You are on page 1of 8

[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.

97]

REVIEW ARTICLE

Antifungal Resistance in Dermatology


Varadraj Pai, Ajantha Ganavalli1, Naveen Narayanshetty Kikkeri2

Abstract From the Department of


Cutaneous fungal infections affect more than one‑fourth of world’s population. The pathogenesis Dermatology, Goa Medical College,
and severity of fungal infection depend on various immunological and nonimmunological Bambolim, Goa, Departments of
1
Microbiology and 2Dermatology,
factors. The rampant use of antifungal therapy in immunocompromised individuals marked
SDM College of Medical Sciences
the onset of antifungal drug resistance. Fungal resistance can be microbiological or clinical. and Hospital, Dharwad,
Microbiological resistance depends on various fungal factors which have established due to Karnataka, India
genetic alteration in the fungi. Clinical resistance is due to host‑ or drug‑related factors. All
these factors may cause fungal resistance individually or in tandem. In addition to standardized
susceptibility testing and appropriate drug dosing, one of the ways to avoid resistance is Address for correspondence:
Dr. Varadraj Pai,
the use of combinational antifungal therapy. Combination therapy also offers advantages in
Department of Dermatology,
increased synergistic action with enhanced spectrum activity. Newer insights into mechanisms Goa Medical College,
of drug resistance will help in the development of appropriate antifungal therapy. Bambolim, Goa, India.
E‑mail: docpai@rediffmail.com
Key Words: Antifungal resistance, dermatophytes, mechanisms

Introduction dermatophytes are divided into zoophilic, geophilic,


Fungi have been present for around 1500 million or anthropophilic, depending on their primary
years with more than 1.5 million species out of which habitat (animals, soil, or humans, respectively).
only about 300 species are known to cause human Zoophilic species are responsible for about 30% of human
infections.[1,2] Fungi were recognized earlier than bacteria dermatophytoses and usually cause acute inflammatory
as a pathogenic agent of human disease with David features. Anthropophilic species represent about 70% of
Gruby describing etiological agent of favus, ectothrix, infections on these hosts, causing a chronic infection
and endothrix into three genera, Epidermophyton, of slow progression, suggesting that the fungus has
Microsporum, and Trichophyton.[3] Despite their early adapted to the human host.[8]
discovery, most widespread infectious diseases in the
19th century were attributed to bacterial, parasitic,
Pathogenesis of Fungal Infections
and to viral origins.[4] From the mid‑20th century, The successful initiation of infection is a process closely
the incidence of severe systemic fungal infections related to the capability of the infecting dermatophyte
increased significantly, mainly due to increase in the to overcome the host resistance mechanisms.[5]
number of patients with compromised immune system Factors affecting fungal virulence are:
such as acquired immunodeficiency syndrome (AIDS) a. Defect in normal physiological barriers such as
or postorgan transplantation and chemotherapy. The physical and chemical structure of skin, normal
indiscriminate use of antibiotics added to the worsening microflora present, exposure to ultraviolet light,
of this picture, leading to the installation of fungal temperature, and humidity[6]
infections.[5] b. Adherence of dermatophytes and hyphae
Among all fungal infections, superficial mycoses are penetration – adherence of dermatophytes is due
the most frequent forms of human infections, affecting to arthroconidia, which are genetically programmed
more than 20%–25% of the world’s population.[6] It is disarticulated septate hyphae. It occurs 3–4 hours
also estimated that 30%–70% of adults are asymptomatic
carriers of these pathogens.[7] In addition, species of
This is an open access journal, and articles are distributed under the terms of
the Creative Commons Attribution‑NonCommercial‑ShareAlike 4.0 License,
Access this article online which allows others to remix, tweak, and build upon the work non‑commercially,
Quick Response Code: as long as appropriate credit is given and the new creations are licensed under
the identical terms.
Website: www.e‑ijd.org
For reprints contact: reprints@medknow.com

How to cite this article: Pai V, Ganavalli A, Kikkeri NN. Antifungal


DOI: 10.4103/ijd.IJD_131_17 resistance in dermatology. Indian J Dermatol 2018;63:361-8.
Received: February, 2017. Accepted: June, 2018.

© 2018 Indian Journal of Dermatology | Published by Wolters Kluwer - Medknow 361


[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.97]

Pai, et al.: Antifungal resistance

after contact.[8,9] Hyphae penetration into the stratum Antifungal Resistance


corneum is a result of breakage of disulfide bridges The evolution of antimicrobial drug resistance is an
followed by secretion of variety of enzymes such as almost inevitable process that is universal in the
metalloendoproteases (fungalysins) formerly called microbial world. Although fungal resistance is not at par
keratinases, multiple serine‑subtilisins, proteases, with bacterial resistance, the economic facets associated
lipases, elastases, collagenases, phosphatases, with fungal infections remains unacceptably high.[1]
and esterases.[7,8,10] Reduction of disulfide bridges Also considering the limited number of antifungal drugs
depends on a sulfite efflux pump encoded by the available, one of the main strategies of improving therapy
TruSSU1 gene, a proposed target for new antifungal in mycoses is overcoming antifungal resistance.[12,18]
treatment. Sulfite secretion by this transporter
allows the cleavage of the cystine present in keratin, In general, clinical resistance is considered to be the
thereby making it accessible to the action of endo persistence or progression of an infection despite
and exoproteases.[8,10,11] appropriate antimicrobial therapy.[5,13] Clinical resistance
to antifungals was rare during the early phases of
c. Host response:
antifungal therapy.[14] Among the earliest reported case of
• Nonimmunological factors such as inhibitory
drug resistance of dermatophytes was to griseofulvin by
effect of sebum, unsaturated transferrin,
Michaelides et al. in 1961.[15] Antifungal resistance to azoles
α2‑macroglobulin keratin inhibitor, long‑chain
was first reported in 1980s to ketoconazole used in patients
saturated fatty acids in the scalp, serum inhibitory
of chronic mucocutaneous candidiasis.[13,16] Mukherjee et al.
factors (beta‑globulins, ferritin, and other metal
in 2003 reported primary resistance of Trichophyton rubrum
chelators) binding to iron essential for growth of
to terbinafine in patients with onychomycosis.[17] A study
dermatophytes inhibit fungal invasion. Macerated
of Candida species in patients with AIDS showed 33% of
and hydrated skin, altered skin barrier, and
late‑stage patients with drug‑resistant strains of Candida
immunodeficiency disorders can promote entry of
albicans in their oral cavities.[13,18]
dermatophytes[8,10]
• The immunological mechanism is cell‑mediated, Azole resistance in dermatophytes has been reported to
also more effective and protective, and is mediated be as high as 19% in certain regions of the world.[19]
by macrophages as effector cells and cytokines Occurrence of antifungal resistance has to be considered
secretion from type 1 T‑helper lymphocytes.[10] independently for each antifungal class and for each
d. Fungal response – dermatophytes have developed fungal genus.[14]
mechanisms that allow them to avoid the host Fungal resistance can be:
response such as reduction of inflammation i. Microbiological resistance or in vitro resistance
and phagocytosis by fungal mannans. Mannans ii. Clinical resistance or in vivo resistance.[5,20]
also inhibit the proliferation of keratinocytes,
allowing the establishment of a persistent chronic The factors responsible for fungal resistance are given in
infection.[5,8] Table 1.[5,13,20]

The common antifungal agents and their mechanisms of Microbiological resistance refers to nonsusceptibility
of a fungus to an antifungal agent by in vitro
action depending on the above are given in Figure 1.
susceptibility testing, in which the minimum inhibitory
concentration (MIC) of the drug exceeds the susceptibility
breakpoint for that organism. Microbiological resistance
can be primary (intrinsic), where the fungi are resistant to
a drug before exposure and secondary (acquired), which
develops in response to exposure to an antimicrobial
agent.[13] Certain fungal species are intrinsically resistant
such as Candida krusei to fluconazole and Cryptococcus
neoformans to echinocandins and nonalbicans Candida to
5‑flucytosine (5FC).[13,20]
Secondary resistance develops among previously
susceptible strains after exposure to the antifungal agent
and is usually dependent on altered gene expression,[20]
for example, terbinafine resistance in T. rubrum,
fluconazole resistance among C. albicans.
Clinical resistance is defined as the failure to eradicate a
Figure 1: The common antifungals and their mechanism of action fungal infection despite the administration of an antifungal

362 Indian Journal of Dermatology | Volume 63 | Issue 5 | September-October 2018


[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.97]

Pai, et al.: Antifungal resistance

Table 1: Factors for fungal resistance encoded an ABC transporter was seen following azole
Fungal factors and terbinafine therapy.[5]
Reducing the accumulation of the drug within the fungal cell Decreasing the affinity of the drug for its
Decreasing the affinity of the drug for its target target
Modifications of metabolism to counterbalance the drug effect
A mutation or overexpression of the gene coding for
Cellular response to stress
target enzymes is another mechanism developed by
Biofilm production
fungi.
Host factors
State of immunosuppression A point mutation in the ERG11 gene that codes for
Site of infection lanosterol 14α‑demethylase leads to the complete inhibition
Severity of infection of the binding capacity of the azole drug to its target.[14]
Onset of treatment Mutation in the squalene epoxide (SE) gene (ERG1) leads
Drug related to an amino acid substitution in the SE making the fungi
Fungistatic nature about 1000‑fold less susceptible to terbinafine.[5]
Dosage The overexpression of target enzymes is either by gene
Pharmacokinetics amplification or upregulation of the corresponding
Drug‑drug interactions gene.[5] In chromosomal anomalies like isochromosome
Environmental factors formation, an increase in the number of azole‑resistant
genes can occur.[21]
agent with in vitro activity against the organism. Although
clinical resistance cannot always be predicted, it highlights Modifications of metabolism to
the importance of individualizing treatment strategies on counterbalance the drug effect
the basis of the clinical situation.[20] De novo synthesis of pyrimidines
The antifungal drug 5FC competes with the regular
Fungal factors, host factors, and drug pharmacology play
pyrimidine intermediate metabolites for incorporation
a role in fungal resistance in isolation or in association
into nucleic acids.[14,22] De Novo increase in pyrimidine
with other.
synthesis leads to 5FC resistance as seen in Candida
The fungal molecular mechanism can result from gene glabrata.[14]
amplification, gene transfer, gene deletion, point
mutations, loss of cis‑ and trans‑acting regulatory Paradoxical effect
elements, and transcriptional activation. All these effects Few yeasts and filamentous fungi are able to grow in
could be on genes directly involved in the combat elevated echinocandin concentrations much higher than
against the cytotoxic compounds and/or could be on the MICs. This phenomenon, called paradoxical effect
genes involved in their regulation or processing.[5] or “eagle effect,” is strain dependent and is due to the
upregulation of the chitin synthesis in the fungal cell
The genes responsible for ergosterol synthesis is given in wall after drug administration.[14,20]
Figure 2.[13]
Modifications of the ergosterol biosynthetic
Fungal Factors pathway
Decreased accumulation of drug within fungi The antifungal activity of azole drugs depends on the
Reducing the accumulation of the drug within the fungal depletion of ergosterol from the fungal membrane and
cell is done by increasing the drug efflux mechanism. accumulation of the toxic product 14α‑methyl‑3,6‑diol,
Multidrug efflux transporters are membrane proteins leading to growth arrest. Alteration in the late steps of
found in all living organisms. These proteins bind the ergosterol biosynthetic pathway through inactivation
to a variety of structurally and chemically dissimilar of the ERG3 gene can lead to the total inactivation
compounds and actively extrude them from the cells.[5] of C5 sterol desaturase and also can give rise to
Mutations (upregulation or overexpression) of the genes cross‑resistance to all azole drugs. This mechanism is
encoding these efflux pumps result in decreased identified in C. albicans.[14,20]
accumulation of the drug in the cell.[21] Plasma membrane composition variation
Efflux systems affecting the antifungal drugs vary. A decrease or total absence of ergosterol in the plasma
Common drug efflux systems that modulate azole membrane through mutations in nonessential genes of
resistance are the ATP‑binding cassette (ABC) superfamily the ergosterol is a rare mechanism of resistance among
and the major facilitator superfamily. Overexpression polyene drugs, for example, ERG3 mutation in clinical
of TruMDR1 and TruMDR2 genes in T. rubrum, which isolates of C. albicans, ERG6 mutation in C. glabrata.[14]

Indian Journal of Dermatology | Volume 63 | Issue 5 | September-October 2018 363


[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.97]

Pai, et al.: Antifungal resistance

Figure 2: The genes involved in ergosterol synthesis

Biofilms Clinical Resistance


Biofilms are sessile microbial communities surrounded Clinical resistance depends on a multiple host‑ and
by extracellular polymeric substances with increased drug‑related factors which are as follows:[13,20]
resistance to antimicrobial agents and host defenses. a. Patients with severe degree of immunosuppression
Both T. rubrum and T. mentagrophytes are capable of with invasive fungal infections may not respond to
producing biofilms.[23] antifungals
Biofilms by yeast and certain molds are frequently b. Delay in initiation of adequate dose of antifungal
polymicrobial and are resistant to almost all the currently results in increased chances of treatment failure
used antifungals, with the exceptions of echinocandins c. Fluconazole has better cerebrospinal fluid (CSF)
and lipid formulations of amphotericin B. Biofilm penetration as compared to itraconazole, therefore,
resistance is probably the result of multiple factors such as making it a better choice in treating fungal
an increased expression of efflux pumps, a modification of meningitis. When the site of infection is necrotic
plasma membrane composition, and the drug sequestration with poor blood supply, a debulking surgery is
essential to overcome antifungal treatment resistance
in biofilm‑produced extracellular matrix.[14,21]
d. Compliance in patients requiring long‑term therapies.
Cellular response to stress or stress
adaptation Drug‑Related Factors
Fungi are remarkably adaptive and have numerous Various factors such as the fungistatic nature of most
signal‑transduction pathways to sense and ensure drugs, inappropriate antifungal usage (in cases where
appropriate physiological mechanisms to adapt to the etiological agent is known), treatment with low
environmental stress following exposure to an antifungal antifungal dosage, long duration of treatment, drug
agent.[5,21] Hsp90, Hsp104, ubiquitin, calcineurin, interactions, and the cost of therapy play a role in
esterases, and oxidative‑stress proteins such as fungal resistance.[21] Combination of polyenes and azoles
glutathione synthatase are key cellular regulators critical with other nephrotoxic drugs can result in treatment
for orchestrating cellular responses to drug‑induced failure.[20]
stress, e.g., in T. rubrum following azoles, griseofulvin Environmental resistance
and amphotericin B.[5]
In recent years, the role of environment as a driver
Stress adaptation may not induce clinical resistance but for resistance has become prominent. Fungicidal use
stabilizes the cell in the presence of drug and allows it in agriculture differs hugely between different regions,
to develop more profound resistance mechanisms over with the United States using about a tenth of as much
time that are manifested as clinical resistance.[21] as Europe. Azole‑based fungicides are used in grape and

364 Indian Journal of Dermatology | Volume 63 | Issue 5 | September-October 2018


[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.97]

Pai, et al.: Antifungal resistance

cereal production in European countries. A strong link Pharmacological measures like prudent use of antifungals
has been found between countries where azole‑based in proper dosing and duration form the mainstay of
fungicides are used and the incidence of antifungal therapy. In general, topical agents should be applied
resistance.[21,24] once to twice daily over the lesions and up to at least
2 cm beyond the margins of the lesions for 2–4 weeks
Antifungal susceptibility testing and continued for 1 week after the rash resolves.[41]
Antifungal susceptibility testing (AFST) has undergone Topical allylamines maintain the mycological cure for
considerable change from a nonstandardized procedure longer periods compared to azole drugs.[42] With respect
that generated results lacking clinical utility to a to systemic antifungals, in addition to a proper
standardized procedure. Standardized microdilution‑based dose, duration, and knowledge of pharmacokinetics,
procedures by the Clinical and Laboratory Standards dosage updosing (pulse or boosted oral antifungal)
Institute (CLSI) and the European Committee on and combination antifungals can be tried in recurrent
Antibiotic Susceptibility Testing are universally accepted dermatophytoses. Boosted oral antifungal treatment
for performing AFST[25] The CLSI, formerly the National is designed to target dormant chlamydospores and
Committee on Clinical Laboratory Standards has released arthroconidia to produce sensitive hyphae which are less
four standard methods for antifungal susceptibility refractory to antifungal treatment.[43,44]
testing, namely, M27‑A3 for macrobroth and microtiter
yeast testing, M38‑A2 for microtiter mold testing, M44‑A Combination Antifungal Therapy
for yeast disc diffusion testing, and M51‑P for mold disc The availability of new antifungal agents with unique
diffusion testing.[26-31] mechanisms of action and improved tolerability has
AFST patterns in dermatophytes show individual and widened the possibilities for the use of combination
regional variation in their outcome. Studies on AFST antifungal therapy for difficult‑to‑treat opportunistic
showed that T. rubrum and T. mentagrophytes, which mycoses.[45] Combining two systemic antifungal agents
were the common species isolated, showed a higher has been used in invasive mycoses caused by Candida,
MIC to fluconazole. Although generally itraconazole, Aspergillus and Cryptococcus.[46] In dermatophytoses,
terbinafine had lower MICs, few studies demonstrated combination antifungal therapy has been used, wherein
a higher MIC to itraconazole and terbinafine. Systemic topical and systemic antifungal are combined. Gupta
griseofulvin and topical amorolfine had lower MIC than et  al. used sequential therapy with itraconazole and
fluconazole. A study done by Koga et  al. showed the terbinafine pulse in toenail onychomycosis.[44,47] The
lowest MIC to be with luliconazole.[32‑36] Rex and Pfaller advantages of combination therapy are increased rate and
proposed the “90–60 Rule” which states that infections extent of fungal killing (synergy), enhanced spectrum
caused by isolates that have MICs considered susceptible of activity, and decreased likelihood of resistance or
respond favorably to appropriate therapy approximately tolerance.[45] Azoles can act in a synergic way when
90% of the time, whereas infections caused by isolates combined with terbinafine providing good results against
with MICs considered resistant respond favorably Candida, dimorphic molds, dematiaceous fungi and
approximately 60% of the time.[37] yeasts, such as C. glabrata. Azoles with amphotericin B
have mixed response.[46,49]
Treatment The disadvantages are increased toxicity, increased
Clinically, antifungal resistance may be suspected drug interactions, increased cost, and poorly
in patients with recurrent episodes of infection, standardized methods of testing efficacy of the
unresponsiveness to the first line of therapy, generalized combination.[45] Itraconazole and voriconazole are potent
involvement, or atypical form of presentation with usual inhibitors of cytochrome P450 3A4 and associated
history of similar lesions in the family. with a greater risk of broader range of potentially
severe drug interactions and hepatotoxicity than
One of the important treatment measures in recurrent
fluconazole.[48] At present, checkerboard dilution assay
cutaneous fungal infections is prevention of disease
is the most common laboratory method for evaluating
among family members. Therefore, nonpharmacological antifungal combinations.[45]
measures like good hygiene play an important adjunct
along with medications.[38] Antifungal Drugs Plus Nonantifungal Drugs
Good skin hygiene measures include handwash; clipping Combination of antifungal drugs with nonantimicrobial
of nail; regular bathing and complete drying of the skin; agents such as calcineurin inhibitors (cyclosporine A
use of nonocclusive shoes, absorbent socks, and powder; and tacrolimus), proton‑pump inhibitors, antiarrhythmic
avoidance of sharing of combs, towels, brushes, bedding, agents, cholesterol‑lowering agents, immunomodulators,
and hats; and avoidance of barefoot walk in public antineoplastic drugs, antiparasitic agents, microbial
bathroom.[39,40] metabolites, and human recombinant antibodies has

Indian Journal of Dermatology | Volume 63 | Issue 5 | September-October 2018 365


[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.97]

Pai, et al.: Antifungal resistance

shown beneficial effects.[46,50] Cyclosporine singularly hygiene measures, prudent use of antifungals in proper
is not able to inhibit the fungal growth but increases dosing and duration, appropriate use of susceptibility
susceptibility to fluconazole due to efflux pump deletion testing, usage of older molecules such as griseofulvin or
or alteration of stress response caused by calcineurin topical keratolytics in combination with newer drugs and
during azole therapy.[51] in appropriate dosages and combination therapy with
two systemic antifungals or systemic antifungal with
Studies show that statins are active against Microsporum
topical antifungals and/or topical keratolytics.
canis and T. mentagrophytes. Synergistic interactions
were noticed when simvastatin was combined with The key to managing the widespread nature of this
terbinafine against dermatophytes.[52] emerging problem would involve development of drugs
with newer targets, decreased drug interactions, at
Hemopoietic growth factors, such as granulocyte colony
affordable price, which at this stage appears distant in
stimulating factor or granulocyte‑macrophage colony
our scenario, knowing the resources required and the
stimulating factor and Th1 cytokines have activity on
economics involved. However, newer insight on drug
the antifungal function of phagocytes and the efficacy
resistance mechanisms could lead to advanced treatment
of antifungal agents.[46,53,54]
strategies in managing fungal infections.
Fractional carbon‑dioxide laser therapy combined with
topical antifungals such as terbinafine, amolorfine, or
Financial support and sponsorship
luliconazole was found to be effective in the treatment Nil.
of onychomycosis.[55] Conflicts of interest
There are no conflicts of interest.
Newer Antifungal Drugs
An efficient antifungal should act against a wide range References
of fungi having no or low toxicity to the host.[5] One of 1. Srinivasan A, Lopez‑Ribot JL, Ramasubramanian AK.
the major challenges in developing antifungal drugs lies Overcoming antifungal resistance. Drug Discov Today Technol
in the similarities shared between fungi and their hosts, 2014;11:65‑71.
thereby restricting the target molecules for antifungal 2. Hawksworth DL. The magnitude of fungal diversity:
drugs. Most newer antifungal drugs are designed The 1.5 million species estimate revisited. Mycol Res
2001;105:1422‑32.
considering the following:
3. Weitzman I, Summerbell RC. The dermatophytes. Clin Microbiol
i. Drugs having pharmacological similarity with the older Rev 1995;8:240‑59.
drugs which share the target molecules, with lower 4. Kapil A, editor. Ananthanarayan & Paniker’s Textbook of
MIC level and specific indications (e.g., isavuconazole, Microbiology. 9th ed. Hyderabad: University Press Private
micafungin, luliconazole)[56,57] Limited; 2013. p. 589‑615.
ii. Repurposing of established medications where an old 5. Martinez‑Rossi NM, Peres NT, Rossi A. Antifungal
compound with a known pharmacology is used alone resistance mechanisms in dermatophytes. Mycopathologia
2008;166:369‑83.
or in combination with another drug for a newer
6. Havlickova B, Czaika VA, Friedrich M. Epidemiological trends
indication, for example, calcineurin inhibitors, target
in skin mycoses worldwide. Mycoses 2008;51 Suppl 4:2‑15.
of rapamycin inhibitors, Hsp90 inhibitors, in synergy 7. Nigam PK. Antifungal drugs and resistance: Current concepts.
with azoles[20,50,58,59] Our Dermatol Online 2015;6:212‑21.
iii. Recent advances in fungal genomics and proteomics 8. Peres NT, Maranhão FC, Rossi A, Martinez‑Rossi NM.
have revealed target genes, proteins, or virulence Dermatophytes: Host‑pathogen interaction and antifungal
factors required during infection of the host resistance. An Bras Dermatol 2010;85:657‑67.
tissues by dermatophytes, for example, efflux pump 9. Coelho LM, Aquino‑Ferreira R, Maffei CM, Martinez‑Rossi NM.
inhibitors (derived compounds of milbemycin), the In vitro antifungal drug susceptibilities of dermatophytes
microconidia and arthroconidia. J Antimicrob Chemother
transcription factor PacC, a wide‑domain regulatory 2008;62:758‑61.
protein involved in pathogenicity events or the sulfite 10. Tainwala R, Sharma Y. Pathogenesis of dermatophytoses.
transporters were proposed as interesting targets for Indian J Dermatol 2011;56:259‑61.
antifungal drugs in dermatology because of their role 11. Léchenne B, Reichard U, Zaugg C, Fratti M, Kunert J,
in the proteolytic digestion of hard keratin).[5,20,60‑62] Boulat O, et al. Sulphite efflux pumps in Aspergillus fumigatus
and dermatophytes. Microbiology 2007;153:905‑13.
Conclusion 12. Sanglard D, Odds FC. Resistance of Candida species to
antifungal agents: Molecular mechanisms and clinical
While deeper mycoses are still difficult to treat, common consequences. Lancet Infect Dis 2002;2:73‑85.
fungal infections in dermatology were considered easily 13. White TC, Marr KA, Bowden RA. Clinical, cellular, and
treatable. Lately, fungi causing superficial mycoses also molecular factors that contribute to antifungal drug resistance.
have evolved to develop resistance against commonly Clin Microbiol Rev 1998;11:382‑402.
used drugs. Current solutions to this could be good skin 14. Vandeputte P, Ferrari S, Coste AT. Antifungal resistance and

366 Indian Journal of Dermatology | Volume 63 | Issue 5 | September-October 2018


[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.97]

Pai, et al.: Antifungal resistance

new strategies to control fungal infections. Int J Microbiol 32. Indira  G. In vitro antifungal susceptibility testing of
2012;2012:713687. 5 antifungal agents against dermatophytic species by
15. Michaelides P, Rosenthal SA, Sulzberger MB, Witten VH. CLSI (M38‑A) micro dilution method. Clin Microbiol
Trichophyton tonsurans infection resistant to griseofulvin. 2014;3:145.
A case demonstrating clinical and in vitro resistance. Arch 33. Favre B, Hofbauer B, Hildering KS, Ryder NS. Comparison of
Dermatol 1961;83:988‑90. in vitro activities of 17 antifungal drugs against a panel of 20
16. Ryley JF, Wilson RG, Barrett‑Bee KJ. Azole resistance in dermatophytes by using a microdilution assay. J Clin Microbiol
Candida albicans. Sabouraudia 1984;22:53‑63. 2003;41:4817‑9.
17. Mukherjee PK, Leidich SD, Isham N, Leitner I, Ryder NS, 34. Jessup CJ, Warner J, Isham N, Hasan I, Ghannoum MA.
Ghannoum MA, et al. Clinical Trichophyton rubrum strain Antifungal susceptibility testing of dermatophytes:
exhibiting primary resistance to terbinafine. Antimicrob Establishing a medium for inducing conidial growth and
Agents Chemother 2003;47:82‑6. evaluation of susceptibility of clinical isolates. J Clin Microbiol
18. Law D, Moore CB, Wardle HM, Ganguli LA, Keaney MG, 2000;38:341‑4.
Denning DW. High prevalence of antifungal resistance in 35. Adimi P, Hashemi SJ, Mahmoudi M, Mirhendi H, Shidfar MR,
Candida spp. from patients with AIDS. J Antimicrob Chemother Emmami M, et al. In vitro activity of 10 antifungal agents
1994;34:659‑68. against 320 dermatophyte strains using microdilution method
19. Ghannoum M. Azole resistance in dermatophytes: Prevalence in Tehran. Iran J Pharm Res 2013;12:537‑45.
and mechanism of action. J Am Podiatr Med Assoc 36. Koga H, Nanjoh Y, Makimura K, Tsuboi R. In vitro antifungal
2016;106:79‑86. activities of luliconazole, a new topical imidazole. Med Mycol
20. Kanafani ZA, Perfect JR. Antimicrobial resistance: Resistance 2009;47:640‑7.
to antifungal agents: Mechanisms and clinical impact. Clin 37. Rex JH, Pfaller MA. Has antifungal susceptibility testing come
Infect Dis 2008;46:120‑8. of age? Clin Infect Dis 2002;35:982‑9.
21. Perlin DS, Shor E, Zhao Y. Update on antifungal drug 38. Rezabek GH, Friedman AD. Superficial fungal infections of
resistance. Curr Clin Microbiol Rep 2015;2:84‑95. the skin. Diagnosis and current treatment recommendations.
22. Polak A, Scholer HJ. Mode of action of 5‑fluorocytosine and Drugs 1992;43:674‑82.
mechanisms of resistance. Chemotherapy 1975;21:113‑30. 39. Clinard VB, Smith JD. Cutaneous fungal infections. US Pharm
23. Costa‑Orlandi CB, Sardi JC, Santos CT, Fusco‑Almeida AM, 2015;40:35‑9.
Mendes‑Giannini MJ. In vitro characterization of Trichophyton 40. Seebacher C, Bouchara JP, Mignon B. Updates on the
rubrum and T. Mentagrophytes biofilms. Biofouling epidemiology of dermatophyte infections. Mycopathologia
2014;30:719‑27. 2008;166:335‑52.
24. van der Linden JW, Camps SM, Kampinga GA, Arends JP, 41. Kim D, Lockey R. Dermatology for the allergist. World Allergy
Debets‑Ossenkopp YJ, Haas PJ, et al. Aspergillosis due to Organ J 2010;3:202‑15.
voriconazole highly resistant Aspergillus fumigatus and 42. Rotta I, Otuki MF, Sanches AC, Correr CJ. Efficacy of topical
recovery of genetically related resistant isolates from antifungal drugs in different dermatomycoses: A systematic
domiciles. Clin Infect Dis 2013;57:513‑20. review with meta‑analysis. Rev Assoc Med Bras (1992)
25. Antifungal Susceptibility Testing: Current role from the clinical 2012;58:308‑18.
laboratory perspective Brunella Posteraro, Riccardo Torelli, 43. Piérard GE, Piérard‑Franchimont C, Arrese JE. The boosted
Elena De Carolis, Patrizia Posteraro and Maurizio Sanguinetti. oral antifungal treatment for onychomycosis beyond the
Mediterr J Hematol Infect Dis 2014;6:e2014030. regular itraconazole pulse dosing regimen. Dermatology
26. CLSI. Reference Method for Broth Dilution Antifungal 2000;200:185‑7.
Susceptibility Testing of Yeasts. Approved Standard‑CLSI 44. Singal A, Khanna D. Onychomycosis: Diagnosis and
Document M27‑A3. 3rd ed. Wayne, PA: Clinical and Laboratory management. Indian J Dermatol Venereol Leprol
Standards Institute; 2008. 2011;77:659‑72.
27. CLSI. Reference Method for Broth Dilution Antifungal 45. Kontoyiannis DP, Lewis RE. Toward more effective antifungal
Susceptibility Testing of Filamentous Fungi. Approved therapy: The prospects of combination therapy. Br J Haematol
Standard CLSI Document M38‑A2. Wayne, PA: Clinical and 2004;126:165‑75.
Laboratory Standards Institute; 2008. 46. Carrillo‑Muñoz AJ, Finquelievich J, Tur‑Tur C, Eraso E,
28. CLSI. Method for Antifungal Disk Diffusion Susceptibility Jauregizar N, Quindós G, et al. Combination antifungal
Testing of Filamentous fungi. Approved Guideline. CLSI therapy: A strategy for the management of invasive fungal
Document M51‑P. Wayne, PA: Clinical and Laboratory Standards infections. Rev Esp Quimioter 2014;27:141‑58.
Institute; 2010. 47. Gupta AK, Lynde CW, Konnikov N. Single‑blind, randomized,
29. NCCLS. Reference Method for Antifungal Disk Diffusion prospective study of sequential itraconazole and terbinafine
Susceptibility Testing of Yeasts. Approved Guideline. NCCLS pulse compared with terbinafine pulse for the treatment of
Document M44‑A. Wayne, PA: National Committee for Clinical toenail onychomycosis. J Am Acad Dermatol 2001;44:485‑91.
Laboratory Standards; 2004. 48. Groll AH, Piscitelli SC, Walsh TJ. Clinical pharmacology
30. Fothergill AW. Antifungal susceptibility testing: Clinical of systemic antifungal agents: A comprehensive review of
laboratory and standards institute (CLSI) methods. In: Hall GS, agents in clinical use, current investigational compounds,
editor. Interactions of Yeasts, Moulds, and Antifungal Agents: and putative targets for antifungal drug development. Adv
How to Detect Resistance. New York: Springer; 2012. p. 65‑74. Pharmacol 1998;44:343‑500.
31. CLSI/NCCLS. Reference Method for Broth Dilution Antifungal 49. Cuenca‑Estrella M. Combinations of antifungal agents in
Susceptibility Testing of Conidial Forming Filamentous Fungi. therapy – What value are they? J Antimicrob Chemother
Approved Standard NCCLS M38‑A. Wayne, PA: National 2004;54:854‑69.
Committee for Clinical Laboratory Standards; 2002. 50. Marchetti O, Moreillon P, Glauser MP, Bille J, Sanglard D.

Indian Journal of Dermatology | Volume 63 | Issue 5 | September-October 2018 367


[Downloaded free from http://www.e-ijd.org on Sunday, June 16, 2019, IP: 106.205.76.97]

Pai, et al.: Antifungal resistance

Potent synergism of the combination of fluconazole and and as prophylaxis against Candida infections. Drugs
cyclosporine in Candida albicans. Antimicrob Agents 2008;68:2225‑55.
Chemother 2000;44:2373‑81. 57. Miceli MH, Kauffman CA. Isavuconazole: A New broad‑spectrum
51. Onyewu C, Eads E, Schell WA, Perfect JR, Ullmann Y, triazole antifungal agent. Clin Infect Dis 2015;61:1558‑65.
Kaufman G, et al. Targeting the calcineurin pathway enhances 58. Cruz MC, Goldstein AL, Blankenship J, Del Poeta M, Perfect JR,
ergosterol biosynthesis inhibitors against Trichophyton McCusker JH, et al. Rapamycin and less immunosuppressive
mentagrophytes in vitro and in a human skin infection model.
analogs are toxic to Candida albicans and Cryptococcus
Antimicrob Agents Chemother 2007;51:3743‑6.
neoformans via FKBP12‑dependent inhibition of TOR.
52. Nyilasi I, Kocsubé S, Krizsán K, Galgóczy L, Papp T, Pesti M, Antimicrob Agents Chemother 2001;45:3162‑70.
et al. Susceptibility of clinically important dermatophytes
against statins and different statin‑antifungal combinations. 59. Butts A, Krysan DJ. Antifungal drug discovery: Something old
Med Mycol 2014;52:140‑8. and something new. PLoS Pathog 2012;8:e1002870.
53. Farmaki E, Roilides E. Immunotherapy in patients with systemic 60. Sharma M, Manoharlal R, Shukla S, Puri N, Prasad T,
mycoses: A promising adjunct. BioDrugs 2001;15:207‑14. Ambudkar SV, et al. Curcumin modulates efflux mediated
54. Casadevall A, Pirofski LA. Adjunctive immune therapy for by yeast ABC multidrug transporters and is synergistic with
fungal infections. Clin Infect Dis 2001;33:1048‑56. antifungals. Antimicrob Agents Chemother 2009;53:3256‑65.
55. Bhatta AK, Keyal U, Huang X, Zhao JJ. Fractional 61. Ferreira‑Nozawa MS, Silveira HC, Ono CJ, Fachin AL, Rossi A,
carbon‑dioxide (CO2) laser‑assisted topical therapy for Martinez‑Rossi NM. The pH signaling transcription factor PacC
the treatment of onychomycosis. J Am Acad Dermatol mediates the growth of Trichophyton rubrum on human nail
2016;74:916‑23. in vitro. Med Mycol 2006;44:641‑5.
56. Cross SA, Scott LJ. Micafungin: A review of its use in adults 62. Odds FC. Genomics, molecular targets and the discovery of
for the treatment of invasive and oesophageal candidiasis, antifungal drugs. Rev Iberoam Micol 2005;22:229‑37.

368 Indian Journal of Dermatology | Volume 63 | Issue 5 | September-October 2018

You might also like