Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Rep ; 41(11): 111835, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516783

ABSTRACT

As opposed to de novo mutation, ß-lactam resistance in S. pneumoniae is often conferred via homologous recombination during horizontal gene transfer. We hypothesize that ß-lactam resistance in pathogenic streptococci is restricted to naturally competent species via intra-/interspecies recombination due to in vivo fitness trade-offs of de novo penicillin-binding protein (PBP) mutations. We show that de novo mutant populations have abrogated invasive disease capacity and are difficult to evolve in vivo. Conversely, serially transformed recombinant strains efficiently integrate resistant oral streptococcal DNA, gain penicillin resistance and tolerance, and retain virulence in mice. Large-scale changes in pbp2X, pbp2B, and non-PBP-related genes occur in recombinant isolates. Our results indicate that horizontal transfer of ß-lactam resistance engenders initially favorable or minimal cost changes in vivo compared with de novo mutation(s), underscoring the importance of recombination in the emergence of ß-lactam resistance and suggesting why some pathogenic streptococci lacking innate competence remain universally susceptible.


Subject(s)
Pneumococcal Infections , Streptococcus pneumoniae , Mice , Animals , Streptococcus pneumoniae/genetics , Gene Transfer, Horizontal , Virulence/genetics , Microbial Sensitivity Tests , beta-Lactam Resistance/genetics , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Mutation/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism
2.
Nat Commun ; 13(1): 3165, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672367

ABSTRACT

Detailed knowledge on how bacteria evade antibiotics and eventually develop resistance could open avenues for novel therapeutics and diagnostics. It is thereby key to develop a comprehensive genome-wide understanding of how bacteria process antibiotic stress, and how modulation of the involved processes affects their ability to overcome said stress. Here we undertake a comprehensive genetic analysis of how the human pathogen Streptococcus pneumoniae responds to 20 antibiotics. We build a genome-wide atlas of drug susceptibility determinants and generated a genetic interaction network that connects cellular processes and genes of unknown function, which we show can be used as therapeutic targets. Pathway analysis reveals a genome-wide atlas of cellular processes that can make a bacterium less susceptible, and often tolerant, in an antibiotic specific manner. Importantly, modulation of these processes confers fitness benefits during active infections under antibiotic selection. Moreover, screening of sequenced clinical isolates demonstrates that mutations in genes that decrease antibiotic sensitivity and increase tolerance readily evolve and are frequently associated with resistant strains, indicating such mutations could be harbingers for the emergence of antibiotic resistance.


Subject(s)
Anti-Bacterial Agents , Streptococcus pneumoniae , Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial , Drug Tolerance , Humans , Microbial Sensitivity Tests
3.
Front Microbiol ; 11: 1543, 2020.
Article in English | MEDLINE | ID: mdl-32714314

ABSTRACT

Streptococcus pneumoniae is an opportunistic pathogen responsible for widespread illness and is a major global health issue for children, the elderly, and the immunocompromised population. Pneumolysin (PLY) is a cholesterol-dependent cytolysin (CDC) and key pneumococcal virulence factor involved in all phases of pneumococcal disease, including transmission, colonization, and infection. In this review we cover the biology and cytolytic function of PLY, its contribution to S. pneumoniae pathogenesis, and its known interactions and effects on the host with regard to tissue damage and immune response. Additionally, we review statins as a therapeutic option for CDC toxicity and PLY toxoid as a vaccine candidate in protein-based vaccines.

4.
J Antimicrob Chemother ; 75(2): 257-270, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31603213

ABSTRACT

Candida albicans is an opportunistic yeast and the major human fungal pathogen in the USA, as well as in many other regions of the world. Infections with C. albicans can range from superficial mucosal and dermatological infections to life-threatening infections of the bloodstream and vital organs. The azole antifungals remain an important mainstay treatment of candidiasis and therefore the investigation and understanding of the evolution, frequency and mechanisms of azole resistance are vital to improving treatment strategies against this organism. Here the organism C. albicans and the genetic changes and molecular bases underlying the currently known resistance mechanisms to the azole antifungal class are reviewed, including up-regulated expression of efflux pumps, changes in the expression and amino acid composition of the azole target Erg11 and alterations to the organism's typical sterol biosynthesis pathways. Additionally, we update what is known about activating mutations in the zinc cluster transcription factor (ZCF) genes regulating many of these resistance mechanisms and review azole import as a potential contributor to azole resistance. Lastly, investigations of azole tolerance in C. albicans and its implicated clinical significance are reviewed.


Subject(s)
Antifungal Agents/pharmacology , Azoles/pharmacology , Candida albicans , Candidiasis , Drug Resistance, Fungal , Antifungal Agents/therapeutic use , Azoles/therapeutic use , Candida albicans/drug effects , Candida albicans/genetics , Candidiasis/drug therapy , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Humans , Microbial Sensitivity Tests , Transcription Factors/genetics
5.
Article in English | MEDLINE | ID: mdl-31383660

ABSTRACT

VT-1161 and VT-1598 are promising investigational tetrazole antifungals that have shown in vitro and in vivo activity against Candida and other fungi. Candida glabrata is a problematic opportunistic pathogen that is associated with high mortality in invasive infection, as well as both intrinsic and rapidly acquired antifungal resistance. The MICs of VT-1161 and VT-1598 were determined by CLSI methodology to evaluate their in vitro activities against clinical C. glabrata isolates and strains containing individual deletions of the zinc cluster transcription factor genes PDR1 and UPC2A as well as the efflux transporter genes CDR1, PDH1, and SNQ2 Overall, both tetrazoles demonstrated relative activities comparable to those of the tested triazole antifungals against clinical C. glabrata isolates (MIC range, 0.25 to 2 mg/liter and 0.5 to 2 µg/ml for VT-1161 and VT-1598, respectively). Deletion of the PDR1 gene in fluconazole-resistant matched clinical isolate SM3 abolished the decreased susceptibility phenotype completely for both VT-1161 and VT-1598, similarly to the triazoles. UPC2A deletion also increased susceptibility to both triazoles and tetrazoles but to a lesser extent than PDR1 deletion. Of the three major transporter genes regulated by Pdr1, CDR1 deletion resulted in the largest MIC reductions for all agents tested, while PDH1 and SNQ2 deletion individually impacted MICs very little. Overall, both VT-1161 and VT-1598 have comparable activities to those of the available triazoles, and decreased susceptibility to these tetrazoles in C. glabrata is driven by many of the same known resistance mechanisms.


Subject(s)
Antifungal Agents/pharmacology , Candida glabrata/drug effects , Pyridines/pharmacology , Tetrazoles/pharmacology , Candida glabrata/genetics , Candida glabrata/metabolism , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Microbial Sensitivity Tests , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Article in English | MEDLINE | ID: mdl-30858206

ABSTRACT

The increasing incidence of and high mortality rates associated with invasive fungal infections (IFIs) impose an enormous clinical, social, and economic burden on humankind. In addition to microbiological resistance to existing antifungal drugs, the large number of unexplained treatment failures is a serious concern. Due to the extremely limited therapeutic options available, it is critical to identify and understand the various causes of treatment failure if patient outcomes are to improve. In this study, we examined one potential source of treatment failure: antagonistic drug interactions. Using a simple screen, we systematically identified currently approved medications that undermine the antifungal activity of three major antifungal drugs-fluconazole, caspofungin, and amphotericin B-on four prevalent human fungal pathogens-Candida albicans, Candida glabrata, Candida parapsilosis, and Candida tropicalis This revealed that a diverse collection of structurally distinct drugs exhibit antagonistic interactions with fluconazole. Several antagonistic agents selected for follow-up studies induce azole resistance through a mechanism that depends on Tac1p/Pdr1p zinc-cluster transcription factors, which activate the expression of drug efflux pumps belonging to the ABC-type transporter family. Few antagonistic interactions were identified with caspofungin or amphotericin B, possibly reflecting their cell surface mode of action that should not be affected by drug efflux mechanisms. Given that patients at greatest risk of IFIs usually receive a multitude of drugs to treat various underlying conditions, these studies suggest that chemically inducible azole resistance may be much more common and important than previously realized.


Subject(s)
Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Candida/drug effects , Azoles/pharmacology , Drug Resistance, Fungal , Echinocandins/pharmacology , Haloperidol/pharmacology , Humans , Morpholines/pharmacology
7.
Article in English | MEDLINE | ID: mdl-30833425

ABSTRACT

Mutations in genes encoding zinc cluster transcription factors (ZCFs) such as TAC1, MRR1, and UPC2 play a key role in Candida albicans azole antifungal resistance. Artificial activation of the ZCF Mrr2 has shown increased expression of the gene encoding the Cdr1 efflux pump and resistance to fluconazole. Amino acid substitutions in Mrr2 have recently been reported to contribute to fluconazole resistance in clinical isolates. In the present study, 57 C. albicans clinical isolates with elevated fluconazole MICs were examined for mutations in MRR2 and expression of CDR1 Mutations in MRR2 resulting in 15 amino acid substitutions were uniquely identified among resistant isolates, including 4 substitutions (S466L, A468G, S469T, T470N) previously reported to reduce fluconazole susceptibility. Three additional, novel amino acid substitutions (R45Q, A459T, V486M) were also discovered in fluconazole-resistant isolates. When introduced into a fluconazole-susceptible background, no change in fluconazole MIC or CDR1 expression was observed for any of the mutations found in this collection. However, introduction of an allele leading to artificial activation of Mrr2 increased resistance to fluconazole as well as CDR1 expression. Moreover, Mrr2 amino acid changes reported previously to have the strongest effect on fluconazole susceptibility and CDR1 expression also exhibited no differences in fluconazole susceptibility or CDR1 expression relative to the parent strain. While all known fluconazole resistance mechanisms are represented within this collection of clinical isolates and contribute to fluconazole resistance to different extents, mutations in MRR2 do not appear to alter CDR1 expression or contribute to resistance in any of these isolates.


Subject(s)
Candida albicans/drug effects , Candida albicans/metabolism , Fluconazole/pharmacology , Fungal Proteins/metabolism , Transcription Factors/metabolism , Azoles/pharmacology , Candida albicans/genetics , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Gene Expression Regulation, Fungal/genetics , Microbial Sensitivity Tests , Mutation/genetics , Transcription Factors/genetics
8.
Article in English | MEDLINE | ID: mdl-30910896

ABSTRACT

The fungal Cyp51-specific inhibitors VT-1161 and VT-1598 have emerged as promising new therapies to combat fungal infections, including Candida spp. To evaluate their in vitro activities compared to other azoles, MICs were determined by Clinical and Laboratory Standards Institute (CLSI) method for VT-1161, VT-1598, fluconazole, voriconazole, itraconazole, and posaconazole against 68 C. albicans clinical isolates well characterized for azole resistance mechanisms and mutant strains representing individual azole resistance mechanisms. VT-1161 and VT-1598 demonstrated potent activity (geometric mean MICs ≤0.15 µg/ml) against predominantly fluconazole-resistant (≥8 µg/ml) isolates. However, five of 68 isolates exhibited MICs greater than six dilutions (>2 µg/ml) to both tetrazoles compared to fluconazole-susceptible isolates. Four of these isolates likewise exhibited high MICs beyond the upper limit of the assay for all triazoles tested. A premature stop codon in ERG3 likely explained the high-level resistance in one isolate. VT-1598 was effective against strains with hyperactive Tac1, Mrr1, and Upc2 transcription factors and against most ERG11 mutant strains. VT-1161 MICs were elevated compared to the control strain SC5314 for hyperactive Tac1 strains and two strains with Erg11 substitutions (Y132F and Y132F&K143R) but showed activity against hyperactive Mrr1 and Upc2 strains. While mutations affecting Erg3 activity appear to greatly reduce susceptibility to VT-1161 and VT-1598, the elevated MICs of both tetrazoles for four isolates could not be explained by known azole resistance mechanisms, suggesting the presence of undescribed resistance mechanisms to triazole- and tetrazole-based sterol demethylase inhibitors.


Subject(s)
Antifungal Agents/pharmacology , Azoles/pharmacology , Candida albicans/drug effects , Drug Resistance, Fungal/drug effects , Pyridines/pharmacology , Tetrazoles/pharmacology , Candida albicans/genetics , Drug Resistance, Fungal/genetics , Fluconazole/pharmacology , Fungal Proteins/genetics , Humans , Microbial Sensitivity Tests/methods , Mutation/genetics , Transcription Factors/genetics
9.
Article in English | MEDLINE | ID: mdl-30783005

ABSTRACT

Recombinant Candida albicans CYP51 (CaCYP51) proteins containing 23 single and 5 double amino acid substitutions found in clinical strains and the wild-type enzyme were expressed in Escherichia coli and purified by Ni2+-nitrilotriacetic acid agarose chromatography. Catalytic tolerance to azole antifungals was assessed by determination of the concentration causing 50% enzyme inhibition (IC50) using CYP51 reconstitution assays. The greatest increase in the IC50 compared to that of the wild-type enzyme was observed with the five double substitutions Y132F+K143R (15.3-fold), Y132H+K143R (22.1-fold), Y132F+F145L (10.1-fold), G307S+G450E (13-fold), and D278N+G464S (3.3-fold). The single substitutions K143R, D278N, S279F, S405F, G448E, and G450E conferred at least 2-fold increases in the fluconazole IC50, and the Y132F, F145L, Y257H, Y447H, V456I, G464S, R467K, and I471T substitutions conferred increased residual CYP51 activity at high fluconazole concentrations. In vitro testing of select CaCYP51 mutations in C. albicans showed that the Y132F, Y132H, K143R, F145L, S405F, G448E, G450E, G464S, Y132F+K143R, Y132F+F145L, and D278N+G464S substitutions conferred at least a 2-fold increase in the fluconazole MIC. The catalytic tolerance of the purified proteins to voriconazole, itraconazole, and posaconazole was far lower and limited to increased residual activities at high triazole concentrations for certain mutations rather than large increases in IC50 values. Itraconazole was the most effective at inhibiting CaCYP51. However, when tested against CaCYP51 mutant strains, posaconazole seemed to be the most resistant to changes in MIC as a result of CYP51 mutation compared to itraconazole, voriconazole, or fluconazole.


Subject(s)
Antifungal Agents/pharmacology , Azoles/pharmacology , Candida albicans/drug effects , Sterol 14-Demethylase/metabolism , Amino Acid Sequence , Candida albicans/genetics , Fluconazole/pharmacology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Itraconazole/pharmacology , Mutation/genetics , Protein Binding/drug effects , Protein Binding/genetics , Sterol 14-Demethylase/genetics , Triazoles/pharmacology , Voriconazole/pharmacology
10.
Article in English | MEDLINE | ID: mdl-30718246

ABSTRACT

Candida auris has rapidly emerged as a health care-associated and multidrug-resistant pathogen of global concern. In this work, we examined the relative expression of the four C. auris genes with the highest degree of homology to Candida albicansCDR1 and MDR1 among three triazole-resistant clinical isolates as compared to the triazole-susceptible genome reference clinical isolate. We subsequently utilized a novel Cas9-mediated system for genetic manipulations to delete C. aurisCDR1 and MDR1 in both a triazole-resistant clinical isolate and a susceptible reference strain and observed that MICs for all clinically available triazoles decreased as much as 128-fold in the CDR1 deletion strains. The findings of this work reveal for the first time that C. aurisCDR1 and MDR1 are more highly expressed among triazole-resistant clinical isolates of C. auris and that the overexpression of CDR1 is a significant contributor to clinical triazole resistance.


Subject(s)
Antifungal Agents/pharmacology , Candida/drug effects , Candida/genetics , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , CRISPR-Associated Protein 9/genetics , Candida/isolation & purification , Candidiasis/microbiology , Drug Resistance, Fungal/drug effects , Fluconazole/pharmacology , Gene Deletion , Gene Expression Regulation, Fungal , Humans , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Microorganisms, Genetically-Modified , Triazoles/pharmacology
11.
mBio ; 9(3)2018 05 22.
Article in English | MEDLINE | ID: mdl-29789366

ABSTRACT

Inactivation of sterol Δ5,6-desaturase (Erg3p) in the prevalent fungal pathogen Candida albicans is one of several mechanisms that can confer resistance to the azole antifungal drugs. However, loss of Erg3p activity is also associated with deficiencies in stress tolerance, invasive hyphal growth, and attenuated virulence in a mouse model of disseminated infection. This may explain why relatively few erg3-deficient strains have been reported among azole-resistant clinical isolates. In this study, we examined the consequences of Erg3p inactivation upon C. albicans pathogenicity and azole susceptibility in mouse models of mucosal and disseminated infection. While a C. albicanserg3Δ/Δ mutant was unable to cause lethality in the disseminated model, it induced pathology in a mouse model of vaginal infection. The erg3Δ/Δ mutant was also more resistant to fluconazole treatment than the wild type in both models of infection. Thus, complete loss of Erg3p activity confers azole resistance but also niche-specific virulence deficiencies. Serendipitously, we discovered that loss of azole-inducible ERG3 transcription (rather than complete inactivation) is sufficient to confer in vitro fluconazole resistance, without compromising C. albicans stress tolerance, hyphal growth, or pathogenicity in either mouse model. It is also sufficient to confer fluconazole resistance in the mouse vaginal model, but not in the disseminated model of infection, and thus confers niche-specific azole resistance without compromising C. albicans pathogenicity at either site. Collectively, these results establish that modulating Erg3p expression or activity can have niche-specific consequences on both C. albicans pathogenicity and azole resistance.IMPORTANCE While conferring resistance to the azole antifungals in vitro, loss of sterol Δ5,6-desaturase (Erg3p) activity has also been shown to reduce C. albicans pathogenicity. Accordingly, it has been presumed that this mechanism may not be significant in the clinical setting. The results presented here challenge this assumption, revealing a more complex relationship between Erg3p activity, azole resistance, C. albicans pathogenicity, and the specific site of infection. Most importantly, we have shown that even modest changes in ERG3 transcription are sufficient to confer azole resistance without compromising C. albicans fitness or pathogenicity. Given that previous efforts to assess the importance of ERG3 as a determinant of clinical azole resistance have focused almost exclusively on detecting null mutants, its role may have been grossly underestimated. On the basis of our results, a more thorough investigation of the contribution of the ERG3 gene to azole resistance in the clinical setting is warranted.


Subject(s)
Antifungal Agents/pharmacology , Azoles/pharmacology , Candida albicans/pathogenicity , Candidiasis/microbiology , Drug Resistance, Fungal , Fungal Proteins/metabolism , Oxidoreductases/metabolism , Trans-Activators/metabolism , Animals , Candida albicans/drug effects , Candida albicans/enzymology , Candida albicans/genetics , Female , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Humans , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Oxidoreductases/genetics , Trans-Activators/genetics , Virulence/drug effects
12.
Article in English | MEDLINE | ID: mdl-28630186

ABSTRACT

Among emerging non-albicans Candida species, Candida parapsilosis is of particular concern as a cause of nosocomial bloodstream infections in neonatal and intensive care unit patients. While fluconazole and echinocandins are considered effective treatments for such infections, recent reports of fluconazole and echinocandin resistance in C. parapsilosis indicate a growing problem. The present study describes a novel mechanism of antifungal resistance in this organism affecting susceptibility to azole and echinocandin antifungals in a clinical isolate obtained from a patient with prosthetic valve endocarditis. Transcriptome analysis indicated differential expression of several genes in the resistant isolate, including upregulation of ergosterol biosynthesis pathway genes ERG2, ERG5, ERG6, ERG11, ERG24, ERG25, and UPC2 Whole-genome sequencing revealed that the resistant isolate possessed an ERG3 mutation resulting in a G111R amino acid substitution. Sterol profiles indicated a reduction in sterol desaturase activity as a result of this mutation. Replacement of both mutant alleles in the resistant isolate with the susceptible isolate's allele restored wild-type susceptibility to all azoles and echinocandins tested. Disruption of ERG3 in the susceptible and resistant isolates resulted in a loss of sterol desaturase activity, high-level azole resistance, and an echinocandin-intermediate to -resistant phenotype. While disruption of ERG3 in C. albicans resulted in azole resistance, echinocandin MICs, while elevated, remained within the susceptible range. This work demonstrates that the G111R substitution in Erg3 is wholly responsible for the altered azole and echinocandin susceptibilities observed in this C. parapsilosis isolate and is the first report of an ERG3 mutation influencing susceptibility to the echinocandins.


Subject(s)
Antifungal Agents/pharmacology , Azoles/pharmacology , Candida parapsilosis/drug effects , Candida parapsilosis/genetics , Echinocandins/pharmacology , Oxidoreductases/genetics , Azoles/metabolism , Candida parapsilosis/isolation & purification , Cross Infection/drug therapy , Cross Infection/microbiology , Cross Infection/prevention & control , Drug Resistance, Multiple, Fungal/genetics , Echinocandins/metabolism , Ergosterol/biosynthesis , Ergosterol/genetics , Fungemia/drug therapy , Fungemia/microbiology , Fungemia/prevention & control , Gene Dosage/genetics , Genome, Fungal/genetics , Humans , Microbial Sensitivity Tests , Polymorphism, Single Nucleotide/genetics
13.
Front Microbiol ; 7: 2173, 2016.
Article in English | MEDLINE | ID: mdl-28127295

ABSTRACT

Within the limited antifungal armamentarium, the azole antifungals are the most frequent class used to treat Candida infections. Azole antifungals such as fluconazole are often preferred treatment for many Candida infections as they are inexpensive, exhibit limited toxicity, and are available for oral administration. There is, however, extensive documentation of intrinsic and developed resistance to azole antifungals among several Candida species. As the frequency of azole resistant Candida isolates in the clinical setting increases, it is essential to elucidate the mechanisms of such resistance in order to both preserve and improve upon the azole class of antifungals for the treatment of Candida infections. This review examines azole resistance in infections caused by C. albicans as well as the emerging non-albicans Candida species C. parapsilosis, C. tropicalis, C. krusei, and C. glabrata and in particular, describes the current understanding of molecular basis of azole resistance in these fungal species.

14.
Pharmacotherapy ; 35(11): 1037-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26598096

ABSTRACT

Coinciding with the continually increasing population of immunocompromised patients worldwide, the incidence of invasive fungal infections has grown over the past 4 decades. Unfortunately, infections caused by both yeasts such as Candida and molds such as Aspergillus or Mucorales remain associated with unacceptably high morbidity and mortality. In addition, the available antifungals with proven efficacy in the treatment of these infections remain severely limited. Although previously available second-generation triazole antifungals have significantly expanded the spectrum of the triazole antifungal class, these agents are laden with shortcomings in their safety profiles as well as formulation and pharmacokinetic challenges. Isavuconazole, administered as the prodrug isavuconazonium, is the latest second-generation triazole antifungal to receive U.S. Food and Drug Administration approval. Approved for the treatment of both invasive aspergillosis and invasive mucormycosis, and currently under investigation for the treatment of candidemia and invasive candidiasis, isavuconazole may have therapeutic advantages over its predecessors. With clinically relevant antifungal potency against a broad range of yeasts, dimorphic fungi, and molds, isavuconazole has a spectrum of activity reminiscent of the polyene amphotericin B. Moreover, clinical experience thus far has revealed isavuconazole to be associated with fewer toxicities than voriconazole, even when administered without therapeutic drug monitoring. These characteristics, in an agent available in both a highly bioavailable oral and a ß-cyclodextrin-free intravenous formulation, will likely make isavuconazole a welcome addition to the triazole class of antifungals.


Subject(s)
Antifungal Agents , Nitriles , Pyridines , Triazoles , Antifungal Agents/pharmacokinetics , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Candidemia/drug therapy , Candidiasis, Invasive/drug therapy , Humans , Mucormycosis/drug therapy , Nitriles/pharmacokinetics , Nitriles/pharmacology , Nitriles/therapeutic use , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/therapeutic use , Triazoles/pharmacokinetics , Triazoles/pharmacology , Triazoles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...