura spp. in the same range as the reference compounds, warrants the testing of further salicylanilides against these pathogens and the consideration of niclosamide as a repurposing candidate for mycetoma.

4. Materials and Methods

4.1. Chemicals

MMV665807 (Princeton Bio Molecular Research) and niclosamide-ethanolamine (2A Biotech) were received from Britta Lundström-Stadelmann (University of Bern, Switzerland), the five peroxides from Jonathan Vennerstrom (University of Nebraska, USA). All other test compounds were obtained from DNDi (Geneva, Switzerland). Ro 15-6547 is 4′[(1-methyl-2-nitroimidazole-5-yl)methoxy]-1-pyrrolidine-acetanilide, and RJ-55 and RJ-164 are 1-aryl-4-nitroimidazoles; the three nitroimidazoles were developed for African trypanosomiasis [10,11]. OZ78 is an ozonide related to arterolane [41,42] that has mainly been studied for its activity against trematodes [43,44,45]. All compounds were dissolved in DMSO.

4.2. Strains

Madurella mycetomatis isolate SO1 was originally isolated from a Somalian patient and CBS131320 from a Sudanese patient. The two PCR-identified strains were obtained from the mycetoma collection of the Erasmus Medical Centre, Rotterdam, The Netherlands. The mycelia were grown at 37 °C in RPMI 1640 medium supplemented with 0.35 g/L l-glutamine and 1.98 mM 4-morpholinepropane sulfonic acid. Actinomadura madurae SAK-A05 and Actinomadura syzygii SAK-A08 were originally isolated from Sudanese patients and grown in the pharmaceutical research laboratory, University of Science and Technology repository (Omdurman, Sudan). The bacterial suspensions were prepared in Mueller Hinton II broth (CAMHB) media and propagated at 35 °C.

4.3. In Vitro Drug Efficacy Testing

M. mycetomatis mycelia in RPMI 1640 medium were sonicated for 10 s (QSONICA Q55) and centrifuged at 2600× g for 5 min. The mycelia were washed and resuspended in fresh RPMI 1640 medium to obtain a fungal suspension of 68% to 72% transmission at 660 nm (UV/Vis Spectrophotometer 6305, Jenway, UK). A 1:2 serial drug dilution covering a range from 256 μg/mL to 0.063 μg/mL of test compound was prepared in a round-bottom 96-well microtiter plate (Corning, CLS379). Itraconazole was used as the positive control at a range from 1 μg/mL to 0.03 μg/mL. Exactly 100 µL of adjusted fungal suspension was added to each well along with 1 µL of the drug. Next, 1 µL of resazurin was added to a final concentration of 0.15 mg/mL [12]. The plates were sealed and incubated at 37 °C for 7 days. All assays were performed in triplicate. For further details see [12].
Actinomadura suspensions were adjusted to absorbance of 0.08–0.1 at 625 nm. A 1:2 serial drug dilution covering a range from 256 μg/mL to 0.063 μg/mL was prepared in a 96-well microtiter plate. Co-trimoxazole (fixed combination of 1 part trimethoprim and 5 parts sulfamethoxazole) was employed as the positive control at a starting concentration of 80 µg/mL. Next, 100 µL of adjusted bacterial suspension were added to each well along with 1 µL of the drug. Precisely 1 µL of resazurin was added to a final concentration of 0.15 mg/mL. The plates were incubated at 35 °C for 5 days. All assays were performed in triplicate.
The assay plates were inspected for visual and spectrometric endpoints. Absorbance was measured at 600 nm (Thermo Scientific Multiskan Spectrum, Thermo Fisher Scien-tific, Vantaa, Finland). The minimal inhibitory concentration (MIC) was defined as the lowest concentration of test compound that produced ≥80% growth inhibition as compared to the untreated cultures (negative control).

Author Contributions

Conceptualization, A.B.M., M.K., S.K. and P.M.; methodology, S.A.A., W.v.d.S., S.K.; validation, S.A.A. and A.B.M.; formal analysis, S.A.A. and A.B.M.; investigation, S.A.A. and A.B.M.; resources, M.K., S.K., W.v.d.S.; data curation, S.A.A. and A.B.M.; writing—original draft preparation, P.M.; writing—review and editing, A.B.M. and S.A.A.; visualization, P.M.; supervision, M.K., S.K. and P.M.; project ad-ministration, P.M.; funding acquisition, A.B.M. and P.M. All authors have read and agreed to the published version of the manuscript.

Funding

We gratefully acknowledge financial support by the Emilia Guggenheim-Schnurr Foundation (www.ngib.ch/stiftung-egs/) to A.B.M. The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are contained within the article.

Acknowledgments

We thank DNDi for the nitroheterocyclic drugs, Jonathan Vennerstrom for the peroxides, and Britta Lundström-Stadelmann and Reto Rufener for MMV665807 and helpful advice. We are most grateful to Marcel Tanner and Suad Sulaiman for their continuous support.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Sample Availability

For samples of the compounds please contact the corresponding author, who will link you up with the compound providers (see Acknowledgments above).

References

  1. Nenoff, P.; van de Sande, W.W.; Fahal, A.H.; Reinel, D.; Schofer, H. Eumycetoma and actinomycetoma--an update on causative agents, epidemiology, pathogenesis, diagnostics and therapy. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 1873–1883. [Google Scholar] [CrossRef] [PubMed]
  2. Zijlstra, E.E.; van de Sande, W.W.J.; Welsh, O.; Mahgoub, E.S.; Goodfellow, M.; Fahal, A.H. Mycetoma: A unique neglected tropical disease. Lancet Infect. Dis. 2016, 16, 100–112. [Google Scholar] [CrossRef]
  3. Ahmed, S.A.; van de Sande, W.W.; Stevens, D.A.; Fahal, A.; van Diepeningen, A.D.; Menken, S.B.; de Hoog, G.S. Revision of agents of black-grain eumycetoma in the order Pleosporales. Persoonia 2014, 33, 141–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. van de Sande, W.; Fahal, A.; Ahmed, S.A.; Serrano, J.A.; Bonifaz, A.; Zijlstra, E. eumycetoma working, g. Closing the mycetoma knowledge gap. Med. Mycol. 2018, 56, 153–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Suleiman, S.H.; Wadaella, E.S.; Fahal, A.H. The Surgical Treatment of Mycetoma. PLoS Negl. Trop. Dis. 2016, 10, e0004690. [Google Scholar] [CrossRef] [Green Version]
  6. Ferreira, L.G.; Andricopulo, A.D. Drug repositioning approaches to parasitic diseases: A medicinal chemistry perspective. Drug Discov. Today 2016, 21, 1699–1710. [Google Scholar] [CrossRef]
  7. Lim, W.; Melse, Y.; Konings, M.; Phat Duong, H.; Eadie, K.; Laleu, B.; Perry, B.; Todd, M.H.; Ioset, J.R.; van de Sande, W.W.J. Addressing the most neglected diseases through an open research model: The discovery of fenarimols as novel drug candidates for eumycetoma. PLoS Negl. Trop. Dis. 2018, 12, e0006437. [Google Scholar] [CrossRef] [Green Version]
  8. Pal, C.; Bandyopadhyay, U. Redox-active antiparasitic drugs. Antioxid. Redox Signal. 2012, 17, 555–582. [Google Scholar] [CrossRef]
  9. Mäser, P. Cherchez l’Electron. Mol. Microbiol. 2017, 106, 183–185. [Google Scholar] [CrossRef] [Green Version]
  10. Richle, R.; Hofheinz, W. Chemotherapeutische Wirksamkeit von 2 neuen 2-Nitroimidazolderivaten gegen Trypanosoma brucei rhodesiense bei der experimentellen Schlafkrankheit von Maus und Kaninchen. Mitt. Österr. Ges. Tropenmed. Parasitol. 1983, 5, 143–149. [Google Scholar]
  11. Trunz, B.B.; Jedrysiak, R.; Tweats, D.; Brun, R.; Kaiser, M.; Suwinski, J.; Torreele, E. 1-Aryl-4-nitro-1H-imidazoles, a new promising series for the treatment of human African trypanosomiasis. Eur. J. Med. Chem. 2011, 46, 1524–1535. [Google Scholar] [CrossRef]
  12. Abd Algaffar, S.O.; Verbon, A.; van de Sande, W.W.J.; Khalid, S.A. Development and Validation of an In Vitro Resazurin-Based Susceptibility Assay against Madurella mycetomatis. Antimicrob. Agents Chemother. 2021, 65. [Google Scholar] [CrossRef] [PubMed]
  13. van de Sande, W.W.; Fahal, A.H.; Riley, T.V.; Verbrugh, H.; van Belkum, A. In vitro susceptibility of Madurella mycetomatis, prime agent of Madura foot, to tea tree oil and artemisinin. J. Antimicrob. Chemother. 2007, 59, 553–555. [Google Scholar] [CrossRef] [PubMed]
  14. Hecht, G.; Gloxhuber, C. Tolerance to 2′, 5-dichloro-4-nitrosalicylanilide ethanolamine salt. Z. Tropenmed. Parasit. 1962, 13, 1–8. [Google Scholar]
  15. Spangenberg, T.; Burrows, J.N.; Kowalczyk, P.; McDonald, S.; Wells, T.N.; Willis, P. The open access malaria box: A drug discovery catalyst for neglected diseases. PLoS ONE 2013, 8, e62906. [Google Scholar] [CrossRef] [Green Version]
  16. Zapotoczna, M.; Boksmati, N.; Donohue, S.; Bahtiar, B.; Boland, A.; Somali, H.A.; Cox, A.; Humphreys, H.; O’Gara, J.P.; Brennan, M.; et al. Novel anti-staphylococcal and anti-biofilm properties of two anti-malarial compounds: MMV665953 {1-(3-chloro-4-fluorophenyl)-3-(3,4-dichlorophenyl)urea} and MMV665807 {5-chloro-2-hydroxy-N-[3-(trifluoromethyl)phenyl]benzamide}. J. Med. Microbiol. 2017, 66, 377–387. [Google Scholar] [CrossRef]
  17. Aleman, A.; Guerra, T.; Maikis, T.J.; Milholland, M.T.; Castro-Arellano, I.; Forstner, M.R.; Hahn, D. The Prevalence of Trypanosoma cruzi, the Causal Agent of Chagas Disease, in Texas Rodent Populations. Ecohealth 2017, 14, 130–143. [Google Scholar] [CrossRef]
  18. Muller, J.; Winzer, P.A.; Samby, K.; Hemphill, A. In Vitro Activities of MMV Malaria Box Compounds against the Apicomplexan Parasite Neospora caninum, the Causative Agent of Neosporosis in Animals. Molecules 2020, 25, 1460. [Google Scholar] [CrossRef] [Green Version]
  19. Ritler, D.; Rufener, R.; Sager, H.; Bouvier, J.; Hemphill, A.; Lundstrom-Stadelmann, B. Development of a movement-based in vitro screening assay for the identification of new anti-cestodal compounds. PLoS Negl. Trop. Dis. 2017, 11, e0005618. [Google Scholar] [CrossRef] [PubMed]
  20. Stadelmann, B.; Rufener, R.; Aeschbacher, D.; Spiliotis, M.; Gottstein, B.; Hemphill, A. Screening of the Open Source Malaria Box Reveals an Early Lead Compound for the Treatment of Alveolar Echinococcosis. PLoS Negl. Trop. Dis. 2016, 10, e0004535. [Google Scholar] [CrossRef] [Green Version]
  21. Mook, R.A.; Jr Wang, J.; Ren, X.R.; Chen, M.; Spasojevic, I.; Barak, L.S.; Lyerly, H.K.; Chen, W. Structure-activity studies of Wnt/beta-catenin inhibition in the Niclosamide chemotype: Identification of derivatives with improved drug exposure. Bioorg. Med. Chem. 2015, 23, 5829–5838. [Google Scholar] [CrossRef] [Green Version]
  22. Alasadi, A.; Chen, M.; Swapna, G.V.T.; Tao, H.; Guo, J.; Collantes, J.; Fadhil, N.; Montelione, G.T.; Jin, S. Effect of mitochondrial uncouplers niclosamide ethanolamine (NEN) and oxyclozanide on hepatic metastasis of colon cancer. Cell Death Dis. 2018, 9, 215. [Google Scholar] [CrossRef] [Green Version]
  23. Kumar, R.; Coronel, L.; Somalanka, B.; Raju, A.; Aning, O.A.; An, O.; Ho, Y.S.; Chen, S.; Mak, S.Y.; Hor, P.Y.; et al. Mitochondrial uncoupling reveals a novel therapeutic opportunity for p53-defective cancers. Nat. Commun. 2018, 9, 3931. [Google Scholar] [CrossRef] [Green Version]
  24. Chen, W.; Mook, R.A.; Premont, R.T., Jr.; Wang, J. Niclosamide: Beyond an antihelminthic drug. Cell Signal 2018, 41, 89–96. [Google Scholar] [CrossRef]
  25. Kadri, H.; Lambourne, O.A.; Mehellou, Y. Niclosamide, a Drug with Many (Re)purposes. ChemMedChem 2018, 13, 1088–1091. [Google Scholar] [CrossRef]
  26. NIH National Center for Advancing Translational Sciences, Inxight: Drugs. Niclosamide. Available online: https://drugs.ncats.io/drug/8KK8CQ2K8G (accessed on 20 May 2021).
  27. Rajamuthiah, R.; Fuchs, B.B.; Conery, A.L.; Kim, W.; Jayamani, E.; Kwon, B.; Ausubel, F.M.; Mylonakis, E. Repurposing salicylanilide anthelmintic drugs to combat drug resistant Staphylococcus aureus. PLoS ONE 2015, 10, e0124595. [Google Scholar] [CrossRef] [Green Version]
  28. Imperi, F.; Massai, F.; Ramachandran Pillai, C.; Longo, F.; Zennaro, E.; Rampioni, G.; Visca, P.; Leoni, L. New life for an old drug: The anthelmintic drug niclosamide inhibits Pseudomonas aeruginosa quorum sensing. Antimicrob. Agents Chemother. 2013, 57, 996–1005. [Google Scholar] [CrossRef] [Green Version]
  29. Garcia, C.; Burgain, A.; Chaillot, J.; Pic, E.; Khemiri, I.; Sellam, A. A phenotypic small-molecule screen identifies halogenated salicylanilides as inhibitors of fungal morphogenesis, biofilm formation and host cell invasion. Sci. Rep. 2018, 8, 11559. [Google Scholar] [CrossRef] [Green Version]
  30. Jeon, S.; Ko, M.; Lee, J.; Choi, I.; Byun, S.Y.; Park, S.; Shum, D.; Kim, S. Identification of Antiviral Drug Candidates against SARS-CoV-2 from FDA-Approved Drugs. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef] [PubMed]
  31. Wu, C.J.; Jan, J.T.; Chen, C.M.; Hsieh, H.P.; Hwang, D.R.; Liu, H.W.; Liu, C.Y.; Huang, H.W.; Chen, S.C.; Hong, C.F.; et al. Inhibition of severe acute respiratory syndrome coronavirus replication by niclosamide. Antimicrob. Agents Chemother. 2004, 48, 2693–2696. [Google Scholar] [CrossRef] [Green Version]
  32. Barbosa, E.J.; Lobenberg, R.; de Araujo, G.L.B.; Bou-Chacra, N.A. Niclosamide repositioning for treating cancer: Challenges and nano-based drug delivery opportunities. Eur. J. Pharm. Biopharm. 2019, 141, 58–69. [Google Scholar] [CrossRef]
  33. Schultz, D.P.; Harman, P.D. Uptake, distribution, and elimination of the lampricide 2’,5-dichloro-4’-nitro[14C]salicylanilide (Bayer 2353) and its 2-aminoethanol salt (Bayer 73) by largemouth bass. J. Agric. Food Chem. 1978, 26, 1226–1230. [Google Scholar] [CrossRef]
  34. Guo, J.; Tao, H.; Alasadi, A.; Huang, Q.; Jin, S. Niclosamide piperazine prevents high-fat diet-induced obesity and diabetic symptoms in mice. Eat. Weight Disord. 2019, 24, 91–96. [Google Scholar] [CrossRef]
  35. Rehman, M.U.; Khan, M.A.; Khan, W.S.; Shafique, M.; Khan, M. Fabrication of Niclosamide loaded solid lipid nanoparticles: In vitro characterization and comparative in vivo evaluation. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1926–1934. [Google Scholar]
  36. Zhang, X.; Zhang, Y.; Zhang, T.; Zhang, J.; Wu, B. Significantly enhanced bioavailability of niclosamide through submicron lipid emulsions with or without PEG-lipid: A comparative study. J. Microencapsul. 2015, 32, 496–502. [Google Scholar] [CrossRef] [PubMed]
  37. Han, P.; Zhan, H.; Shao, M.; Wang, W.; Song, G.; Yu, X.; Zhang, C.; Ge, N.; Yi, T.; Li, S.; et al. Niclosamide ethanolamine improves kidney injury in db/db mice. Diabetes Res. Clin. Pract. 2018, 144, 25–33. [Google Scholar] [CrossRef]
  38. Li, S.L.; Yan, J.; Zhang, Y.Q.; Zhen, C.L.; Liu, M.Y.; Jin, J.; Gao, J.L.; Xiao, X.L.; Shen, X.; Tai, Y.; et al. Niclosamide ethanolamine inhibits artery constriction. Pharmacol. Res. 2017, 115, 78–86. [Google Scholar] [CrossRef]
  39. Park, J.S.; Lee, Y.S.; Lee, D.H.; Bae, S.H. Repositioning of niclosamide ethanolamine (NEN), an anthelmintic drug, for the treatment of lipotoxicity. Free Radic Biol. Med. 2019, 137, 143–157. [Google Scholar] [CrossRef]
  40. Tao, H.; Zhang, Y.; Zeng, X.; Shulman, G.I.; Jin, S. Niclosamide ethanolamine-induced mild mitochondrial uncoupling improves diabetic symptoms in mice. Nat. Med. 2014, 20, 1263–1269. [Google Scholar] [CrossRef] [Green Version]
  41. Lanteri, C.A.; Chaorattanakawee, S.; Lon, C.; Saunders, D.L.; Rutvisuttinunt, W.; Yingyuen, K.; Bathurst, I.; Ding, X.C.; Tyner, S.D. Ex vivo activity of endoperoxide antimalarials, including artemisone and arterolane, against multidrug-resistant Plasmodium falciparum isolates from Cambodia. Antimicrob. Agents Chemother. 2014, 58, 5831–5840. [Google Scholar] [CrossRef] [Green Version]
  42. Vennerstrom, J.L.; Arbe-Barnes, S.; Brun, R.; Charman, S.A.; Chiu, F.C.; Chollet, J.; Dong, Y.; Dorn, A.; Hunziker, D.; Matile, H.; et al. Identification of an antimalarial synthetic trioxolane drug development candidate. Nature 2004, 430, 900–904. [Google Scholar] [CrossRef] [PubMed]
  43. Keiser, J.; Utzinger, J.; Tanner, M.; Dong, Y.; Vennerstrom, J.L. The synthetic peroxide OZ78 is effective against Echinostoma caproni and Fasciola hepatica. J. Antimicrob. Chemother. 2006, 58, 1193–1197. [Google Scholar] [CrossRef] [Green Version]
  44. Keiser, J.; Xiao, S.H.; Dong, Y.; Utzinger, J.; Vennerstrom, J.L. Clonorchicidal properties of the synthetic trioxolane OZ78. J. Parasitol. 2007, 93, 1208–1213. [Google Scholar] [CrossRef] [PubMed]
  45. Xiao, S.H.; Keiser, J.; Chollet, J.; Utzinger, J.; Dong, Y.; Endriss, Y.; Vennerstrom, J.L.; Tanner, M. In vitro and in vivo activities of synthetic trioxolanes against major human schistosome species. Antimicrob. Agents Chemother. 2007, 51, 1440–1445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Chemical structure of niclosamide and MMV665807: Both compounds are salicylanilides, i.e., amides of salicylic acid and aniline.
Figure 1. Chemical structure of niclosamide and MMV665807: Both compounds are salicylanilides, i.e., amides of salicylic acid and aniline.
Molecules 26 04005 g001
Table 1. Selected redox-active agents and their in vitro activity against M. mycetomatis: All assays were performed in triplicate. Values are minimal inhibitory concentrations (MIC) in µg/mL.
Table 1. Selected redox-active agents and their in vitro activity against M. mycetomatis: All assays were performed in triplicate. Values are minimal inhibitory concentrations (MIC) in µg/mL.
CompoundClassIndication 1SO1 2CBS131320 2
NiclosamideSalicylanilideTapeworms0.781.6
SecnidazoleNitroimidazoleBacterial vaginosis>256>256
MetronidazoleNitroimidazoleBroad spectrum antibiotic>256>256
FexinidazoleNitroimidazoleHuman African trypanosomiasis>256>256
RJ-164Nitroimidazole(Human African trypanosomiasis)>256>256
RJ-55Nitroimidazole(Human African trypanosomiasis)>256>256
Ro 15-6547Nitroimidazole(Human African trypanosomiasis)>256>256
NifurtimoxNitrofuranChagas’ disease, HAT>256>256
NifuroxazideNitrofuranColitis and diarrhea>256>256
NitrofurantoineNitrofuranUrinary tract infections>256>256
OZ 78Peroxide(Malaria, trematodes)>256>256
ArtemisininPeroxideMalaria1616
DihydroartemisininPeroxideMalaria>256>256
ArtesunatePeroxideMalaria>256>256
ArtemetherPeroxideMalaria6464
ItraconazoleTriazoleAntifungal0.130.25
1 For experimental compounds, the envisaged indication is in parentheses; 2 SO1 and CBS131320 are two different isolates of M. mycetomatis.
Table 2. In vitro activity of niclosamide and related compounds against causative agents of mycetoma: All assays were performed in triplicate. Values are MIC in µg/mL.
Table 2. In vitro activity of niclosamide and related compounds against causative agents of mycetoma: All assays were performed in triplicate. Values are MIC in µg/mL.
CompoundSO1 1CBS131320 1SAK-A05 2SAK-A08 2
Niclosamide0.781.60.390.39
Niclosamide-ethanolamine0.781.60.190.39
MMV6658071.61.60.390.39
Itraconazole0.130.25n.d.n.d.
Cotrimoxazolen.d.n.d.2010
1 SO1 and CBS131320 are two different isolates of M. mycetomatis.2 SAK-A05 and SAK-A08 are two species of Actinomadura (A. madurae and A. syzygii, respectively).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mahmoud, A.B.; Abd Algaffar, S.; van de Sande, W.; Khalid, S.; Kaiser, M.; Mäser, P. Niclosamide Is Active In Vitro against Mycetoma Pathogens. Molecules 2021, 26, 4005. https://doi.org/10.3390/molecules26134005

AMA Style

Mahmoud AB, Abd Algaffar S, van de Sande W, Khalid S, Kaiser M, Mäser P. Niclosamide Is Active In Vitro against Mycetoma Pathogens. Molecules. 2021; 26(13):4005. https://doi.org/10.3390/molecules26134005

Chicago/Turabian Style

Mahmoud, Abdelhalim B., Shereen Abd Algaffar, Wendy van de Sande, Sami Khalid, Marcel Kaiser, and Pascal Mäser. 2021. "Niclosamide Is Active In Vitro against Mycetoma Pathogens" Molecules 26, no. 13: 4005. https://doi.org/10.3390/molecules26134005

Article Metrics

Back to TopTop