Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A screening of the MMV Pathogen Box® reveals new potential antifungal drugs against the etiologic agents of chromoblastomycosis

  • Rowena Alves Coelho ,

    Roles Conceptualization, Data curation, Methodology, Writing – original draft

    rowena.alves@ini.fiocruz.br, rodrigo.paes@ini.fiocruz.br

    Affiliation Mycology Laboratory at the Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil

  • Luna Sobrino Joffe,

    Roles Formal analysis, Methodology

    Affiliation Department of Microbiology and Immunology, Stony Brook University, New York, NY, United States of America

  • Gabriela Machado Alves,

    Roles Methodology

    Affiliation Mycology Laboratory at the Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil

  • Maria Helena Galdino Figueiredo-Carvalho,

    Roles Data curation, Methodology, Writing – review & editing

    Affiliation Mycology Laboratory at the Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil

  • Fábio Brito-Santos,

    Roles Formal analysis, Writing – review & editing

    Affiliation Mycology Laboratory at the Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil

  • Ana Claudia Fernandes Amaral,

    Roles Visualization, Writing – review & editing

    Affiliation Laboratory of Natural Products Chemistry, Farmanguinhos, Fiocruz, Rio de Janeiro, Brazil

  • Marcio L. Rodrigues,

    Roles Formal analysis, Project administration, Resources, Validation, Writing – review & editing

    Affiliations Carlos Chagas Institute, Fiocruz, Paraná, Brazil, Microbiology Institute, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil

  • Rodrigo Almeida-Paes

    Roles Conceptualization, Formal analysis, Funding acquisition, Project administration, Resources, Supervision, Validation, Writing – original draft, Writing – review & editing

    Affiliation Mycology Laboratory at the Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil

Abstract

Chromoblastomycosis (CBM) is a chronic subcutaneous mycosis caused by traumatic implantation of many species of black fungi. Due to the refractoriness of some cases and common recurrence of CBM, a more effective and less time-consuming treatment is mandatory. The aim of this study was to identify compounds with in vitro antifungal activity in the Pathogen Box® compound collection against different CBM agents. Synergism of these compounds with drugs currently used to treat CBM was also assessed. An initial screening of the drugs present in this collection at 1 μM was performed with a Fonsecaea pedrosoi clinical strain according to the EUCAST protocol. The compounds with activity against this fungus were also tested against other seven etiologic agents of CBM (Cladophialophora carrionii, Phialophora verrucosa, Exophiala jeanselmei, Exophiala dermatitidis, Fonsecaea monophora, Fonsecaea nubica, and Rhinocladiella similis) at concentrations ranging from 0.039 to 10 μM. The analysis of potential synergism of these compounds with itraconazole and terbinafine was performed by the checkerboard method. Eight compounds inhibited more than 60% of the F. pedrosoi growth: difenoconazole, bitertanol, iodoquinol, azoxystrobin, MMV688179, MMV021013, trifloxystrobin, and auranofin. Iodoquinol produced the lowest MIC values (1.25–2.5 μM) and MMV688179 showed MICs that were higher than all compounds tested (5 - >10 μM). When auranofin and itraconazole were tested in combination, a synergistic interaction (FICI = 0.37) was observed against the C. carrionii isolate. Toxicity analysis revealed that MMV021013 showed high selectivity indices (SI ≥ 10) against the fungi tested. In summary, auranofin, iodoquinol, and MMV021013 were identified as promising compounds to be tested in CBM models of infection.

Introduction

Chromoblastomycosis (CBM) is a subcutaneous neglected mycosis [1]. It is caused by the implantation of one of its etiological agents through a trauma to the skin. Black fungi from the genera Fonsecaea, Cladophialophora, Rhinocladiella, Phialophora, Exophiala, among others, can cause CBM. Since this mycosis is not of compulsory notification in most countries where it occurs, its distribution is based on case reports and case series, with most of them occurring in Latin America, Africa, and Asia. In South America, most of the cases of CBM have been described in Brazil, in Africa most cases come from Madagascar, and China is the leading Asian country on number of cases [2]. In Brazil, the most frequent CBM agents belong to the genus Fonsecaea [36]. F. pedrosoi is the predominant species in South America, followed by F. monophora [7]. F. pedrosoi also predominates in humid regions of most countries where CBM is endemic, including Madagascar and China. Cladophialophora carrionii is more frequent in arid areas of these countries [810]. The frequency of infections by species belonging to other genera of black fungi varies among different geographic regions [2].

CBM mainly affects agricultural and construction workers, which develop extensive injuries with damage to the affected limb [11]. Most patients often take a long time to seek medical help, therefore their lesions are usually extensive [2]. Although there is no official therapeutic protocol for CBM, itraconazole is the most frequently used drug, followed by terbinafine [2]. In developing countries, including Brazil, treatment based on the use of these drugs is long and expensive, and some patients show recurrence and refractoriness [2,12]. These patients usually require more than one therapeutic method, often including physical methods, such as cryosurgery or laser therapy [13]. The in vitro response of the CBM agents to other antifungal drugs currently used to treat mycotic infections, such as amphotericin B, fluconazole, flucytosine, and micafungin is not satisfactory [14]. A more effective and less time-consuming treatment is clearly required to combat CBM.

The discovery of new pharmacological agents, however, is costly and time-consuming. Moreover, most of compounds in pre-clinical or clinical studies will never be approved for human use [15]. A useful approach to bypass these problems is drug repurposing, where drugs already studied, and sometimes approved, to treat other medical conditions are redirected to target a new disease [16].

In order to identify novel drugs with activity against neglected diseases, the Medicines for Malaria Venture (MMV, Switzerland; http://www.pathogenbox.org/), developed a collection of 400 compounds named Pathogen Box®. This initiative provides the tools for identifying active compounds against neglected pathogens [17,18]. The aim of this study was to identify in vitro antifungal activity within the Pathogen Box® compounds against the CBM agents and to investigate the synergism of these new compounds with drugs currently used to treat CBM.

Materials and methods

Fungal strains and growth conditions

The eight strains used in the study were obtained from the Collection of Pathogenic Fungi (CFP) of Fiocruz, as well as from the American Type Culture Collection (ATCC). Fonsecaea pedrosoi CFP00791 was used throughout the study. This species is one of the major agents of CBM in most regions where this disease is endemic. Cladophialophora carrionii CFP 00910, Phialophora verrucosa CFP 00937, Fonsecaea monophora CFP 00911, Fonsecaea nubica CFP 00912, Rhinocladiela similis CFP 00790, Exophiala jeanselmei var. heteromorpha ATCC 28180, and Exophiala dermatitidis ATCC 28869 were used for minimal inhibitory concentration (MIC) and synergism assays. Strains were maintained on potato dextrose agar (PDA) (Sigma Chemical Corporation, St. Louis, MO, USA). Seven-day-old cultures incubated at 30°C were used in the assays.

The Pathogen Box® compounds

The Pathogen Box® was kindly provided by Medicines for Malaria Venture (MMV, Geneva, Switzerland). It contains 400 different compounds tested for cytotoxicity with values within levels considered acceptable for an initial drug discovery programme [19]. The Pathogen Box® contains compounds with proven activity against neglected pathogens, including those causing tuberculosis, malaria, helminthiasis, cryptosporidiosis, toxoplasmosis, and dengue [2023]. The compounds were supplied in 96-well microtiter plates containing 10 μl/well of 10 mM compound solutions in dimethylsulfoxide (DMSO). The plates were diluted to a final drug concentration of 1 mM in DMSO (Sigma Chemical Corporation, St. Louis, MO, USA), as recommended by the fabricant, for the drug screening. The first and last columns in each plate were left as blank for negative and positive controls, respectively. All plates were stored at −20°C until their use in the following experiments.

Screening for antifungal activity

For the initial screening, all compounds were tested in 96-well plates (Kasvi Ltda, São José dos Pinhais, PR, Brazil) at a final concentration of 1μM in 100 μl of RPMI 1640 medium, with phenol red, with L-glutamine, and without sodium bicarbonate (Sigma Chemical Corporation), buffered with morpholine propanesulfonic acid (MOPS) (Vetec Química Fina Ltda, Rio de Janeiro, RJ, Brazil) at pH 7.0, and supplemented for a final 2% glucose (Neon Comercial Ltda, São Paulo, SP, Brazil) concentration. The antifungal drugs available as reference compounds in the MMV collection were used as controls of fungal growth inhibition. DMSO concentration in all wells, including those used for fungal control growth, at this point, corresponded to 1%. For preparation of the fungal inoculum, the F. pedrosoi strain CFP 00791 was grown as described above and then its conidia were suspended in sterile distilled water supplemented with 0.1% Tween 20 (Sigma Chemical Corporation) and vortexed, with the suspension turbidity adjusted to the 0.5 McFarland scale. This suspension was further diluted 1:10 and then 100 μl of the fungal inoculum was added to each well containing the compounds, generating a final working inoculum density of 2–5 × 105 CFU/ml and a final DMSO concentration of 0.5% in each well, including controls [21]. Plates were incubated at 35°C for 72–96 hours. The optical density at 530 nm (OD530) was recorded using the Epoch microplate reader (Biotek Instruments Inc, Winooski, VT, USA). The percentage of inhibition of fungal growth (%IG) was calculated according to the formula: %IG = (1- (OD1 / OD2)) × 100, where OD1 = fungal optical density in the presence of the drug; OD2 = optical density of the fungal growth control well without any drug. The %IG data of the 400 compounds was plotted using the Prism 8 software (GraphPad Software, San Diego, CA, USA). Compounds that presented %IG values greater than 60% were selected for further assays.

Minimal inhibitory concentration (MIC) of selected drugs

Assays for MIC determination were performed in sterile polystyrene flat-bottom 96-well microtiter plates using the broth microdilution method according to the EUCAST guidelines [24]. The final concentration of the selected compounds of the Pathogen Box® ranged from 0.015 to 10 μM. All isolates were tested at a final cellular density of 2 to 5 × 105 CFU/ml. Plates were incubated at 35°C for 72–96 h. All experiments were repeated at least two times. The MIC was defined as the lowest drug concentration (μM) that inhibited 100% of fungal growth.

Fungicidal activity

The minimal fungicidal concentration (MFC) was determined by transferring an aliquot of 5 μl of each well without fungal growth of the microdilution plates used for the determination of MIC, as described above, on Sabouraud 2% glucose agar (Sigma Chemical Corporation). The MFC was determined as the lowest drug concentration without fungal growth on the Sabouraud agar after five days of incubation at 35°C. When the MFC:MIC ratio of an agent is 1 or 2, the compound is considered fungicidal against the pathogen. If the ratio is higher than 2, the mode of action is probably fungistatic [25,26].

Evaluation of compounds applicability in CBM treatment

Cytotoxicity data of the compounds was provided by the MMV. The selectivity index (SI) was calculated as follows: SI = CC50 (μM) / MIC (μM), where CC50 is the drug concentration that kills 50% of the cells tested. Compounds with selectivity index lower than 1 were not considered for the synergistic assays. The original use of the compounds and their known side-effects were also verified in the literature, to evaluate their safety for human use. Compounds found to have dangerous side-effects were further excluded from the analysis.

Synergism evaluation

Synergistic activity between the selected compounds and standard antifungal drugs used in the treatment of CBM (itraconazole and terbinafine) was tested by the checkerboard method [27]. Two drugs were loaded into a single 96-well plate, so that in each of the wells there were different concentrations of the compound-antifungal combination. Compound/antifungal dilutions were prepared following the methodology proposed by the EUCAST, starting from a 100-fold concentrated stock compound/antifungal solution according to the MIC determination methodology [24]. Final concentrations of the standard drugs corresponded to 0.06–4 μg/ml (itraconazole) and 0.015–1 μg/ml (terbinafine). Serial two-fold dilutions of the selected Pathogen Box® compounds were performed so that the MIC of that compound was in a central position of the plate (lines 5–7). Fungal inocula and incubation conditions were the same that were described for the previous experiments. The drug interaction was classified according to the fractional inhibitory concentration index (FICI). The FICI was obtained by the formula: FICI ═ (A/MIC (a)) + (B/MIC (b)), where: A = MIC of the drug (a) in combination; MIC (a) = MIC of drug (a) alone; B = MIC of the drug (b) in combination; MIC (b) = MIC of drug (b) alone [28]. The type of interaction between the antifungal agents in combination was classified as the following: synergism if FICI ≤ 0.5; indifference if 0.5 < FICI < 4 and antagonism if FICI 4 [27,28].

Results

Identification of compounds with antifungal activity against CBM agents

The internal reference drug posaconazole inhibited 99% of the F. pedrosoi growth, validating the assay. Amphotericin B, which is also present in the drug collection, inhibited F. pedrosoi growth by 29%. Among the 400 compounds present in the Pathogen Box®, another eight drugs were found to have antifungal activity against the F. pedrosoi CFP00791 strain (Fig 1). These compounds were identified as difenoconazole (%IG = 98%), bitertanol (%IG = 98%), iodoquinol (%IG = 99%), azoxytrobin (%IG = 95%), MMV688179 (%IG = 73%), MMV021013 (%IG = 62%), trifloxystrobin (%IG = 82%), and auranofin (%IG = 82%). The chemical structure of these compounds is depicted in Fig 2. Their original applications, as described by Medicines for Malaria Venture, are as follows: difenoconazole, bitertanol, MMV688179, and trifloxystrobin have anti-kinetoplastid action, iodoquinol has anti-onchocerciasis action, azoxystrobin has anti-malaria action, MMV021013 has anti-tuberculosis action, and auranofin, a regulated drug for rheumatoid arthritis treatment, is another reference compound present in the collection.

thumbnail
Fig 1. Screening of 400 compounds present in the MMV Pathogen Box® collection against a Fonsecaea pedrosoi strain (CFP00791).

Results are presented as the percent inhibition of growth (%IG) of the fungal strain. The compounds tested are grouped according their original application against some infectious diseases, as described by Medicines for Malaria Venture. The dashed line represents the threshold applied in this study to select compounds with anti-chromoblastomycosis activity. Red symbols represent compounds that were discarded, due to low %IG. Green symbols represent compounds selected for further studies. The blue symbol represents the internal control drug posaconazole.

https://doi.org/10.1371/journal.pone.0229630.g001

thumbnail
Fig 2.

Chemical structures of the eight compounds with anti-Fonsecaea pedrosoi activity present in the MMV Pathogen Box® collection: (A) MMV688943 (difenoconazole); (B) MMV688942 (bitertanol); (C) MMV688179 (2-[3-chloro-4-[5-[2-chloro-4-(diaminomethylideneamino)phenyl]furan-2-yl]phenyl]guanidine); (D) MMV002817 (iodoquinol); (E) MMV688754 (trifloxystrobin); (F) MMV021013 (N-cyclohexyl-6-cyclopropyl-2-pyridin-2-ylpyrimidin-4-amine); (G) MMV021057 (azoxystrobin); (H) MMV688978 (auranofin).

https://doi.org/10.1371/journal.pone.0229630.g002

Efficacy and spectrum of antifungal activity of the selected compounds

Table 1 depicts the MIC and MFC values for each selected compound and the eight CBM agents tested. In brief, iodoquinol (MMV002817) presented the lowest MIC values among the compounds that we tested for antifungal activity. Seven out of the eight strains had MIC values corresponding to 1.25 μM and one MIC was equal to 2.5 μM to this compound. MMV688179 showed the highest MIC values against all species (5 - >10μM). Auranofin (MMV688978) was fungicidal against P. verrucosa and E. dermatitidis, and presented fungistatic activity against the other species. Iodoquinol (MMV002817) and bitertanol (MMV688942) were fungicidal against P. verrucosa and fungistatic against all other species. MMV688179 was fungicidal against P. verrucosa and F. monophora, and fungistatic against the other species. All other compounds had fungistatic activity against the set of isolates tested in this study.

thumbnail
Table 1. Determination of minimal inhibitory concentration (MIC) and minimal fungicidal concentration (MFC) (μM) against chromoblastomycosis agents.

https://doi.org/10.1371/journal.pone.0229630.t001

Analysis of synergistic effects

The compound MMV688179 was excluded from further analyses because it showed the highest MIC values (MIC >10 μM) and low selectivity index (Table 2). Trifloxystrobin was found to be toxic to keratinocytes [29], discouraging its use to treat CBM, which affects the skin. From the set of the other agricultural fungicides, we selected bitertanol for synergistic studies, on the basis of its higher selectivity index (Table 2). Therefore, iodoquinol, bitertanol, MMV021013, and auranofin were selected for the determination of FICI in the in vitro combination test with itraconazole and terbinafine. The concentration ranges of the compounds tested corresponded to 0.01–6 μg/ml for auranofin, 0.003–2.5 μg/ml for MMV021013, 0.005–3 μg/ml for bitertanol, and 0.007–4 μg/ml for iodoquinol. The results obtained from the checkerboard method with these four compounds in combination with itraconazole or terbinafine are detailed in Tables 3 and 4, respectively. The auranofin and itraconazole combination produced the single synergistic interaction (FICI < 0.5), against the C. carrionii strain. The combinations of iodoquinol, bitertanol, and MMV021013 with itraconazole produced indifferent interactions (FICI between 0.5 and 4) for all isolates tested. All compounds in combination with terbinafine produced indifferent interactions.

thumbnail
Table 2. Cytotoxicity values, minimal inhibitory concentration (MIC), and Selectivity Index (SI) of selected compounds with activity against the chromoblastomycosis agents.

https://doi.org/10.1371/journal.pone.0229630.t002

thumbnail
Table 3. Interaction of selected compounds with itraconazole.

https://doi.org/10.1371/journal.pone.0229630.t003

thumbnail
Table 4. Interaction of selected compounds with terbinafine.

https://doi.org/10.1371/journal.pone.0229630.t004

Discussion

CBM is difficult to treat because there is no standardized drug of choice. In addition, relapses are frequent for this mycosis [13]. The taxonomic diversity of the CBM agents adds another obstacle to the development of new drugs against this disease, because most patients are diagnosed exclusively on the basis of the presence of muriform cells in infected tissues [2]. Therefore, general therapeutic decisions are made regardless of the etiology of CBM, which may be deleterious for the patient. In this study, for instance, E. dermatitidis often presented MIC values higher than other species for the studied drugs. In this context, we recommend that repurposing studies for CBM use a large number of etiologic agents of this disease in the antifungal activity experiments.

The compound collection used in this study, the Pathogen Box® library, was previously demonstrated to contain molecules with antifungal activity. Mayer and Kronstad identified five compounds (tolfenpyrad, difenoconazole, bitertanol, posaconazole, and MMV688271) that exhibited antifungal activity against Cryptococcus neoformans and Candida albicans [18]. In our study, antifungal activities were identified for three of these compounds (difenoconazole, posaconazole, and bitertanol). It is noteworthy that tolfenpyrad inhibited F. pedrosoi growth by 49%. This compound is an agricultural insecticide and with well-known poisoning effects [30]. MMV688271, which completely inhibited the growth of C. neoformans and C. albicans [18], showed only 36% inhibition against F. pedrosoi in our screening assay. Vila and Lopez-Ribot evaluated three of the compounds that we investigated (MMV687807, MMV687273, and MMV688768) against C. albicans biofilms [17]. In the present study, these compounds showed 13% (MMV687807), 37% (MMV687273) and 24% (MMV688768) inhibition of the F. pedrosoi growth. Wall et al identified three compounds with anti-C. auris activity in the Pathogen Box® at a 5 μM concentration, including pentamidine, MMV659010, and iodoquinol. In this group, iodoquinol and MMV659010 showed %IG of 86 and 62, respectively, against plant cells. Pentamidine showed 61% inhibition in biofilms of C. auris [31]. In the present study, these compounds presented 34% (MMV659010), 49% (pentamidine), and 99% (iodoquinol) inhibition of the F. pedrosoi growth. These results clearly demonstrate that major fungal pathogens respond differently to the Pathogen Box® compounds (S1 Table).

The MMV Pathogen Box® contains two antifungal drugs as reference compounds. Inhibition of F. pedrosoi growth by posaconazole (99%) was observed, but amphotericin B inhibited less fungal growth (29%). This data can be explained by the high MIC values of amphotericin B (4–16 μg/ml) against this species [14].

In our study, three compounds were selected as promising anti-CBM agents: auranofin, iodoquinol, and MMV021013. Auranofin has been in clinical use as the gold standard for the oral treatment of rheumatoid arthritis since the 1980s [32,33]. The oral bioavailability of auranofin and its reduced side effects offer significant advantages over traditional injectable drugs [32,33]. Several studies were conducted to identify alternative therapeutic applications for auranofin, particularly in the area of infectious diseases. For example, antifungal activity against C. albicans [34] and C. neoformans [3436] has been described. In this study, low MIC values for this drug against eight different CBM agents were observed. Furthermore, the synergism found in the combination of auranofin with itraconazole against C. carrionii, the main CBM agent in arid and semi-arid areas [37], as well as its fungicidal activity against E. dermatitidis support the use of auranofin as a therapeutic option for CBM. These findings justify future in vivo testing and clinical studies to evaluate auranofin as an antifungal drug.

Iodoquinol (diiodohydroxyquinoline) is a luminal amebicide. Its use is recommended after treatment with a tissue amebicide, such as metronidazole, to eradicate surviving parasites in the colon [38]. This drug has low absorption through the gastrointestinal tract, so only the topical formulation would be applicable against CBM. Burnett and Mitchell demonstrated that the topical formulation of 1% iodoquinol in combination with 2% hydrocortisone acetate had antifungal activity against C. albicans, dermatophytes, and Malassezia furfur [39]. The study by Wall et al. demonstrated the in vitro efficacy of iodoquinol against several Candida species under planktonic conditions. As expected from the initial results with C. auris, iodoquinol was virtually ineffective against preformed biofilms of all species tested. However this compound displayed potent activity against planktonic growth of all strains tested, with IG50 (50% inhibition growth) values generally lower than 1 μg/ml [31]. In the present study, lower MIC values of iodoquinol (100% inhibition) were observed against all studied CBM agents, supporting its use against this group of fungi.

MMV021013 is a 2-pyridyl-4-aminopyrimidine. Antifungal activity has been described for some aminopyrimidines, including4-(2-Fluorophenyl)-6-trifluoromethyl-2-aminopyrimidine, active against Botrytis cinera [40], and 4-(1H-benzo[d]imidazol-2-yl)-6-(p-tolyl)pyrimidin-2-amine, active against Aspergillus flavus, Candida utilis, and Saccharomyces cerevisiae [41]. The Pathogen Box® classifies this compound as a tuberculosis targeted drug. Also, it was found to be active against Leishmania donovani and Trypanosoma cruzi [19]. To the best of our knowledge, no antifungal activity has been described for MMV021013. Some studies have identified molecules with in vitro activity against mycotic and tuberculosis agents [42,43], as well as against protozoa and fungi [44]. These observations suggest that these pathogens may share common antimicrobial targets that may propel drug discovery against a broad range of infectious diseases.

In summary, auranofin was identified in our study as a promising compound to be tested as a CBM therapeutic agent, on the basis of its low MIC values against CBM agents, synergism with itraconazole against a C. carrionii strain, and fungicidal action against P. verrucosa and E. dermatitidis. Iodoquinol has also proved promising for future studies involving CBM agents due to its low MIC values, fungicidal activity against P. verrucosa, and approved topical use in some countries. The 2-pyridyl-4-aminopyrimidine MMV021013 showed good selectivity and a combined activity against mycobacteria, Leishmania, Trypanosoma, and CBM agents. These data suggest the need for further in vitro and in vivo studies with these drugs aiming to develop new therapeutic tools against the CBM agents.

Supporting information

S1 Table. Screening of 400 compounds present in the MMV Pathogen Box® collection against a Fonsecaea pedrosoi strain (CFP00791).

Results are presented as the percent inhibition of growth (%IG) of the fungal strain.

https://doi.org/10.1371/journal.pone.0229630.s001

(PDF)

Acknowledgments

We are grateful for support from the Coordination for the Improvement of Higher Education Personnel (CAPES). We thank Medicines for Malaria Venture (MMV, Switzerland) for support, design and supply of the Pathogen Box®. We thank for Ingrid Ludmilla Rodrigues da Cruz, Iara Bastos de Andrade, Mônica dos Santos Elias, and Jonas Campos Pereira that contributed with technical assistance in this study.

References

  1. 1. Queiroz-Telles F. Chromoblastomycosis: a neglected tropical disease. Rev Inst Med Trop São Paulo. 2015;57: 46–50. pmid:26465369
  2. 2. Queiroz-Telles F, de Hoog S, Santos DWCL, Salgado CG, Vicente VA, Bonifaz A, et al. Chromoblastomycosis. Clin Microbiol Rev. 2017;30: 233–276. pmid:27856522
  3. 3. Najafzadeh MJ, Sun J, Vicente V, Xi L, van den Ende AHGG, de Hoog GS. Fonsecaea nubica sp. nov, a new agent of human chromoblastomycosis revealed using molecular data. Med Mycol. 2010;48: 800–806. pmid:20302550
  4. 4. Najafzadeh M, Gueidan C, Badali H, Van Den Ende AG, Xi L, Hoog G de. Genetic diversity and species delimitation in the opportunistic genus Fonsecaea. Med Mycol. 2009;47: 17–25. pmid:19107635
  5. 5. Gomes RR, Vicente VA, de Azevedo CM, Salgado CG, da Silva MB, Queiroz-Telles F, et al. Molecular epidemiology of agents of human chromoblastomycosis in Brazil with the description of two novel species. PLoS Negl Trop Dis. 2016;10: e0005102. pmid:27893750
  6. 6. de Azevedo CMPS, Gomes RR, Vicente VA, Santos DWCL, Marques SG, do Nascimento MMF, et al. Fonsecaea pugnacius, a novel agent of disseminated Chromoblastomycosis. J Clin Microbiol. 2015;53: 2674–2685. pmid:26085610
  7. 7. Queiroz-Telles F, Santos DW de CL. Challenges in the therapy of chromoblastomycosis. Mycopathologia. 2013;175: 477–488. pmid:23636730
  8. 8. Cohen J, Powderly WG, Opal SM. Infectious diseases. 2017.
  9. 9. Lu S, Lu C, Zhang J, Hu Y, Li X, Xi L. Chromoblastomycosis in Mainland China: A systematic review on clinical characteristics. Mycopathologia. 2013;175: 489–495. pmid:23086329
  10. 10. Xi L, Sun J, Lu C, Liu H, Xie Z, Fukushima K, et al. Molecular diversity of Fonsecaea (Chaetothyriales) causing chromoblastomycosis in southern China. Med Mycol. 2009;47: 27–33. pmid:18951291
  11. 11. Queiroz-Telles F, Nucci M, Colombo AL, Tobón A, Restrepo A. Mycoses of implantation in Latin America: an overview of epidemiology, clinical manifestations, diagnosis and treatment. Med Mycol. 2011;49: 225–236. pmid:21128710
  12. 12. Torres-Guerrero E, Isa-Isa R, Isa M, Arenas R. Chromoblastomycosis. Clin Dermatol. 2012;30: 403–408. pmid:22682188
  13. 13. Brito AC de, Bittencourt M de JS. Chromoblastomycosis: an etiological, epidemiological, clinical, diagnostic, and treatment update. An Bras Dermatol. 2018;93: 495–506. pmid:30066754
  14. 14. Coelho RA, Brito-Santos F, Figueiredo-Carvalho MHG, Silva JVDS, Gutierrez-Galhardo MC, do Valle ACF, et al. Molecular identification and antifungal susceptibility profiles of clinical strains of Fonsecaea spp. isolated from patients with chromoblastomycosis in Rio de Janeiro, Brazil. PLoS Negl Trop Dis. 2018;12: e0006675. pmid:30048450
  15. 15. Austin BA, Gadhia AD. New therapeutic uses for existing drugs. Adv Exp Med Biol. 2017;1031: 233–247. pmid:29214576
  16. 16. Breckenridge A, Jacob R. Overcoming the legal and regulatory barriers to drug repurposing. Nat Rev Drug Discov. 2019;18: 1–2. pmid:29880920
  17. 17. Vila T, Lopez-Ribot JL. Screening the Pathogen Box for identification of Candida albicans biofilm inhibitors. Antimicrob Agents Chemother. 2017;61. pmid:27795383
  18. 18. Mayer FL, Kronstad JW. Discovery of a novel antifungal agent in the Pathogen Box. mSphere. 2017;2. pmid:28435886
  19. 19. Duffy S, Sykes ML, Jones AJ, Shelper TB, Simpson M, Lang R, et al. Screening the Medicines for Malaria Venture Pathogen Box across multiple pathogens reclassifies starting points for open-source drug discovery. Antimicrob Agents Chemother. 2017;61. pmid:28674055
  20. 20. Jeong J, Kim G, Moon C, Kim HJ, Kim TH, Jang J. Pathogen Box screening for hit identification against Mycobacterium abscessus. PLOS ONE. 2018;13: e0195595. pmid:29698397
  21. 21. Hennessey KM, Rogiers IC, Shih H-W, Hulverson MA, Choi R, McCloskey MC, et al. Screening of the Pathogen Box for inhibitors with dual efficacy against Giardia lamblia and Cryptosporidium parvum. PLoS Negl Trop Dis. 2018;12: e0006673. pmid:30080847
  22. 22. Veale CGL, Hoppe HC. Screening of the Pathogen Box reveals new starting points for anti-trypanosomal drug discovery. MedChemComm. 2018;9: 2037–2044. pmid:30647879
  23. 23. Kipandula W, Young SA, MacNeill SA, Smith TK. Screening of the MMV and GSK open access chemical boxes using a viability assay developed against the kinetoplastid Crithidia fasciculata. Mol Biochem Parasitol. 2018;222: 61–69. pmid:29782894
  24. 24. Arendrup MC, Meletiadis J, Mouton JW, Lagrou K, Petr Hamal, Guinea J, et al. Method for the determination of broth dilution minimum inhibitory concentrations of antifungal agents for conidia forming moulds. EUCAST; 2017. Available: http://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/AFST/Files/EUCAST_E_Def_9_3_1_Mould_testing__definitive.pdf
  25. 25. CLSI—Clinical and Laboratory Standars Institute. Reference method for broth dilution antifungal susceptibility testing of filamentous fungi. Approved standard-second edition. Wayne, PA. USA: Clinical and Laboratory Standards Institute.; 2008.
  26. 26. Hafidh RR, Abdulamir AS, Vern LS, Abu Bakar F, Abas F, Jahanshiri F, et al. Inhibition of growth of highly resistant bacterial and fungal pathogens by a natural product. Open Microbiol J. 2011;5: 96–106. pmid:21915230
  27. 27. Dannaoui E, Afeltra J, Meis JF, Verweij PE, Network E, others. In vitro susceptibilities of zygomycetes to combinations of antimicrobial agents. Antimicrob Agents Chemother. 2002;46: 2708–2711. pmid:12121963
  28. 28. Odds FC. Synergy, antagonism, and what the chequerboard puts between them. J Antimicrob Chemother. 2003;52: 1–1. pmid:12805255
  29. 29. Jang Y, Kim J-E, Jeong S-H, Paik M-K, Kim JS, Cho M-H. Trifloxystrobin-induced mitophagy through mitochondrial damage in human skin keratinocytes. J Toxicol Sci. 2016;41: 731–737. pmid:27853101
  30. 30. Hikiji W, Yamaguchi K, Saka K, Hayashida M, Ohno Y, Fukunaga T. Acute fatal poisoning with Tolfenpyrad. J Forensic Leg Med. 2013;20: 962–964. pmid:24237799
  31. 31. Wall G, Herrera N, Lopez-Ribot JL. Repositionable compounds with antifungal activity against multidrug resistant Candida auris identified in the Medicines for Malaria Venture’s Pathogen Box. J Fungi Basel Switz. 2019;5. pmid:31581540
  32. 32. Bernhard GC. Auranofin therapy in rheumatoid arthritis. J Lab Clin Med. 1982;100: 167–177. pmid:6808066
  33. 33. Furst DE. Mechanism of action, pharmacology, clinical efficacy and side effects of auranofin: an orally administered organic gold compound for the treatment of rheumatoid arthritis. Pharmacother J Hum Pharmacol Drug Ther. 1983;3: 284–296. pmid:6417628
  34. 34. Fuchs BB, RajaMuthiah R, Souza ACR, Eatemadpour S, Rossoni RD, Santos DA, et al. Inhibition of bacterial and fungal pathogens by the orphaned drug auranofin. Future Med Chem. 2016;8: 117–132. pmid:26808006
  35. 35. Thangamani S, Maland M, Mohammad H, Pascuzzi PE, Avramova L, Koehler CM, et al. Repurposing approach identifies auranofin with broad spectrum antifungal activity that targets Mia40-Erv1 pathway. Front Cell Infect Microbiol. 2017;7: 4. pmid:28149831
  36. 36. Wiederhold NP, Patterson TF, Srinivasan A, Chaturvedi AK, Fothergill AW, Wormley FL, et al. Repurposing auranofin as an antifungal: In vitro activity against a variety of medically important fungi. Virulence. 2017;8: 138–142. pmid:27268469
  37. 37. de Hoog GS, Nishikaku AS, Fernandez-Zeppenfeldt G, Padín-González C, Burger E, Badali H, et al. Molecular analysis and pathogenicity of the Cladophialophora carrionii complex, with the description of a novel species. Stud Mycol. 2007;58: 219–234. pmid:18491001
  38. 38. Marie C, Petri WA. Amoebic dysentery. BMJ Clin Evid. 2013;2013.
  39. 39. Burnett BP, Mitchell CM. Antimicrobial activity of iodoquinol 1%-hydrocortisone acetate 2% gel against ciclopirox and clotrimazole. Cutis. 2008;82: 273–280. pmid:19055171
  40. 40. Liu C, Cui Z, Yan X, Qi Z, Ji M, Li X. Synthesis, fungicidal activity and mode of action of 4-Phenyl-6-trifluoromethyl-2-aminopyrimidines against Botrytis cinerea. Mol Basel Switz. 2016;21. pmid:27347910
  41. 41. Liu H-B, Gao W-W, Tangadanchu VKR, Zhou C-H, Geng R-X. Novel aminopyrimidinyl benzimidazoles as potentially antimicrobial agents: Design, synthesis and biological evaluation. Eur J Med Chem. 2018;143: 66–84. pmid:29172083
  42. 42. Capasso C, Supuran CT. Bacterial, fungal and protozoan carbonic anhydrases as drug targets. Expert Opin Ther Targets. 2015;19: 1689–1704. pmid:26235676
  43. 43. Kucerova-Chlupacova M, Kunes J, Buchta V, Vejsova M, Opletalova V. Novel pyrazine analogs of chalcones: synthesis and evaluation of their antifungal and antimycobacterial activity. Mol Basel Switz. 2015;20: 1104–1117. pmid:25587786
  44. 44. Borba-Santos LP, Gagini T, Ishida K, de Souza W, Rozental S. Miltefosine is active against Sporothrix brasiliensis isolates with in vitro low susceptibility to amphotericin B or itraconazole. J Med Microbiol. 2015;64: 415–422. pmid:25681323