Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 164: 273-281, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30597328

ABSTRACT

N,N'-Diaryl-bishydrazones of [1,1'-biphenyl]-3,4'-dicarboxaldehyde, [1,1'-biphenyl]-4,4'-dicarboxaldehyde, and 4,4'-bisacetyl-1,1-biphenyl exhibited excellent antifungal activity against a broad spectrum of filamentous and non-filamentous fungi. These N,N'-diaryl-bishydrazones displayed no antibacterial activity in contrast to previously reported N,N'-diamidino-bishydrazones and N-amidino-N'-aryl-bishydrazones. The leading candidate, 4,4'-bis((E)-1-(2-(4-fluorophenyl)hydrazono)ethyl)-1,1'-biphenyl, displayed less hemolysis of murine red blood cells at concentrations at or below that of a control antifungal agent (voriconazole), was fungistatic in a time-kill study, and possessed no mammalian cytotoxicity and no toxicity with respect to hERG inhibition.


Subject(s)
Antifungal Agents/chemistry , Biphenyl Compounds/pharmacology , Hydrazones/pharmacology , Animals , Antifungal Agents/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/therapeutic use , Drug-Related Side Effects and Adverse Reactions , Erythrocytes/drug effects , Fungicides, Industrial , Hemolysis/drug effects , Hydrazones/chemistry , Hydrazones/therapeutic use , Mice
2.
ACS Infect Dis ; 4(7): 1121-1129, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29714997

ABSTRACT

Here, we describe the preparation and evaluation of α,ß-unsaturated carbonyl derivatives of the bacterial translation inhibiting antibiotic chloramphenicol (CAM). Compared to the parent antibiotic, two compounds containing α,ß-unsaturated ketones (1 and 4) displayed a broader spectrum of activity against a panel of Gram-positive pathogens with a minimum inhibitory concentration range of 2-32 µg/mL. Interestingly, unlike the parent CAM, these compounds do not inhibit bacterial translation. Microscopic evidence and metabolic labeling of a cell wall peptidoglycan suggested that compounds 1 and 4 caused extensive damage to the envelope of Staphylococcus aureus cells by inhibition of the early stage of cell wall peptidoglycan biosynthesis. Unlike the effect of membrane-disrupting antimicrobial cationic amphiphiles, these compounds did not rapidly permeabilize the bacterial membrane. Like the parent antibiotic CAM, compounds 1 and 4 had a bacteriostatic effect on S. aureus. Both compounds 1 and 4 were cytotoxic to immortalized nucleated mammalian cells; however, neither caused measurable membrane damage to mammalian red blood cells. These data suggest that the reported CAM-derived antimicrobial agents offer a new molecular scaffold for development of novel bacterial cell wall biosynthesis inhibiting antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacterial Physiological Phenomena , Cell Wall/drug effects , Cell Wall/metabolism , Chloramphenicol/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Chloramphenicol/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Peptidoglycan/biosynthesis , Protein Biosynthesis/drug effects , Rats , Ribosomes/chemistry , Ribosomes/metabolism , Structure-Activity Relationship
3.
Molecules ; 23(4)2018 Apr 13.
Article in English | MEDLINE | ID: mdl-29652845

ABSTRACT

As the threat associated with fungal infections continues to rise and the availability of antifungal drugs remains a concern, it becomes obvious that the need to bolster the antifungal armamentarium is urgent. Building from our previous findings of tobramycin (TOB) derivatives with antifungal activity, we further investigate the effects of various linkers on the biological activity of these aminoglycosides. Herein, we analyze how thioether, sulfone, triazole, amide, and ether functionalities affect the antifungal activity of alkylated TOB derivatives against 22 Candida, Cryptococcus, and Aspergillus species. We also evaluate their impact on the hemolysis of murine erythrocytes and the cytotoxicity against mammalian cell lines. While the triazole linker appears to confer optimal activity overall, all of the linkers incorporated into the TOB derivatives resulted in compounds that are very effective against the Cryptococcus neoformans species, with MIC values ranging from 0.48 to 3.9 µg/mL.


Subject(s)
Aminoglycosides/chemical synthesis , Aminoglycosides/pharmacology , Antifungal Agents/chemical synthesis , Antifungal Agents/pharmacology , Tobramycin/chemistry , A549 Cells , Aminoglycosides/chemistry , Animals , Antifungal Agents/chemistry , Aspergillus/drug effects , Candida/drug effects , Cell Line , Cryptococcus neoformans/drug effects , Erythrocytes/drug effects , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure
4.
J Med Chem ; 61(1): 158-173, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29256601

ABSTRACT

The extensive use of fluconazole (FLC) and other azole drugs has caused the emergence and rise of azole-resistant fungi. The fungistatic nature of FLC in combination with toxicity concerns have resulted in an increased demand for new azole antifungal agents. Herein, we report the synthesis and antifungal activity of novel alkylated piperazines and alkylated piperazine-azole hybrids, their time-kill studies, their hemolytic activity against murine erythrocytes, as well as their cytotoxicity against mammalian cells. Many of these molecules exhibited broad-spectrum activity against all tested fungal strains, with excellent minimum inhibitory concentration (MIC) values against non-albicans Candida and Aspergillus strains. The most promising compounds were found to be less hemolytic than the FDA-approved antifungal agent voriconazole (VOR). Finally, we demonstrate that the synthetic alkylated piperazine-azole hybrids do not function by fungal membrane disruption, but instead by disruption of the ergosterol biosynthetic pathway via inhibition of the 14α-demethylase enzyme present in fungal cells.


Subject(s)
Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Azoles/chemistry , Piperazines/chemistry , Piperazines/pharmacology , 14-alpha Demethylase Inhibitors/chemistry , 14-alpha Demethylase Inhibitors/metabolism , 14-alpha Demethylase Inhibitors/pharmacology , 14-alpha Demethylase Inhibitors/toxicity , Alkylation , Animals , Antifungal Agents/metabolism , Antifungal Agents/toxicity , Aspergillus/drug effects , Candida albicans/drug effects , Cell Line , Hemolysis/drug effects , Humans , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Piperazines/metabolism , Piperazines/toxicity , Protein Conformation , Sterol 14-Demethylase/chemistry , Sterol 14-Demethylase/metabolism
5.
Bioorg Med Chem ; 26(3): 573-580, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29279242

ABSTRACT

The fungistatic nature and toxicity concern associated with the azole drugs currently on the market have resulted in an increased demand for new azole antifungal agents for which these problematic characteristics do not exist. The extensive use of azoles has resulted in fungal strains capable of resisting the action of these drugs. Herein, we report the synthesis and antifungal activity of novel fluconazole (FLC) analogues with alkyl-, aryl-, cycloalkyl-, and dialkyl-amino substituents. We evaluated their antifungal activity by MIC determination and time-kill assay as well as their safety profile by hemolytic activity against murine erythrocytes as well as cytotoxicity against mammalian cells. The best compounds from our study exhibited broad-spectrum activity against most of the fungal strains tested, with excellent MIC values against a number of clinical isolates. The most promising compounds were found to be less hemolytic than the least hemolytic FDA-approved azole antifungal agent voriconazole (VOR). Finally, we demonstrated that the synthetic alkyl-amino FLC analogues displayed chain-dependent fungal membrane disruption as well as inhibition of ergosterol biosynthesis as possible mechanisms of action.


Subject(s)
Antifungal Agents/pharmacology , Fluconazole/pharmacology , Fungi/drug effects , Animals , Antifungal Agents/chemistry , Antifungal Agents/toxicity , Candida/drug effects , Cell Line , Cell Survival/drug effects , Drug Design , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/metabolism , Fluconazole/chemistry , Fluconazole/toxicity , Hemolysis/drug effects , Mice , Microbial Sensitivity Tests , Structure-Activity Relationship , Voriconazole/pharmacology , Voriconazole/toxicity
6.
ACS Infect Dis ; 4(2): 196-207, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29227087

ABSTRACT

Alkylated aminoglycosides and bisbenzimidazoles have previously been shown to individually display antifungal activity. Herein, we explore for the first time the antifungal activity (in liquid cultures and in biofilms) of ten alkylated aminoglycosides covalently linked to either mono- or bisbenzimidazoles. We also investigate their toxicity against mammalian cells, their hemolytic activity, and their potential mechanism(s) of action (inhibition of fungal ergosterol biosynthetic pathway and/or reactive oxygen species (ROS) production). Overall, many of our hybrids exhibited broad-spectrum antifungal activity. We also found them to be less cytotoxic to mammalian cells and less hemolytic than the FDA-approved antifungal agents amphotericin B and voriconazole, respectively. Finally, we show with our best derivative (8) that the mechanism of action of our compounds is not the inhibition of ergosterol biosynthesis, but that it involves ROS production in yeast cells.


Subject(s)
Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Bisbenzimidazole/chemistry , Framycetin/chemistry , Biofilms/drug effects , Candida albicans/drug effects , Candida albicans/metabolism , Dose-Response Relationship, Drug , Hemolysis , Microbial Sensitivity Tests , Molecular Structure , Reactive Oxygen Species/metabolism , Sterols/chemistry , Time Factors
7.
Eur J Med Chem ; 139: 12-21, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-28797882

ABSTRACT

As the number of hospitalized and immunocompromised patients continues to rise, invasive fungal infections, such as invasive candidiasis and aspergillosis, threaten the life of millions of patients every year. The azole antifungals are currently the most prescribed drugs clinically that display broad-spectrum antifungal activity and excellent oral bioavailability. Yet, the azole antifungals have their own limitations and are unable to meet the challenges associated with increasing fungal infections and the accompanied development of resistance against azoles. Exploring combination therapy that involves the current azoles and another drug has been shown to be a promising strategy. Haloperidol and its derivative, bromperidol, were originally discovered as antipsychotics. Herein, we synthesize and report a series of bromperidol derivatives and their synergistic antifungal interactions in combination with a variety of current azole antifungals against a wide panel of fungal pathogens. We further select two representative combinations and confirm the antifungal synergy by performing time-kill assays. Furthermore, we evaluate the ability of selected combinations to destroy fungal biofilm. Finally, we perform mammalian cytotoxicity assays with the representative combinations against three mammalian cell lines.


Subject(s)
Antifungal Agents/pharmacology , Antipsychotic Agents/pharmacology , Azoles/pharmacology , Fungi/drug effects , Haloperidol/analogs & derivatives , Mycoses/drug therapy , Animals , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antipsychotic Agents/chemical synthesis , Antipsychotic Agents/chemistry , Azoles/chemical synthesis , Azoles/chemistry , Biofilms/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Haloperidol/chemical synthesis , Haloperidol/chemistry , Haloperidol/pharmacology , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
8.
Biochemistry ; 56(34): 4457-4467, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28762729

ABSTRACT

The initiation of the nonribosomal peptide synthetase (NRPS) assembly of the bisintercalator natural product thiocoraline involves key enzymatic steps for AMP activation and carrier protein loading of the starter unit 3-hydroxyquinaldic acid (3HQA). Gene cluster data combined with protein sequence homology analysis originally led us to propose that TioJ could be responsible for the AMP activation step, whereas TioO could act as the thiolation (T) domain, facilitating the transfer of 3HQA to the next NRPS module, TioR. Herein, we confirmed the involvement of TioJ in thiocoraline biosynthesis by tioJ knockout and in vitro activation of 3HQA studies. However, we demonstrated that TioJ-activated 3HQA is not loaded onto the T domain TioO, as originally believed, but instead onto a fatty acid synthase (FAS) acyl carrier protein (ACP) domain FabC, which is located outside of the thiocoraline gene cluster. We showed a strong interaction between TioJ and FabC. By generating TioJ point mutants mimicking the active site of highly homologous enzymes activating different molecules, we showed that the identity of the substrate activated by adenylation domains such as TioJ is not determined by only the active site residues that directly interact with the substrate. The insights gained from these enzymatic transformations are valuable in the efforts toward deciphering the complete biosynthetic pathway of thiocoraline and bisintercalators in general.


Subject(s)
Depsipeptides/biosynthesis , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Depsipeptides/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics
9.
Eur J Med Chem ; 133: 309-318, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28395217

ABSTRACT

Fluconazole (FLC) is the drug of choice when it comes to treat fungal infections such as invasive candidiasis in humans. However, the widespread use of FLC has resulted in the development of resistance to this drug in various fungal strains and, simultaneously has occasioned the need for new antifungal agents. Herein, we report the synthesis of 27 new FLC derivatives along with their antifungal activity against a panel of 13 clinically relevant fungal strains. We also explore their toxicity against mammalian cells, their hemolytic activity, as well as their mechanism of action. Overall, many of our FLC derivatives exhibited broad-spectrum antifungal activity and all compounds displayed an MIC value of <0.03 µg/mL against at least one of the fungal strains tested. We also found them to be less hemolytic and less cytotoxic to mammalian cells than the FDA approved antifungal agent amphotericin B. Finally, we demonstrated with our best derivative that the mechanism of action of our compounds is the inhibition of the sterol 14α-demethylase enzyme involved in ergosterol biosynthesis.


Subject(s)
Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Azoles/chemistry , Azoles/pharmacology , Fungi/drug effects , Mycoses/drug therapy , Alkylation , Animals , Antifungal Agents/adverse effects , Antifungal Agents/chemical synthesis , Azoles/adverse effects , Azoles/chemical synthesis , Candida albicans/drug effects , Candidiasis/drug therapy , Cell Line , Ergosterol/metabolism , Fungi/enzymology , Fungi/metabolism , Hemolysis/drug effects , Humans , Mice , Microbial Sensitivity Tests
10.
Bioorg Med Chem ; 25(1): 58-66, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27769670

ABSTRACT

The emergence of multidrug-resistant bacterial and fungal strains poses a threat to human health that requires the design and synthesis of new classes of antimicrobial agents. We evaluated bis(N-amidinohydrazones) and N-(amidino)-N'-aryl-bishydrazones for their antibacterial and antifungal activities against panels of Gram-positive/Gram-negative bacteria as well as fungi. We investigated their potential to develop resistance against both bacteria and fungi by a multi-step resistance-selection method, explored their potential to induce the production of reactive oxygen species, and assessed their toxicity. In summary, we found that these compounds exhibited broad-spectrum antibacterial and antifungal activities against most of the tested strains with minimum inhibitory concentration (MIC) values ranging from <0.5 to >500µM against bacteria and 1.0 to >31.3µg/mL against fungi; and in most cases, they exhibited either superior or similar antimicrobial activity compared to those of the standard drugs used in the clinic. We also observed minimal emergence of drug resistance to these newly synthesized compounds by bacteria and fungi even after 15 passages, and we found weak to moderate inhibition of the human Ether-à-go-go-related gene (hERG) channel with acceptable IC50 values ranging from 1.12 to 3.29µM. Overall, these studies show that bis(N-amidinohydrazones) and N-(amidino)-N'-aryl-bishydrazones are potentially promising scaffolds for the discovery of novel antibacterial and antifungal agents.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Hydrazones/chemistry , Hydrazones/pharmacology , Bacteria/drug effects , Bacterial Infections/drug therapy , Candida albicans/drug effects , Candidiasis/drug therapy , Cell Line , Drug Discovery , Drug Resistance, Multiple , Fungi/drug effects , Humans , Microbial Sensitivity Tests , Mycoses/drug therapy
11.
Bioorg Med Chem ; 24(16): 3680-6, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27301676

ABSTRACT

The rise and emergence of resistance to antifungal drugs by diverse pathogenic fungal strains have resulted in an increase in demand for new antifungal agents. Various heterocyclic scaffolds with different mechanisms of action against fungi have been investigated in the past. Herein, we report the synthesis and antifungal activities of 18 alkylated mono-, bis-, and trisbenzimidazole derivatives, their toxicities against mammalian cells, as well as their ability to induce reactive oxygen species (ROS) in yeast cells. Many of our bisbenzimidazole compounds exhibited moderate to excellent antifungal activities against all tested fungal strains, with MIC values ranging from 15.6 to 0.975µg/mL. The fungal activity profiles of our bisbenzimidazoles were found to be dependent on alkyl chain length. Our most potent compounds were found to display equal or superior antifungal activity when compared to the currently used agents amphotericin B, fluconazole, itraconazole, posaconazole, and voriconazole against many of the strains tested.


Subject(s)
Antifungal Agents/chemical synthesis , Antifungal Agents/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Antifungal Agents/chemistry , Benzimidazoles/chemistry , Carbon-13 Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Proton Magnetic Resonance Spectroscopy , Reactive Oxygen Species/metabolism , Yeasts/drug effects , Yeasts/metabolism
12.
ChemMedChem ; 11(14): 1507-16, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27334363

ABSTRACT

Invasive fungal infections are on the rise due to an increased population of critically ill patients as a result of HIV infections, chemotherapies, and organ transplantations. Current antifungal drugs are helpful, but are insufficient in addressing the problem of drug-resistant fungal infections. Thus, there is a growing need for novel antimycotics that are safe and effective. The ebselen scaffold has been evaluated in clinical trials and has been shown to be safe in humans. This makes ebselen an attractive scaffold for facile translation from bench to bedside. We recently reported a library of ebselen-inspired ebsulfur analogues with antibacterial properties, but their antifungal activity has not been characterized. In this study, we repurposed ebselen, ebsulfur, and 32 additional ebsulfur analogues as antifungal agents by evaluating their antifungal activity against a panel of 13 clinically relevant fungal strains. The effect of induction of reactive oxygen species (ROS) by three of these compounds was evaluated. Their hemolytic and cytotoxicity activities were also determined using mouse erythrocytes and mammalian cells. The MIC values of these compounds were found to be in the range of 0.02-12.5 µg mL(-1) against the fungal strains tested. Notably, yeast cells treated with our compounds showed an accumulation of ROS, which may further contribute to the growth-inhibitory effect against fungi. This study provides new lead compounds for the development of antimycotic agents.


Subject(s)
Antifungal Agents/pharmacology , Thiazoles/pharmacology , Amphotericin B/pharmacology , Animals , Antifungal Agents/chemistry , Antifungal Agents/toxicity , Aspergillus/drug effects , Candida/drug effects , Fluconazole/pharmacology , HEK293 Cells , Hemolysis , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Itraconazole/pharmacology , Mice , Microbial Sensitivity Tests , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/toxicity , Triazoles/pharmacology , Voriconazole/pharmacology
13.
Bioorg Med Chem ; 24(24): 6298-6306, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27073054

ABSTRACT

Antibiotic resistance is a worldwide problem that needs to be addressed. Staphylococcus aureus is one of the dangerous "ESKAPE" pathogens that rapidly evolve and evade many current FDA-approved antibiotics. Thus, there is an urgent need for new anti-MRSA compounds. Ebselen (also known as 2-phenyl-1,2-benzisoselenazol-3(2H)-one) has shown promising activity in clinical trials for cerebral ischemia, bipolar disorder, and noise-induced hearing loss. Recently, there has been a renewed interest in exploring the antibacterial properties of ebselen. In this study, we synthesized an ebselen-inspired library of 33 compounds where the selenium atom has been replaced by sulfur (ebsulfur derivatives) and evaluated them against a panel of drug-sensitive and drug-resistant S. aureus and non-S. aureus strains. Within our library, we identified three outstanding analogues with potent activity against all S. aureus strains tested (MIC values mostly ⩽2µg/mL), and numerous additional ones with overall very good to good antibacterial activity (1-7.8µg/mL). We also characterized the time-kill analysis, anti-biofilm ability, hemolytic activity, mammalian cytotoxicity, membrane-disruption ability, and reactive oxygen species (ROS) production of some of these analogues.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azoles/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Organoselenium Compounds/pharmacology , Anti-Bacterial Agents/chemistry , Azoles/chemical synthesis , Azoles/chemistry , Dose-Response Relationship, Drug , Isoindoles , Microbial Sensitivity Tests , Molecular Structure , Organoselenium Compounds/chemical synthesis , Organoselenium Compounds/chemistry , Structure-Activity Relationship
14.
Chembiochem ; 17(14): 1328-32, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27128382

ABSTRACT

Nonribosomal peptides synthetases (NRPSs), which are multifunctional mega-enzymes producing many biologically active metabolites, are ideal targets for enzyme engineering. NRPS adenylation domains play a critical role in selecting/activating the amino acids to be transferred to downstream NRPS domains in the biosynthesis of natural products. Both monofunctional and bifunctional A domains interrupted with an auxiliary domain are found in nature. Here, we show that a bifunctional interrupted A domain can be uninterrupted by deleting its methyltransferase auxiliary domain portion to make an active monofunctional enzyme. We also demonstrate that a portion of an auxiliary domain with almost no sequence identity to the original auxiliary domain can be insert into naturally interrupted A domain to develop a new active bifunctional A domain with increased substrate profile. This work shows promise for the creation of new interrupted A domains in engineered NRPS enzymes.


Subject(s)
Peptide Synthases/chemistry , Protein Engineering/methods , Adenosine Monophosphate , Amino Acids/metabolism , Biological Products/chemical synthesis , Methylation , Protein Domains , Substrate Specificity
15.
Sci Rep ; 5: 17070, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26594050

ABSTRACT

Azoles are antifungal drugs used to treat fungal infections such as candidiasis in humans. Their extensive use has led to the emergence of drug resistance, complicating antifungal therapy for yeast infections in critically ill patients. Combination therapy has become popular in clinical practice as a potential strategy to fight resistant fungal isolates. Recently, amphiphilic tobramycin analogues, C12 and C14, were shown to display antifungal activities. Herein, the antifungal synergy of C12 and C14 with four azoles, fluconazole (FLC), itraconazole (ITC), posaconazole (POS), and voriconazole (VOR), was examined against seven Candida albicans strains. All tested strains were synergistically inhibited by C12 when combined with azoles, with the exception of C. albicans 64124 and MYA-2876 by FLC and VOR. Likewise, when combined with POS and ITC, C14 exhibited synergistic growth inhibition of all C. albicans strains, except C. albicans MYA-2876 by ITC. The combinations of FLC-C14 and VOR-C14 showed synergistic antifungal effect against three C. albicans and four C. albicans strains, respectively. Finally, synergism between C12/C14 and POS were confirmed by time-kill and disk diffusion assays. These results suggest the possibility of combining C12 or C14 with azoles to treat invasive fungal infections at lower administration doses or with a higher efficiency.


Subject(s)
Antifungal Agents/pharmacology , Fluconazole/pharmacology , Itraconazole/pharmacology , Tobramycin/pharmacology , Triazoles/pharmacology , Voriconazole/pharmacology , Candida albicans/drug effects , Candida albicans/growth & development , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Fungal/drug effects , Drug Synergism , Drug Therapy, Combination , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Microbial Sensitivity Tests , Species Specificity
16.
J Med Chem ; 58(23): 9124-32, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26592740

ABSTRACT

Cationic amphiphiles derived from aminoglycosides (AGs) have been shown to exhibit enhanced antimicrobial activity. Through the attachment of hydrophobic residues such as linear alkyl chains on the AG backbone, interesting antibacterial and antifungal agents with a novel mechanism of action have been developed. Herein, we report the design and synthesis of seven kanamycin B (KANB) derivatives. Their antibacterial and antifungal activities, along with resistance/enzymatic, hemolytic, and cytotoxicity assays were also studied. Two of these compounds, with a C12 and C14 aliphatic chain attached at the 6″-position of KANB through a thioether linkage, exhibited good antibacterial and antifungal activity, were poorer substrates than KANB for several AG-modifying enzymes, and could delay the development of resistance in bacteria and fungi. Also, they were both relatively less hemolytic than the known membrane targeting antibiotic gramicidin and the known antifungal agent amphotericin B and were not toxic at their antifungal MIC values. Their oxidation to sulfones was also demonstrated to have no effect on their activities. Moreover, they both acted synergistically with posaconazole, an azole currently used in the treatment of human fungal infections.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Kanamycin/analogs & derivatives , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Antifungal Agents/chemical synthesis , Antifungal Agents/toxicity , Bacteria/drug effects , Bacterial Infections/drug therapy , Cell Line , Cell Survival/drug effects , Drug Design , Drug Resistance, Microbial , Fungi/drug effects , Hemolysis/drug effects , Humans , Kanamycin/chemical synthesis , Kanamycin/chemistry , Kanamycin/pharmacology , Kanamycin/toxicity , Mice , Mycoses/drug therapy , Surface-Active Agents/chemical synthesis , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacology , Surface-Active Agents/toxicity
17.
Med Mycol ; 53(8): 837-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26260746

ABSTRACT

Several azoles are widely used to treat human fungal infections. Increasing resistance to these azoles has prompted exploration of their synergistic antifungal activities when combined with other agents. The amphiphilic aminoglycoside, K20, was recently shown to inhibit filamentous fungi, yeasts and heterokonts, but not bacteria. In this study, in vitro synergistic growth inhibition by combinations of K20 and azoles (fluconazole, itraconazole, voriconazole, clotrimazole, or posaconazole) were examined against Candida species and Cryptococcus neoformans. Checkerboard microbroth dilution, time-kill curve, and disk diffusion assays revealed that K20 has synergistic inhibitory activities with all five azoles against C. albicans including azole-resistant C. albicans strains ATCC 64124 and ATCC 10231. Four (fluconazole, itraconazole, clotrimazole, posaconazole) and three (fluconazole, itraconazole, voriconazole) azoles were synergistically inhibitory with K20 against C. lusitaniae and C. tropicalis, respectively. Only posaconazole showed synergy with K20 against two Cryptococcus neoformans strains (90-26 and VR-54). Time-kill curves with azole-resistant C. albicans 64124 and azole-sensitive C. albicans MYA-2876 confirmed the K20-azole synergistic interactions with a ≥ 2 log10 decrease in colony-forming units (CFU)/ml compared with the corresponding azoles alone. These results suggest that combinations of K20 and azoles offer a possible strategy for developing therapies against candidiasis.


Subject(s)
Aminoglycosides/pharmacology , Antifungal Agents/pharmacology , Azoles/pharmacology , Candida/drug effects , Cryptococcus neoformans/drug effects , Drug Synergism , Candida/growth & development , Cryptococcus neoformans/growth & development , Humans , Microbial Sensitivity Tests
18.
Antimicrob Agents Chemother ; 59(8): 4861-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26033722

ABSTRACT

In this study, we investigated the in vitro antifungal activities, cytotoxicities, and membrane-disruptive actions of amphiphilic tobramycin (TOB) analogues. The antifungal activities were established by determination of MIC values and in time-kill studies. Cytotoxicity was evaluated in mammalian cell lines. The fungal membrane-disruptive action of these analogues was studied by using the membrane-impermeable dye propidium iodide. TOB analogues bearing a linear alkyl chain at their 6″-position in a thioether linkage exhibited chain length-dependent antifungal activities. Analogues with C12 and C14 chains showed promising antifungal activities against tested fungal strains, with MIC values ranging from 1.95 to 62.5 mg/liter and 1.95 to 7.8 mg/liter, respectively. However, C4, C6, and C8 TOB analogues and TOB itself exhibited little to no antifungal activity. Fifty percent inhibitory concentrations (IC50s) for the most potent TOB analogues (C12 and C14) against A549 and Beas 2B cells were 4- to 64-fold and 32- to 64-fold higher, respectively, than their antifungal MIC values against various fungi. Unlike conventional aminoglycoside antibiotics, TOB analogues with alkyl chain lengths of C12 and C14 appear to inhibit fungi by inducing apoptosis and disrupting the fungal membrane as a novel mechanism of action. Amphiphilic TOB analogues showed broad-spectrum antifungal activities with minimal mammalian cell cytotoxicity. This study provides novel lead compounds for the development of antifungal drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Tobramycin/pharmacology , Aminoglycosides/metabolism , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Fungi/drug effects , Humans , Microbial Sensitivity Tests/methods
19.
J Org Chem ; 80(9): 4398-411, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25826012

ABSTRACT

Novel fungicides are urgently needed. It was recently reported that the attachment of an octyl group at the O-4″ position of kanamycin B converts this antibacterial aminoglycoside into a novel antifungal agent. To elucidate the structure-activity relationship (SAR) for this phenomenon, a lead compound FG03 with a hydroxyl group replacing the 3″-NH2 group of kanamycin B was synthesized. FG03's antifungal activity and synthetic scheme inspired the synthesis of a library of kanamycin B analogues alkylated at various hydroxyl groups. SAR studies of the library revealed that for antifungal activity the O-4″ position is the optimal site for attaching a linear alkyl chain and that the 3″-NH2 and 6″-OH groups of the kanamycin B parent molecule are not essential for antifungal activity. The discovery of lead compound, FG03, is an example of reviving clinically obsolete drugs like kanamycin by simple chemical modification and an alternative strategy for discovering novel antimicrobials.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Antifungal Agents/chemical synthesis , Kanamycin/chemistry , Surface-Active Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antifungal Agents/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Drug Discovery , Molecular Sequence Data , Structure-Activity Relationship , Surface-Active Agents/chemistry
20.
Front Microbiol ; 5: 671, 2014.
Article in English | MEDLINE | ID: mdl-25538692

ABSTRACT

K20 is a novel amphiphilic antifungal aminoglycoside that is synthetically derived from the antibiotic kanamycin A. Reported here are investigations of K20's antimicrobial activities, cytotoxicity, and fungicidal mechanism of action. In vitro growth inhibitory activities against a variety of human and plant pathogenic yeasts, filamentous fungi, and bacteria were determined using microbroth dilution assays and time-kill curve analyses, and hemolytic and animal cell cytotoxic activities were determined. Effects on Cryptococcus neoformans H-99 infectivity were determined with a preventive murine lung infection model. The antifungal mechanism of action was studied using intact fungal cells, yeast lipid mutants, and small unilamellar lipid vesicles. K20 exhibited broad-spectrum in vitro antifungal activities but not antibacterial activities. Pulmonary, single dose-administration of K20 reduced C. neoformans lung infection rates 4-fold compared to controls. Hemolysis and half-maximal cytotoxicities of mammalian cells occurred at concentrations that were 10 to 32-fold higher than fungicidal MICs. With fluorescein isothiocyanate (FITC), 20-25 mg/L K20 caused staining of >95% of C. neoformans and Fusarium graminearum cells and at 31.3 mg/L caused rapid leakage (30-80% in 15 min) of calcein from preloaded small unilamellar lipid vesicles. K20 appears to be a broad-spectrum fungicide, capable of reducing the infectivity of C. neoformans, and exhibits low hemolytic activity and mammalian cell toxicity. It perturbs the plasma membrane by mechanisms that are lipid modulated. K20 is a novel amphiphilic aminoglycoside amenable to scalable production and a potential lead antifungal for therapeutic and crop protection applications.

SELECTION OF CITATIONS
SEARCH DETAIL
...