Acessibilidade / Reportar erro

Recent developments on the anti-Candida effect of amphotericin B combined with a second drug - a mini-review

Abstract

Invasive Candida infections threaten human health due to the increasing incidence of resistance to the currently available antifungal agents. Amphotericin B (AMB) is the gold standard therapy to treat these infections. Nevertheless, the use of such substance in the clinic is aggravated by its toxicity. Since AMB binds to membrane sterols, it forms pores on human plasma membranes, mainly in kidney cells, leading to nephrotoxicity. The combination of this drug to a second substance could allow for the use of smaller concentrations of AMB, consequently lowering the probability of adverse effects. This mini-review summarizes information regarding an array of substances that enhance AMB antifungal activity. It may be noticed that several of these compounds target plasma membrane. Interestingly, substances approved for human use also presented combinatory anti-Candida activity with AMB. These data reinforce the potential of associating AMB to another drug as a promising therapeutical alternative to treat Candida infections. Further studies, regarding mechanism of action, pharmacokinetics and toxicity parameters must be conducted to confirm the role of these substances as adjuvant agents in candidiasis therapy.

Key words
Amphotericin B; antifungal activity; Candida spp

INTRODUCTION

Candida spp. is a major fungal pathogen that affects human beings. Three main classes of antifungal agents are currently used to treat Candida. infections - azoles, polyenes, and echinocandins -, however these compounds may not be effective against all strains, due to the development of resistance by the fungus (Kotey et al. 2021KOTEY FC, DAYIE NT, TETTEH-UARCOO PB & DONKOR ES. 2021. Candida Bloodstream Infections: Changes in Epidemiology and Increase in Drug Resistance . Infect Dis Res Treat 14: 117863372110269.). Because of the small number of substances available to treat Candida spp. infections and the increasing incidence of resistance to them, high mortality rates of invasive Candida infections can be observed, mainly on immunocompromised individuals. Annualy, more than 400,000 invasive Candida spp. infections occur, with a mortality rate of 46 - 75% (Brown et al. 2012BROWN GD. ET AL. 2012. Hidden Killers: Human Fungal Infections. Sci Transl Med 4: 1-10.). These alarming numbers show that it is imperative the development of new effective treatments against candidiasis.

Amphotericin B (AMB) is a polyene drug widely employed to treat candidiasis, especially when the infection becomes invasive, and visceral leishmaniasis. It is well accepted that this substance binds ergosterol located in the cell membrane, causing the formation of pores and hence the leakage of ions, both in fungi and Leishmania spp. Nonetheless, the mechanism of action of AMB may be more complex. According to Chattopadhyay & Jafurulla (2011)CHATTOPADHYAY A & JAFURULLA M. 2011. A novel mechanism for an old drug: amphotericin B in the treatment of visceral leishmaniasis. Biochem Biophys Res Commun 416: 7-12., the antileishmanial activity of AMB is consequence of its binding to protozoan ergosterol and macrophage cholesterol, avoiding the entry of Leishmania spp. into the macrophages. Anderson et al. (2014)ANDERSON TM ET AL. 2014. Amphotericin B forms an extramembranous and fungicidal sterol sponge. Nat Chem Biol 10: 400-406. proposed that instead of forming an ion channel, AMB originates an extramembranous structure that extracts ergosterol from plasma membrane, therefore killing the microorganisms. Using a confocal fluorescence lifetime imaging microscopy of giant unilamellar vesicles, Grudzinski et al. (2016)GRUDZINSKI W, SAGAN J, WELC R, LUCHOWSKI R & GRUSZECKI WI. 2016. Molecular organization, localization and orientation of antifungal antibiotic amphotericin B in a single lipid bilayer. Sci Rep 6:32780. did not detect these extramembranous structures, but observed that membrane sterols may allow AMB to adopt a vertical orientation within the lipid phase of the membrane, originating an aggregate that promotes osmotic imbalance. In C. albicans, Grela et al. (2019)GRELA E ET AL. 2019. Modes of the antibiotic activity of amphotericin B against Candida albicans. Sci Rep 9: 17029. showed that AMB binds to the cell membrane of budding yeasts, impairs the formation of cell walls, and penetrate into the cytoplasm. Recently, Dong et al. (2021)DONG PT ET AL. 2021. Polarization-sensitive stimulated Raman scattering imaging resolves amphotericin B orientation in Candida membrane. Sci Adv 7: eabd5230. observed by stimulated Raman scattering imaging that AMB molecules are indeed organized vertically in the plasma membrane, parallel to phospholipid acyl chains, corroborating to the ion channel model. Besides affecting membrane permeability, AMB may present a second mechanism of action, related to its ability to induce the production of reactive oxygen species (ROS) within the fungus, damaging essential structures such as the membrane itself, DNA, and mitochondria (Mesa-Arango et al. 2014MESA-ARANGO AC ET AL. 2014. The Production of Reactive Oxygen Species Is a Universal Action Mechanism of Amphotericin B against Pathogenic Yeasts and Contributes to the Fungicidal Effect of This Drug. Antimicrob Agents Chemother 58: 6627-6638.).

Resistance to AMB has already been described, but its incidence is low when compared to azoles and echinocandins. The main disadvantage of administering AMB is its nephrotoxicity, a consequence of the binding of this compound to human cells membrane.

Combinatory approaches have been extensively used in clinical practice to manage infectious diseases. For instance, sulfamethoxazole and trimethoprim, two drugs that inhibit folic acid biosynthesis pathway, are commonly used in association to treat bacterial infections (John 2020JOHN J. 2020. Review the Treatment of Resistant Staphylococcal Infections. F1000Research 9: 1-7.). A strategy that could be approached to diminish AMB toxicity would be administering smaller doses of this drug combined to a second substance. Attempts of combining AMB with antifungal drugs currently used in clinical practice have been made, but the results obtained against Candida spp. were not satisfactory. Scheid et al. (2012)SCHEID LA, MARIO DAN, KUBIÇA TF, SANTURIO JM & ALVES SH. 2012. In vitro activities of antifungal agents alone and in combination against fluconazole-susceptible and -resistant strains of Candida dubliniensis. Braz J Infect Dis 16: 78-81. evaluated the combinatory effect of itraconazole or voriconazole with terbinafine or AMB against C. dubliniensis. Treating the yeasts simultaneously with AMB and azole drugs led to a high degree of indifference and antagonism, precluding the use of these substances combined (Scheid et al. 2012SCHEID LA, MARIO DAN, KUBIÇA TF, SANTURIO JM & ALVES SH. 2012. In vitro activities of antifungal agents alone and in combination against fluconazole-susceptible and -resistant strains of Candida dubliniensis. Braz J Infect Dis 16: 78-81.). Combining AMB to fluconazole, itraconazole, voriconazole, posaconazole, caspofungin, or flucytosine also presented mainly indifferent interactions against C. glabrata (Khalifa et al. 2021KHALIFA HO, MAJIMA H, WATANABE A & KAMEI K. 2021. In vitro characterization of twenty-one antifungal combinations against echinocandin-resistant and -susceptible Candida glabrata. J Fungi 7: 108.). Interestingly, Rosato et al. (2012)ROSATO A ET AL. 2012. In vitro synergy testing of anidulafungin with fluconazole, tioconazole, 5-flucytosine and amphotericin B against some Candida spp. Med Chem 8: 690-698. observed that AMB was synergic with anidulafungin against C. albicans, C. parapsilosis, C. guilliermondii, C. krusei, C. glabrata, and C. tropicalis. Considering the low number of antifungal drugs available to treat candidiasis and the limited efficacy of combining them to AMB, a feasible strategy to overcome Candida spp. infections could be associating AMB to non-antifungal agents. Then, the aim of this mini-review is to provide an overview of the recent developments regarding the anticandidal activity of AMB combined to compounds that are not currently used as antifungal drugs.

MATERIALS AND METHODS

Methods performed to assess the interaction between two drugs

Several assays are available to evaluate the interaction between two drugs, as described below. Initially, in vitro assays are performed to observe whether the compounds interact with each other. A deep knowledge of these assays is essential to correctly interpret the data obtained from the experiments. Incubation time, concentrations of each drug, and type of antifungal activity (fungistatic / fungicidal) must be considered in order to accurately describe the combinatory effect of two compounds.

Among the methodologies that can be carried out in order to evaluate the combined antifungal activity of two substances in vitro, checkerboard stands out as the most commonly performed (Bidaud et al. 2021BIDAUD AL, SCHWARZ P, HERBRETEAU G & DANNAOUI E. 2021. Techniques for the Assessment of in Vitro and in Vivo Antifungal Combinations. J Fungi 7: 1-16.). This assay is conducted in 96-well microplates and consists in a two-dimensional microbroth dilution method. Each column and row of the microplate present a two-fold serial dilution of drugs A and B, respectively. Therefore, each well consists of a combination of different concentrations of drugs A and B. Also, the antifungal effect of each drug alone must be assessed. Then, one column and one row are designated to present only drugs A and B, respectively. After incubation at a specific period, the absorbance of the wells is obtained on a microplate reader, cell growth is measured (Figure 1), and the concentrations that present antifungal activity are determined, such as MIC (minimum inhibitory concentration, i.e., the lowest concentration that completely inhibit cell growth) or IC50 (the concentration that inhibits 50% of cell growth when compared to untreated control). The fractional inhibitory concentration index (FICI) model is often used to evaluate drug interaction. FICI is defined as the sum of the fractional inhibitory concentration (FIC) of each drug, while FIC is the ratio MIC combined / MIC alone. There are different interpretations for FICI values. Usually, synergistic (one drug enhances the activity of another), indifferent (drugs do not enhance each other activity), and antagonistic (one drug decreases the activity of another) interactions are defined by FICI ≤0.5, >0.5 - 4.0, and > 4.0, respectively (Odds 2003ODDS FC. 2003. Synergy, Antagonism, and What the Chequerboard Puts between Them. J Antimicrob Chemother 52: 1-1.). There are some studies that consider an extra type of interaction, namely “additive”, when FICI value is between 0.5 and 1.0 (Trifan et al. 2021TRIFAN A ET AL. 2021. Honokiol and Magnolol: Insights into Their Antidermatophytic Effects. Plants 10(11).). The combinatory antifungal activity may also be determined by adding a specific concentration of drug A to different concentrations of drug B, generally obtained by serial dilution. FICI values can be obtained, but the number of combinations between the drugs is very limited (Fukuda et al. 2019FUKUDA T ET AL. 2019. Nectriatide, a Potentiator of Amphotericin B Activity from Nectriaceae sp. BF-0114. J Nat Prod 82: 2673-2681.). The concentration of drug B that presents synergism with drug A is designated as the combined MIC of this substance.

Figure 1
Checkerboard assay scheme. Drug A is vertically diluted from well A to well G within the columns, while Drug B is horizontally diluted from well 12 to well 2 within the rows. White and yellow wells represent no visible cell growth and visible cell growth, respectively. The purple well indicates the combined concentration of drugs A and B in which the FICI value is 0.500, the usual breakpoint used to classify a drug interaction as synergic. Created with BioRender.com.

Another common method performed to evaluate the interaction between two drugs is the time-kill assay (Bidaud et al. 2021BIDAUD AL, SCHWARZ P, HERBRETEAU G & DANNAOUI E. 2021. Techniques for the Assessment of in Vitro and in Vivo Antifungal Combinations. J Fungi 7: 1-16.). In this experiment, two drugs (alone and combined) are incubated with the cells in a tube. Then, serial dilutions of these systems are carried out, and cells are spread onto the surface of a solid medium. After incubation during predetermined periods, colony forming units (CFU) are counted. If the treatment with the combined drugs decreases or increases cell concentration by ≥ 2log10 CFU/ml (in comparison to the drugs alone), the interaction is classified as synergistic or antagonistic, respectively. Thus, time-kill assay measures the fungicidal effect of drugs combination through time. A disadvantage of this method is that testing several concentrations is laborious and, usually, only one concentration of each compound is tested (Figure 2). In order to overcome the time-consumption of this assay, an end-point measurement can be performed. Cells are incubated with the drugs alone and combined, and after a specific period CFU counting is done. In this case, the antifungal effect of the drugs is not monitored over time.

Figure 2
Time-kill assay scheme. Cells are incubated with the drugs, alone and combined, for predetermined periods, and then are spread onto solid medium to allow colony forming unit (CFU) counting. The result is usually expressed as a graphic that describes cellular concentration over time. Created with BioRender.com.

RESULTS AND DISCUSSION

Amphotericin B associated to natural products

Natural products have been used throughout history as therapeutical agents to treat a wide array of diseases. There are indications that 60.000 years BC Neanderthals might have used flowers for healing purposes (Lietava 1992LIETAVA J. 1992. Medicinal plants in a middle paleolithic grave Shanidar IV? J Ethnopharmacol 35: 263-266.). Nowadays, half of the drugs prescribed in industrialized countries present substances directly or indirectly obtained by natural products (Palmeira-de-Oliveira et al. 2013PALMEIRA-DE-OLIVEIRA A, SILVA BM, PALMEIRA-DE-OLIVEIRA R, MARTINEZ-DE-OLIVEIRA J & SALGUEIRO L. 2013. Are plants extracts a potential therapeutic approach for genital infections? Curr Med Chem 20: 2914-2928.). Due to the extensive production of secondary metabolites, plants stand out as invaluable sources of bioactive compounds, such as the well-known molecules salicylic acid (used to obtain the anti-inflammatory drug acetylsalicylic acid), the analgesic morphine, and the cardiotonic digoxin (Rishton 2008RISHTON GM. 2008. Natural products as a robust source of new drugs and drug leads: past successes and present day issues. Am J Cardiol 101: 43D-49D.). The high biodiversity of plants and their ability of producing several distincts molecules increases the possibility of finding substances capable of inhibiting fungal growth or enhancing the antifungal activity of another drug.

In order to survive in an environment that presents a limited amount of nutrients, bacteria and fungi may produce substances to kill each other (Kong et al. 2010KONG KF, SCHNEPER L & MATHEE K. 2010. Beta-lactam antibiotics: from antibiosis to resistance and bacteriology. APMIS 118: 1-36.). The first observation of this phenomena, known as antibiosis, was made by Fleming in 1929, which allowed the discovery of penicillin. Then, several antimicrobial agents have been extracted from microorganisms, such as bacitracin, streptomycin, polymyxins, and amphotericin B itself. Therefore, besides plants, microorganisms appear as attractive sources of antifungal or adjuvant agents to be used against fungal infections.

Below, this review describes substances obtained from plants or microorganisms that are able to enhance the antifungal activity of amphotericin B against Candida spp.

Amphotericin B associated to substances obtained from plants

Using a panel of 76 C. albicans strains, Zhu et al. (2021)ZHU C ET AL. 2021. Artemisinin Elevates Ergosterol Levels of Candida albicans to Synergise with Amphotericin B against Oral Candidiasis. Int J Antimicrob Agents 58: 106394. observed that artemisinin, an antimalarial drug obtained from Artemisia annua, did not inhibit fungal growth alone. On the other hand, this compound at 25 μg/ml presented synergism against all the tested strains when combined to AMB. Interestingly, this combined activity was also seen in a murine oropharyngeal candidiasis model. Authors observed that artemisinin upregulated the expression of genes related to ergosterol biosynthesis, namely ERG1, ERG3, ERG9, and ERG11, therefore increasing the fungal ergosterol level and the binding of AMB to the plasma membrane (Zhu et al. 2021ZHU C ET AL. 2021. Artemisinin Elevates Ergosterol Levels of Candida albicans to Synergise with Amphotericin B against Oral Candidiasis. Int J Antimicrob Agents 58: 106394.) (Table I).

Table I
Natural products obtained from plants with combined anti-Candida activity with amphotericin B, their active concentrations (alone and combined), the active concentration of amphotericin B combined to these substances and method to assess the combinatory effect.

Yamada et al. (2021)YAMADA N ET AL. 2021. Enhancing the Fungicidal Activity of Amphotericin B via Vacuole Disruption by Benzyl Isothiocyanate, a Cruciferous Plant Constituent. Lett Appl Microbiol 72: 390-398. observed that the viability of C. albicans was significantly reduced after a 3-hour incubation with 50 μM benzyl isothiocyanate (BITC), a natural substance found in Alliaria petiolata. Further, this study verified that combining 0.15 μM (0.139 μg/ml) AMB to 40 μM BITC promoted vacuolar membrane disruption, leading to cell death possibly due to the release of hydrolytic enzymes (Yamada et al. 2021YAMADA N ET AL. 2021. Enhancing the Fungicidal Activity of Amphotericin B via Vacuole Disruption by Benzyl Isothiocyanate, a Cruciferous Plant Constituent. Lett Appl Microbiol 72: 390-398.).

Silva et al. (2020)SILVA D ET AL. 2020. (R)-(+)-β-Citronellol and (S)-(-)-β-Citronellol in Combination with Amphotericin B against Candida spp. Int J Mol Sci 21: 1785. assessed the anti-Candida activity of the monoterpene enantiomers R-(+)-β-citronellol and S-(-)-β-citronellol, and observed MIC values of 32 - 256 μg/ml against C. albicans and C. tropicalis strains. Both compounds presented synergism with AMB against the growth of two C. albicans strains, while indifference was observed when the combinations were tested against C. tropicalis. Considering that in the presence of exogenous ergosterol MIC values of the enantiomers increased 64 - 128 times, it may be hypothesized that the compounds bind to the plasma membrane ergosterol and increase its fluidity, enhancing AMB antifungal activity (Silva et al. 2020SILVA D ET AL. 2020. (R)-(+)-β-Citronellol and (S)-(-)-β-Citronellol in Combination with Amphotericin B against Candida spp. Int J Mol Sci 21: 1785.). More studies need to be conducted to clarify why synergism was not observed against C. tropicalis, and whether the combination display antifungal activity against other Candida species.

The phenolic compounds hydroxychavicol and isoquercitrin showed combined activity with AMB against Candida spp. Himratul-Aznita et al. (2016)HIMRATUL-AZNITA WH, NOR-ZULAILA CO & NURUL-FATIHAH K. 2016. Antifungal Activity of Dual Combination of Hydroxychavicol with Commercialized Agents against Oral Candida Species. SpringerPlus 5. observed that 2 - 8 μg/ml hydroxychavicol presented synergism with AMB against C. albicans, C. parapsilosis, C. dubliniensis, and C. lusitaniae, but the mechanism of the combinatory activity is unclear (Himratul-Aznita et al. 2016HIMRATUL-AZNITA WH, NOR-ZULAILA CO & NURUL-FATIHAH K. 2016. Antifungal Activity of Dual Combination of Hydroxychavicol with Commercialized Agents against Oral Candida Species. SpringerPlus 5.). Kim et al. (2019)KIM S, WOO ER & LEE DG. 2019. Synergistic Antifungal Activity of Isoquercitrin: Apoptosis and Membrane Permeabilization Related to Reactive Oxygen Species in Candida albicans. IUBMB Life 71: 283-292. observed synergism between isoquercitrin and AMB against a C. albicans strain. Combining the compounds promoted intracellular ROS accumulation, increased mitochondrial ROS production, and decreased superoxide dismutase activity. The consequent oxidative stress led to lipid peroxidation and membrane disruption, therefore causing cell death (Kim et al. 2019KIM S, WOO ER & LEE DG. 2019. Synergistic Antifungal Activity of Isoquercitrin: Apoptosis and Membrane Permeabilization Related to Reactive Oxygen Species in Candida albicans. IUBMB Life 71: 283-292.).

The phenolic aldehyde vanillin displayed antifungal activity alone against C. albicans, with MIC of 125 μg/ml. At 62.5 μg/ml, the compound decreased ergosterol levels by approximately 70%. Combining vanillin with AMB presented a remarkable synergistic interaction, with FICI value of 0.1. Vanillin was able to inhibit CaCdr2p, an efflux pump related to fluconazole resistance. Then, a synergistic effect when combined to this antifungal agent was also observed (Saibabu et al. 2020SAIBABU V ET AL. 2020. Vanillin Confers Antifungal Drug Synergism in Candida Albicans by Impeding CaCdr2p Driven Efflux. J Mycol Med 30: 100921.). Nonetheless, the mechanism involved with the interaction between vanillin and AMB needs to be clarified.

The terpenes carvacrol and eugenol also possess anti-Candida activity when combined to AMB. Carvacrol at 125 μg/ml presented antifungal activity alone against 25 strains of C. auris. At 4 μg/ml, carvacrol lowered AMB MIC of sensitive strains from 0.5 μg/ml to 0.25 μg/ml. Moreover, at 8 μg/ml, carvacrol led to a 4-fold decrease of resistant strains AMB MIC (Shaban et al. 2020SHABAN S, PATEL M & AHMAD A. 2020. Improved Efficacy of Antifungal Drugs in Combination with Monoterpene Phenols against Candida Auris. Sci Rep 10: 1-8.). Eugenol presented weaker antifungal activity, with MIC value of 625 μg/ml against C. albicans. Combining 156.3 μg/ml eugenol to 2 μg/ml AMB completely inhibited cell growth. The synergism between the substances was also observed by the time-kill assay. The findings of this study indicate that eugenol blocks Ca2+ channel, impairing its influx and hence promoting ROS production, therefore enhancing AMB antifungal activity and leading the yeast to apoptosis (Khan et al. 2019KHAN SN ET AL. 2019. Synergistic Fungicidal Activity with Low Doses of Eugenol and Amphotericin B against Candida albicans. Biochem Biophys Res Commun 518: 459-464.).

Amphotericin B associated to substances obtained from microorganisms

Teixeira-Santos et al. (2016)TEIXEIRA-SANTOS R ET AL. 2016. Unveiling the Synergistic Interaction between Liposomal Amphotericin B and Colistin. Front Microbiol 7: 1-10. observed that colistin, an antibacterial drug produced by Bacillus colistinus, does not display anti-Candida activity alone, but decreases AMB MIC value by 4 - 8 times. Colistin may enhance AMB activity due to the formation of a stable complex with this antifungal, which may lead to pore formation and membrane disruption (Teixeira-Santos et al. 2016TEIXEIRA-SANTOS R ET AL. 2016. Unveiling the Synergistic Interaction between Liposomal Amphotericin B and Colistin. Front Microbiol 7: 1-10.) (Table II).

Table II
Natural products obtained from microorganisms with combined anti-Candida activity with amphotericin B, their active concentrations (alone and combined), the active concentration of amphotericin B combined to these substances and method to assess the combinatory effect.

Rossi et al. (2020)ROSSI SA ET AL. 2020. “Identification of Off-Patent Drugs That Show Synergism with Amphotericin B or That Present Antifungal Action against Cryptococcus neoformans and Candida Spp. Antimicrob Agents Chemother 64: 1-16. observed that 0.025 - 0.1 mM erythromycin, an antibacterial drug produced by Saccharopolyspora erythraea, enhanced AMB activity against C. albicans, C. glabrata, C. parapsilosis, C. auris, C. tropicalis, and C. krusei. Moreover, the combination erythromycin + AMB did not show toxicity in vitro (against RAW264.7 macrophages) and in vivo (against zebrafish embryos) (Rossi et al. 2020ROSSI SA ET AL. 2020. “Identification of Off-Patent Drugs That Show Synergism with Amphotericin B or That Present Antifungal Action against Cryptococcus neoformans and Candida Spp. Antimicrob Agents Chemother 64: 1-16.). The in vitro efficacy and in vivo safety of this combination point out to the potential of repurposed drugs to be used to treat Candida spp. infections.

Yang et al. (2021)YANG X ET AL. 2021. Study on the Inhibitory Activity and Possible Mechanism of Myriocin on Clinically Relevant Drug-Resistant Candida albicans and Its Biofilms. Biol Pharm Bull 44: 305-315. observed that myriocin, an amino fatty acid produced by certain fungi, such as Myriococcum albomyces, inhibited the growth of 20 Candida strains, including C. albicans, C. tropicalis, C. krusei, C. glabrata, and C. parapsilosis, with MIC values ranging from 0.125 to 4 μg/ml. In the presence of ergosterol or phytosphingosine, myriocin MIC values against a C. albicans strain increased from 0.5 μg/ml to 8 μg/ml and >64 μg/ml, respectively. Using the fluorescent probe propidium iodide, it was observed that myriocin promotes membrane damage. The combination myriocin + AMB presented an additive interaction against C. albicans growth, which may be explained by the action of myriocin on plasma membrane. Although no synergism was observed, it is worth mentioning that combining both substances reduced AMB MIC against one of the tested strains by 128 times (Yang et al. 2021YANG X ET AL. 2021. Study on the Inhibitory Activity and Possible Mechanism of Myriocin on Clinically Relevant Drug-Resistant Candida albicans and Its Biofilms. Biol Pharm Bull 44: 305-315.).

Fukuda et al. (2019)FUKUDA T ET AL. 2019. Nectriatide, a Potentiator of Amphotericin B Activity from Nectriaceae sp. BF-0114. J Nat Prod 82: 2673-2681. isolated the cyclotetrapeptide nectriatide from the fungus Nectriaceae sp., and also synthesized eight derivatives. These compounds did not present antifungal activity alone against C. albicans but, at 8 - 32 μg/ml, decreased AMB MIC from 0.5 μg/ml to 0.031 - 0.25 μg/ml. Using cells from human embryonic kidney (HEK293 cell line), authors observed IC50 values of 15 μg/ml and >128 μg/ml for AMB and nectriatide, respectively. Moreover, 32 μg/ml nectriatide did not change AMB IC50 against these cells, but decreased AMB MIC from 0.5 μg/ml to 0.031 μg/ml against C. albicans, pointing out the selectivity of the combination to this yeast (Fukuda et al. 2019FUKUDA T ET AL. 2019. Nectriatide, a Potentiator of Amphotericin B Activity from Nectriaceae sp. BF-0114. J Nat Prod 82: 2673-2681.). HEK293 lineage is an interesting model of study to evaluate AMB toxicity, since it consists on kidney-derived cells, and the most relevant adverse effect of AMB is nephrotoxicity.

From the rare fungus Pseudophialophora sp., Yagi et al. (2020)YAGI A, UCHIDA R, KOBAYASHI K & TOMODA H. 2020. Polyketide Glycosides Phialotides A to H, New Potentiators of Amphotericin B Activity, Produced by Pseudophialophora sp. BF-0158. J Antibiot 73: 211-223. isolated eight phialotides that do not impair fungal growth alone, but enhance AMB activity against C. albicans. At 4 μg/ml, all the compounds reduced AMB MIC from 0.5 μg/ml to 0.031 μg/ml (Yagi et al. 2020YAGI A, UCHIDA R, KOBAYASHI K & TOMODA H. 2020. Polyketide Glycosides Phialotides A to H, New Potentiators of Amphotericin B Activity, Produced by Pseudophialophora sp. BF-0158. J Antibiot 73: 211-223.). Further studies must be carried out to assess whether these compounds may be used as AMB potentiators.

Uchida et al. (2019)UCHIDA R ET AL. 2019. Simpotentin, a New Potentiator of Amphotericin B Activity against Candida albicans, Produced by Simplicillium minatense FKI-4981. J Antibiot 72: 134-140. purified a novel compound, namely simpotentin, from the fungus Simplicillium minatense. This substance did not inhibit C. albicans growth at the tested concentrations. However, 64 μg/ml simpotentin decreased AMB MIC from 0.5 μg/ml to 0.0625 μg/ml. This compound is structurally related to rhamnolipids. Therefore, it may enhance AMB activity by interacting with plasma membrane. Interestingly, simpotentin led to an 8-fold decrease of AMB MIC against C. albicans, and did not change AMB IC50 against HEK293 cells, showing that the compound potentiate the antifungal activity of AMB without increasing its toxicity (Uchida et al. 2019UCHIDA R ET AL. 2019. Simpotentin, a New Potentiator of Amphotericin B Activity against Candida albicans, Produced by Simplicillium minatense FKI-4981. J Antibiot 72: 134-140.). Ohtawa et al. (2019)OHTAWA M ET AL. 2019. Total Synthesis and Absolute Configuration of Simpotentin, a Potentiator of Amphotericin B Activity. Org Lett 21: 5596-5599. synthesized eight simpotentin stereoisomers and observed that all of them were able to diminish AMB MIC against C. albicans. Data revealed that the isomer (3R,5S,13S) was the most active, decreasing AMB MIC from 0.5 μg/ml to <0.0313 μg/ml (Ohtawa et al. 2019OHTAWA M ET AL. 2019. Total Synthesis and Absolute Configuration of Simpotentin, a Potentiator of Amphotericin B Activity. Org Lett 21: 5596-5599.).

Amphotericin B associated to synthetic substances

The development of the synthetic organic chemistry in the last century allowed the obtention of drugs completely built in laboratories, such as the antifungal agent fluconazole. It is possible to generate a group of derivatives from a core molecule in order to evaluate which compound possesses the greatest pharmacological activity and toxicity profile. A possibility is to evaluate the ability of these derivatives in enhancing the antimicrobial activity of another compound, and compare the results obtained to determine the most active substance. For example, FICI values can be compared between analogue molecules to understand which modifications emerge as the most promising ones. In this context, some studies tested the combinatory effect of synthetic substances and amphotericin B against Candida spp, as depicted below.

Sixteen alkylated tetrahydro-β-carboline derivatives were synthesized from L-Tryptophan by Singh et al. (2019) and had their anti-Candida activity evaluated. One compound, namely 12c, presented antifungal activity alone, with MIC values ranging from 3.67 to 14.8 μg/ml against six Candida species. Moreover, 12c at 0.46 μg/ml presented synergism with AMB against C. glabrata, leading to a 11-fold decrease of AMB MIC. Scanning electron microscopy showed that 12c affects fungal cell surface. Further studies need to be conducted to clarify whether this compound targets plasma membrane and/or cell wall (Singh et al. 2020SINGH R, JAISINGH A, MAURYA IK & SALUNKE DB. 2020. Design, Synthesis and Bio-Evaluation of C-1 Alkylated Tetrahydro-β-Carboline Derivatives as Novel Antifungal Lead Compounds. Bioorganic Med Chem Lett 30: 126869.) (Table III).

Table III
Synthetic compounds with combined anti-Candida activity with amphotericin B, their active concentrations (alone and combined), the active concentration of amphotericin B combined to these substances and method to assess the combinatory effect.

Patil et al. (2020)PATIL M ET AL. 2020. Arginolipid: A Membrane-Active Antifungal Agent and Its Synergistic Potential to Combat Drug Resistance in Clinical Candida Isolates.” Arch Pharm 353: 1-13. synthesized three arginine-based fatty acids named arginolipids, and evaluated their anti-Candida activity against 21 strains of different species. Besides being able to inhibit Candida growth alone, one of the substances (oleoyl arginine ethyl ester, or OAEE) was synergic with fluconazole and AMB against all strains tested. It was observed that OAEE alone depolarizes and damages plasma membrane, which in turn may favor the antifungal activity of fluconazole and AMB. Moreover, complexation between OAEE and AMB may fluidize plasma membrane, enhancing AMB antifungal activity (Patil et al. 2020PATIL M ET AL. 2020. Arginolipid: A Membrane-Active Antifungal Agent and Its Synergistic Potential to Combat Drug Resistance in Clinical Candida Isolates.” Arch Pharm 353: 1-13.).

Lazić et al. (2018)LAZIĆ J ET AL. 2018. Bis-Guanylhydrazones as Efficient Anti-Candida Compounds through DNA Interaction. Appl Microbiol and Biotechnol 102: 1889-1901. synthesized four bis-guanylhydrazones, and observed MIC values from 2 to 15.62 μg/ml against C. albicans, C. glabrata, and C. parapsilosis. BG3, the most active compound, synergized with AMB, but not with nystatin, another polyene drug. Also, BG3 induced ROS production and depolarized mitochondrial membrane potential at high concentrations. These mechanisms could explain the synergism with AMB, however they were not observed at sub-inhibitory concentrations (Lazić et al. 2018LAZIĆ J ET AL. 2018. Bis-Guanylhydrazones as Efficient Anti-Candida Compounds through DNA Interaction. Appl Microbiol and Biotechnol 102: 1889-1901.).

A group of cationic steroidal antimicrobials, namely ceragenins, was synthesized by Bozkurt-Guzel et al. (2018)BOZKURT-GUZEL C, HACIOGLU M & SAVAGE PB. 2018. Investigation of the in Vitro Antifungal and Antibiofilm Activities of Ceragenins CSA-8, CSA-13, CSA-44, CSA-131, and CSA-138 against Candida Species. Diagn Microbiol Infect Dis 91: 324-330., and the anti-Candida activity of five compounds (CSA-8, CSA-13, CSA-44, CSA-131, and CSA-138) was evaluated. All the compounds presented antifungal activity alone, with MIC values ranging from 0.25 μg/ml to 128 μg/ml. Since CSA-8 was the less active compound, it was not tested in combination to AMB. The other four ceragenins presented synergism with AMB against some of the strains tested. For example, the most active compound (CSA-13) was synergic with AMB against 22 out of the 50 strains tested (Bozkurt-Guzel et al. 2018BOZKURT-GUZEL C, HACIOGLU M & SAVAGE PB. 2018. Investigation of the in Vitro Antifungal and Antibiofilm Activities of Ceragenins CSA-8, CSA-13, CSA-44, CSA-131, and CSA-138 against Candida Species. Diagn Microbiol Infect Dis 91: 324-330.). More studies must be conducted to evaluate the applicability of ceragenins either as antifungal or as adjuvant agents in candidiasis treatment.

In a study conducted by Lu et al. (2019)LU Y ET AL. 2019. Fluphenazine Antagonizes with Fluconazole but Synergizes with Amphotericin B in the Treatment of Candidiasis. Appl Microbiol Biotechnol 103: 6701-6709., the antipsychotic drug fluphenazine did not present anti-Candida activity alone at concentrations below 25 μg/ml. However, at 0.78 μg/ml, this substance decreased AMB MIC from 0.25 μg/ml to 0.0625 μg/ml. Treating C. albicans with 3.13 μg/ml fluphenazine for 1 hour increased ERG11 expression, therefore explaining the synergism between this substance and AMB. Interestingly, the combination fluphenazine + AMB downregulated the expression of ALS3 and HWP1, genes that encode surface proteins involved in C. albicans virulence. Using a immunocompromised mouse model of C. albicans disseminated infection, authors observed that combining fluphenazine and AMB enhanced the survival rate and body weight of the animals, and decreased kidney and brain fungal burden (Lu et al. 2019LU Y ET AL. 2019. Fluphenazine Antagonizes with Fluconazole but Synergizes with Amphotericin B in the Treatment of Candidiasis. Appl Microbiol Biotechnol 103: 6701-6709.).

Caldara & Marmiroli (2018)CALDARA M & MARMIROLI N. 2018. Tricyclic Antidepressants Inhibit Candida albicans Growth and Biofilm Formation. Int J Antimicrob Agents 52: 500-505. assessed the anti-Candida activity of the tricyclic antidepressants doxepin, imipramine, and nortriptyline. The MIC values obtained against C. albicans were 200 μg/ml, 40 μg/ml, and 50 μg/ml, respectively. At concentrations that inhibit 10% (MIC10) and 75% (MIC75) of cell growth, imipramine and nortriptyline decreased AMB MIC at 32 - 450 times and 40 - 900 times, respectively, with FICI values ranging from 0.001 to 0.032, pointing out to their potential use as potentiators of AMB activity (Caldara & Marmiroli 2018CALDARA M & MARMIROLI N. 2018. Tricyclic Antidepressants Inhibit Candida albicans Growth and Biofilm Formation. Int J Antimicrob Agents 52: 500-505.).

Faria et al. (2021)FARIA DR ET AL. 2021. Fungicidal Activity of a Safe 1,3,4-Oxadiazole Derivative against Candida albicans. Pathogens 10: 1-19. evaluated the antifungal activity of the synthetic 1,3,4-oxadiazole derivative 4-[cyclohexyl(ethyl)sulfamoyl]N[5-(4-fluorophenyl)-1,3,4-oxadiazol-2-yl]benzamide, also known as LMM6, and observed MIC values of 8 - 32 μg/ml against C. albicans. The combination LMM6 + AMB presented synergistic interaction against C. albicans, with FICI values ranging from 0.531 to 0.75 (Faria et al. 2021FARIA DR ET AL. 2021. Fungicidal Activity of a Safe 1,3,4-Oxadiazole Derivative against Candida albicans. Pathogens 10: 1-19.). It is interesting that results obtained by checkerboard assay may have different interpretations. Results obtained by checkerboard assay may have different interpretations. Based on the FICI values found in this study, interaction would be classified as “additive” or “indifferent” if one consider distinct breakpoints. Therefore, this substance could be discarded even with a FICI of 0.531, that is close to 0.500 (the FICI value that is often used as synergism breakpoint). Thus, assuming strict interpretations based solely on FICI values may not be advised.

Wani et al. (2017)WANI MY, YOUNUS M, AHMAD A, KUMAR S & SOBRAL AJFN. 2017. Flucytosine Analogues Obtained through Biginelli Reaction as Efficient Combinative Antifungal Agents. Microb Pathog 105: 57-62. synthesized four pyrimidinone/thione analogues of the antifungal drug flucytosine, namely BG1, BG2, BG3, and BG4, and obtained MIC values of 1 - 500 μg/ml against eight C. albicans strains. When associated to AMB, BG4 was the most promising substance, since the combination was synergic against 7 out of the 8 strains tested (Wani et al. 2017WANI MY, YOUNUS M, AHMAD A, KUMAR S & SOBRAL AJFN. 2017. Flucytosine Analogues Obtained through Biginelli Reaction as Efficient Combinative Antifungal Agents. Microb Pathog 105: 57-62.). BG4 was also the most active analogue when tested alone. These results show that using old antifungal agents as scaffolds to the development of new ones may be a proper strategy to overcome fungal infections.

Wei et al. (2021)WEI LQ ET AL. 2021. Fingolimod Potentiates the Antifungal Activity of Amphotericin B. Front Cell Infect Microbiol 11: 1-11. assessed the anti-Candida effect of fingolimod, a derivative of the natural product myriocin. This study observed that fingolimod does not inhibit C. albicans growth alone, but enhances AMB activity. Combining 1.38 μg/ml fingolimod with 0.04 μg/ml AMB presented a synergic interaction against C. albicans, with a FICI value of 0.19. This combination also impaired the growth of C. glabrata and C. tropicalis strains. Since myriocin affects Candida plasma membrane, further studies must be conducted to unveil whether fingolimod also targets this structure, in order to clarify the mechanism of synergism with AMB (Wei et al. 2021WEI LQ ET AL. 2021. Fingolimod Potentiates the Antifungal Activity of Amphotericin B. Front Cell Infect Microbiol 11: 1-11.).

The anti-Candida effect of the antihistamine drug promethazine has also been investigated (Brilhante et al. 2018BRILHANTE RSN ET AL. 2018. In Vitro Effects of Promethazine on Cell Morphology and Structure and Mitochondrial Activity of Azole-Resistant Candida tropicalis. Med Mycol 56: 1012-1022.). This substance inhibited C. tropicalis growth alone, with MIC value of 64 μg/ml and, at 8 μg/ml, decreased AMB MIC by 4 to 8 times. This study also observed that 64 μg/ml promethazine damaged plasma membrane and depolarized mitochondria. Although these effects justify the antifungal activity of this drug, the mechanism of synergism with AMB remains unclear, since the combinatory activity was achieved with lower concentrations of promethazine.

Malik et al. (2018)MALIK MA ET AL. 2018. Efficacy of Novel Schiff Base Derivatives as Antifungal Compounds in Combination with Approved Drugs Against Candida albicans. Med Chem 15: 648-658. synthesized and evaluated the antifungal activity of seven novel Schiff base derivatives against ten C. albicans strains. These compounds exhibited MIC values from 6 to 1250 μg/ml. Only 2 out of the 7 substances did not present synergism with AMB against C. albicans. Data show that the compounds at MIC/2 and MIC decreased ergosterol content by 10 - 97%. The most active compound, SB5, may impair ergosterol biosynthesis by binding to lanosterol 14α-demethylase (Malik et al. 2018MALIK MA ET AL. 2018. Efficacy of Novel Schiff Base Derivatives as Antifungal Compounds in Combination with Approved Drugs Against Candida albicans. Med Chem 15: 648-658.). It must be noticed that decreasing ergosterol amount in plasma membrane diminishes AMB affinity but may increase membrane fluidity, enhancing AMB antifungal activity (Mukhopadhyay et al. 2002MUKHOPADHYAY K, KOHLI A & PRASAD R. 2002. Drug Susceptibilities of Yeast Cells Are Affected by Membrane Lipid Composition. Antimicrob Agents Chemother 46: 3695-3705.). Further studies using lower concentrations of the Schiff base derivatives, alone and combined to AMB should be performed to enlighten the mechanisms involved in their combinatory antifungal activity.

CONCLUSION

The development of resistance against azoles and echinocandins by Candida spp. and the adverse effects related to the use of AMB jeopardize antifungal therapy. Then, it is essential that new treatments be created in order to achieve the desired therapeutical success.

Among the compounds described in this study, isoquercitrin, myriocin, alkylated tetrahydro-β-carboline derivatives, and fluphenazine appear as the most promising candidates to be used in combination with AMB, when considered only their active concentrations. Unfortunately, some studies did not report the concentration of the drugs that were synergic with AMB, therefore precluding a more accurate analysis. Moreover, only three studies assessed the toxicity of AMB combined to a second drug. The mechanisms related to the anti-Candida activity of these combinations are mostly related to the plasma membrane, which is also the target of AMB. Therefore, toxicity determination is essential to ensure that the combinations are suitable for clinical use. In vitro experiments may be employed as screen methodologies to choose the less toxic combinations towards mammalian cells, using viability dyes such as tetrazolium salts (Wijesinghe et al. 2021WIJESINGHE ET AL. 2021. Efficacy of true cinnamon (Cinnamomum verum) leaf essential oil as a therapeutic alternative for Candida biofilm infections. Iran J Basic Med Sci 24: 787-795.). Then, in vivo assays should be performed to measure the efficacy and toxicity of the combinations in more complex organisms. Finally, the most promising combinations may be submitted to clinical trials, in order to allow their use to treat fungal infections.

It may be highlighted that artemisinin, colistin, erythromycin, fluphenazine, imipramine, nortriptyline, and promethazine are already approved for human use to treat other diseases. Repositioning “old” substances is an important approach to accelerate drug discovery. However, as aforementioned, clinical trials must be performed to assure the suitability of the combinations as antifungal drugs.

In summary, this review shows that combining AMB to a second substance may be a promising strategy to treat patients with candidiasis. Further studies regarding mechanism of action, pharmacokinetics and toxicity parameters must be conducted to confirm the role of these substances as adjuvant agents in candidiasis therapy.

ACKNOWLEDGMENTS

This study was supported by Universidade Estácio de Sá (UNESA) - Bolsa de Pesquisa Produtividade.

REFERENCES

  • ANDERSON TM ET AL. 2014. Amphotericin B forms an extramembranous and fungicidal sterol sponge. Nat Chem Biol 10: 400-406.
  • BIDAUD AL, SCHWARZ P, HERBRETEAU G & DANNAOUI E. 2021. Techniques for the Assessment of in Vitro and in Vivo Antifungal Combinations. J Fungi 7: 1-16.
  • BOZKURT-GUZEL C, HACIOGLU M & SAVAGE PB. 2018. Investigation of the in Vitro Antifungal and Antibiofilm Activities of Ceragenins CSA-8, CSA-13, CSA-44, CSA-131, and CSA-138 against Candida Species. Diagn Microbiol Infect Dis 91: 324-330.
  • BRILHANTE RSN ET AL. 2018. In Vitro Effects of Promethazine on Cell Morphology and Structure and Mitochondrial Activity of Azole-Resistant Candida tropicalis. Med Mycol 56: 1012-1022.
  • BROWN GD. ET AL. 2012. Hidden Killers: Human Fungal Infections. Sci Transl Med 4: 1-10.
  • CALDARA M & MARMIROLI N. 2018. Tricyclic Antidepressants Inhibit Candida albicans Growth and Biofilm Formation. Int J Antimicrob Agents 52: 500-505.
  • CHATTOPADHYAY A & JAFURULLA M. 2011. A novel mechanism for an old drug: amphotericin B in the treatment of visceral leishmaniasis. Biochem Biophys Res Commun 416: 7-12.
  • DONG PT ET AL. 2021. Polarization-sensitive stimulated Raman scattering imaging resolves amphotericin B orientation in Candida membrane. Sci Adv 7: eabd5230.
  • FARIA DR ET AL. 2021. Fungicidal Activity of a Safe 1,3,4-Oxadiazole Derivative against Candida albicans. Pathogens 10: 1-19.
  • FUKUDA T ET AL. 2019. Nectriatide, a Potentiator of Amphotericin B Activity from Nectriaceae sp. BF-0114. J Nat Prod 82: 2673-2681.
  • GRELA E ET AL. 2019. Modes of the antibiotic activity of amphotericin B against Candida albicans. Sci Rep 9: 17029.
  • GRUDZINSKI W, SAGAN J, WELC R, LUCHOWSKI R & GRUSZECKI WI. 2016. Molecular organization, localization and orientation of antifungal antibiotic amphotericin B in a single lipid bilayer. Sci Rep 6:32780.
  • HIMRATUL-AZNITA WH, NOR-ZULAILA CO & NURUL-FATIHAH K. 2016. Antifungal Activity of Dual Combination of Hydroxychavicol with Commercialized Agents against Oral Candida Species. SpringerPlus 5.
  • JOHN J. 2020. Review the Treatment of Resistant Staphylococcal Infections. F1000Research 9: 1-7.
  • KHALIFA HO, MAJIMA H, WATANABE A & KAMEI K. 2021. In vitro characterization of twenty-one antifungal combinations against echinocandin-resistant and -susceptible Candida glabrata. J Fungi 7: 108.
  • KHAN SN ET AL. 2019. Synergistic Fungicidal Activity with Low Doses of Eugenol and Amphotericin B against Candida albicans. Biochem Biophys Res Commun 518: 459-464.
  • KIM S, WOO ER & LEE DG. 2019. Synergistic Antifungal Activity of Isoquercitrin: Apoptosis and Membrane Permeabilization Related to Reactive Oxygen Species in Candida albicans. IUBMB Life 71: 283-292.
  • KONG KF, SCHNEPER L & MATHEE K. 2010. Beta-lactam antibiotics: from antibiosis to resistance and bacteriology. APMIS 118: 1-36.
  • KOTEY FC, DAYIE NT, TETTEH-UARCOO PB & DONKOR ES. 2021. Candida Bloodstream Infections: Changes in Epidemiology and Increase in Drug Resistance . Infect Dis Res Treat 14: 117863372110269.
  • LAZIĆ J ET AL. 2018. Bis-Guanylhydrazones as Efficient Anti-Candida Compounds through DNA Interaction. Appl Microbiol and Biotechnol 102: 1889-1901.
  • LIETAVA J. 1992. Medicinal plants in a middle paleolithic grave Shanidar IV? J Ethnopharmacol 35: 263-266.
  • LU Y ET AL. 2019. Fluphenazine Antagonizes with Fluconazole but Synergizes with Amphotericin B in the Treatment of Candidiasis. Appl Microbiol Biotechnol 103: 6701-6709.
  • MALIK MA ET AL. 2018. Efficacy of Novel Schiff Base Derivatives as Antifungal Compounds in Combination with Approved Drugs Against Candida albicans. Med Chem 15: 648-658.
  • MESA-ARANGO AC ET AL. 2014. The Production of Reactive Oxygen Species Is a Universal Action Mechanism of Amphotericin B against Pathogenic Yeasts and Contributes to the Fungicidal Effect of This Drug. Antimicrob Agents Chemother 58: 6627-6638.
  • MUKHOPADHYAY K, KOHLI A & PRASAD R. 2002. Drug Susceptibilities of Yeast Cells Are Affected by Membrane Lipid Composition. Antimicrob Agents Chemother 46: 3695-3705.
  • ODDS FC. 2003. Synergy, Antagonism, and What the Chequerboard Puts between Them. J Antimicrob Chemother 52: 1-1.
  • OHTAWA M ET AL. 2019. Total Synthesis and Absolute Configuration of Simpotentin, a Potentiator of Amphotericin B Activity. Org Lett 21: 5596-5599.
  • PALMEIRA-DE-OLIVEIRA A, SILVA BM, PALMEIRA-DE-OLIVEIRA R, MARTINEZ-DE-OLIVEIRA J & SALGUEIRO L. 2013. Are plants extracts a potential therapeutic approach for genital infections? Curr Med Chem 20: 2914-2928.
  • PATIL M ET AL. 2020. Arginolipid: A Membrane-Active Antifungal Agent and Its Synergistic Potential to Combat Drug Resistance in Clinical Candida Isolates.” Arch Pharm 353: 1-13.
  • RISHTON GM. 2008. Natural products as a robust source of new drugs and drug leads: past successes and present day issues. Am J Cardiol 101: 43D-49D.
  • ROSATO A ET AL. 2012. In vitro synergy testing of anidulafungin with fluconazole, tioconazole, 5-flucytosine and amphotericin B against some Candida spp. Med Chem 8: 690-698.
  • ROSSI SA ET AL. 2020. “Identification of Off-Patent Drugs That Show Synergism with Amphotericin B or That Present Antifungal Action against Cryptococcus neoformans and Candida Spp. Antimicrob Agents Chemother 64: 1-16.
  • SAIBABU V ET AL. 2020. Vanillin Confers Antifungal Drug Synergism in Candida Albicans by Impeding CaCdr2p Driven Efflux. J Mycol Med 30: 100921.
  • SCHEID LA, MARIO DAN, KUBIÇA TF, SANTURIO JM & ALVES SH. 2012. In vitro activities of antifungal agents alone and in combination against fluconazole-susceptible and -resistant strains of Candida dubliniensis. Braz J Infect Dis 16: 78-81.
  • SHABAN S, PATEL M & AHMAD A. 2020. Improved Efficacy of Antifungal Drugs in Combination with Monoterpene Phenols against Candida Auris. Sci Rep 10: 1-8.
  • SILVA D ET AL. 2020. (R)-(+)-β-Citronellol and (S)-(-)-β-Citronellol in Combination with Amphotericin B against Candida spp. Int J Mol Sci 21: 1785.
  • SINGH R, JAISINGH A, MAURYA IK & SALUNKE DB. 2020. Design, Synthesis and Bio-Evaluation of C-1 Alkylated Tetrahydro-β-Carboline Derivatives as Novel Antifungal Lead Compounds. Bioorganic Med Chem Lett 30: 126869.
  • TEIXEIRA-SANTOS R ET AL. 2016. Unveiling the Synergistic Interaction between Liposomal Amphotericin B and Colistin. Front Microbiol 7: 1-10.
  • TRIFAN A ET AL. 2021. Honokiol and Magnolol: Insights into Their Antidermatophytic Effects. Plants 10(11).
  • UCHIDA R ET AL. 2019. Simpotentin, a New Potentiator of Amphotericin B Activity against Candida albicans, Produced by Simplicillium minatense FKI-4981. J Antibiot 72: 134-140.
  • WANI MY, YOUNUS M, AHMAD A, KUMAR S & SOBRAL AJFN. 2017. Flucytosine Analogues Obtained through Biginelli Reaction as Efficient Combinative Antifungal Agents. Microb Pathog 105: 57-62.
  • WEI LQ ET AL. 2021. Fingolimod Potentiates the Antifungal Activity of Amphotericin B. Front Cell Infect Microbiol 11: 1-11.
  • WIJESINGHE ET AL. 2021. Efficacy of true cinnamon (Cinnamomum verum) leaf essential oil as a therapeutic alternative for Candida biofilm infections. Iran J Basic Med Sci 24: 787-795.
  • YAGI A, UCHIDA R, KOBAYASHI K & TOMODA H. 2020. Polyketide Glycosides Phialotides A to H, New Potentiators of Amphotericin B Activity, Produced by Pseudophialophora sp. BF-0158. J Antibiot 73: 211-223.
  • YAMADA N ET AL. 2021. Enhancing the Fungicidal Activity of Amphotericin B via Vacuole Disruption by Benzyl Isothiocyanate, a Cruciferous Plant Constituent. Lett Appl Microbiol 72: 390-398.
  • YANG X ET AL. 2021. Study on the Inhibitory Activity and Possible Mechanism of Myriocin on Clinically Relevant Drug-Resistant Candida albicans and Its Biofilms. Biol Pharm Bull 44: 305-315.
  • ZHU C ET AL. 2021. Artemisinin Elevates Ergosterol Levels of Candida albicans to Synergise with Amphotericin B against Oral Candidiasis. Int J Antimicrob Agents 58: 106394.

Publication Dates

  • Publication in this collection
    08 May 2023
  • Date of issue
    2023

History

  • Received
    14 Jan 2022
  • Accepted
    10 May 2022
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br