Review

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

Future directions for anti-biofilm therapeutics targeting Candida Expert Rev. Anti Infect. Ther. 12(3), 375–382 (2014)

Jeniel E Nett Department of Medicine, Department of Medical Microbiology and Immunology, University of Wisconsin, 4153 Microbial Sciences Building, 1550 Linden Drive, Madison, WI 53705, USA Tel.: +1 608 262 7494 Fax: +1 608 263 4464 [email protected]

While proliferating in its most common mode of growth, a biofilm, Candida spp. exhibit increased resistance to available antifungal agents. These adherent communities are difficult to eradicate and often responsible for treatment failures. New therapies are urgently needed to treat a variety of Candida biofilm infections in the medical setting. This review discusses the medical relevance of Candida biofilms, the drug resistance associated with this mode of growth, and approaches to combat these resilient infections. KEYWORDS: amphotericin B • antifungal • azole • biofilm • Candida • echinocandin • lock therapy • matrix • resistance

Role of biofilm in Candida infection

Many fungal and bacterial pathogens thrive as biofilms, causing infections in a variety of clinical and environmental niches [1,2]. As a biofilm, a microbial population demonstrates properties distinct from non-biofilm, free-floating cells. These cohesive communities of organisms are adherent to a surface and surrounded by an extracellular, polymeric matrix. They resist host defenses and may be extraordinarily tolerant to antimicrobials, withstanding anti-infectives at concentrations many times greater than those needed to eradicate non-biofilm cells [1,3]. The importance of biofilms has been increasingly evident with the widespread use of medical devices, as nearly all device-associated infections involve the biofilm mode of growth [2,4]. Vulnerable devices are diverse and include catheters, implants, pacemakers, artificial heart valves and CNS shunts [2,5]. Infection by Candida, the most common hospital-acquired fungal pathogen, frequently involves biofilm growth [5,6]. This organism has a propensity to adhere to the surface of catheters and other commonly used medical devices (FIGURE 1). In the hospital setting where medical device use is commonplace, Candida is the fourth most common cause of bloodstream infection and the third most common cause of urinary tract infection [7–10]. Exhibiting increased resistance to available antifungal

informahealthcare.com

10.1586/14787210.2014.885838

therapies, these infections are notoriously difficult to treat. Most often, cure of biofilm infections requires removal of the infected device [11]. This is reflected in current guidelines that recommend removal of Candidainfected medical devices as treatment of the infection [12]. However, procedural risk to the patient may be significant, especially in the setting of permanent devices, such as pacemakers or hemodialysis access devices. Efficacy of available antifungal therapies

The majority of medically important Candida spp. have now been shown to generate biofilms, including C. albicans, C. dubliniensis, C. glabrata, C. krusei, C. tropicalis and C. parapsilosis [6,13]. Candida biofilms exhibit resistance to agents from all available, commonly used antifungal drug classes, including the azoles (fluconazole, itraconazole, voriconazole, posaconazole), the echinocandins (caspofungin, micafungin, anidulafungin), the amphotericin B formulations and flucytosine [14–19]. These antifungal drugs have diverse mechanisms of action. The azole drugs and amphotericin B formulations interfere with cell membrane ergosterol, while the echinocandins inhibit cell wall b-1,3 glucan synthesis and flucytosine, a pyrimidine analog, inhibits nucleic acid synthesis. [20]. The biofilmassociated drug-resistant phenotype has been replicated and studied using numerous in vitro models of biofilm infections [11,12,21–24]. In vivo

 2014 Informa UK Ltd

ISSN 1478-7210

375

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

Review

Nett

10 µm

Figure 1. Candida albicans biofilm infection of rat venous catheter. C. albicans was instilled in the lumen of a jugular venous catheter, allowed to dwell for 6 h, and then flushed. After a growth period of 24 h, the catheter was harvested, fixed and dehydrated. Catheter segments were imaged by scanning electron microscopy on a JEOL JSM-6100 at 10 kV (2000). The biofilm is composed of both yeast and hyphae encased in an extracellular matrix.

studies using rat or rabbit models of vascular catheter infection show Candida biofilms can withstand fluconazole concentrations up to 1000-fold higher than the minimal inhibitory concentration (MIC) for planktonic organisms [25,26]. Clinically, outcomes of patients with invasive candidiasis are improved when infected medical devices are removed, signifying that antifungal therapy alone is not adequate for treatment of Candida biofilm infection. Although resistance has been described for commonly used antifungals, the degree of drug resistance associated with biofilm growth appears to vary quite significantly among the drugs. Studies examining the impact of fluconazole, the most commonly used azole drug, demonstrate that biofilms are tolerant to concentrations up to 2000-fold above the MIC of planktonic cells [18,27]. Comparatively, the antifungals with the most activity against biofilms are the liposomal formulation of amphotericin B and agents in the echinocandin drug class, which have been shown to be active at concentrations 2- to 25-fold above their planktonic MICs [28]. As might be expected by this observation, the mechanism of biofilm drug resistance is a complex phenomenon that varies by drug class. These investigations have included diverse in vivo and in vitro experimental systems, which differ with regard to substrate, flow, media and environmental conditions. The degree of biofilm resistance has been shown to vary among the in vitro and in vivo models employed [29]. Studies have also found considerable variation in drug resistance among biofilms formed by different Candida species and strains [30].

collectively promote resistance to anti-infectives during biofilm growth. There appears to be interplay between several resistance mechanisms, which vary during the phases of biofilm growth. Also, there is diversity among the biofilm cells in these heterogeneous structures. For example, subpopulations of Candida within biofilms are highly resistant to amphotericin B [33,34]. Upon drug treatment, these ‘persisters’ are proposed to act as a reservoir, allowing the biofilm to repopulate. An example of a phasespecific mechanism of resistance is the alteration of efflux pump activity. Upon transitioning to the biofilm lifestyle, the activity of C. albicans efflux pumps increases, ultimately lowering the intracellular azole drug concentrations and promoting resistance [18,31]. The contribution of the mechanism appears to be greatest early in biofilm formation, likely due to the lower cell density [37]. Other mechanisms are more specific to mature biofilm formation. For example, later in biofilm development, the increased cell density and extracellular matrix promotes resistance to amphotericin B, azoles and echinocandins [3,35–40]. In addition, the altered ergosterol content found in mature biofilm cells is hypothesized to influence drug resistance during the later growth phase [18]. The presence of an adhesive extracellular matrix, one of the unique and defining biofilm traits, has been linked to the multidrug-resistant phenotype observed for biofilms formed by many Candida spp., including C. albicans, C. parapsilosis, C. tropicalis, C. glabrata and C. krusei [3,35,36,38–40]. This mechanism has been shown to play a key role in biofilm resistance to amphotericin B, echinocandin drugs, flucytosine and fluconazole [40–43]. Several components of the extracellular matrix have been linked to biofilm resistance. The b-1,3 glucan content of the biofilm matrix correlates the antifungal-resistant phenotype and this substance has been shown to be a key factor in the binding or sequestering of both fluconazole and amphotericin B [43,44]. More recently, the importance of C. albicans eDNA has been recognized. Interestingly, this substance is critical for biofilm resistance to echinocandins and amphotericin B, but does not appear to contribute greatly to azole resistance [42]. While the importance of the biofilm matrix in biofilm drug resistance has become increasing appreciated, how the individual components cooperate to produce this characteristic remains unclear, and may vary for the individual drugs. As Candida transitions to the biofilm lifestyle, cellular stress responses become activated. These pathways contribute to the ability of Candida biofilms to resistant environmental insults, including antifungal therapy. Pathways shown to contribute to azole resistance in Candida biofilms involve the mitogen-activated protein kinase, calcineurin and heat shock protein 90 (Hsp90p) [32,45–47]. These resistance pathways have been of great interest, as there are pharmaceutical agents that target calcineurin and Hsp90, which will be discussed below [45,47,48].

Mechanisms of Candida biofilm resistance

Identification of the mechanisms underlying the resistance of Candida biofilms to antifungals has been of great interest [18,31–36]. Studies to date support a model in which multiple factors 376

Strategies to combat Candida biofilms infections

As the available antifungal agents have minimal activity against biofilm infections, innovative approaches have been undertaken Expert Rev. Anti Infect. Ther. 12(3), (2014)

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

Future directions for anti-biofilm therapeutics targeting Candida

Review

Local/lock therapy to eradicate these infections (FIGURE 2). One Enhance antifungal activity – Antifungals of the strategies to optimize the efficacy – Inhibition of stress responses – Antiseptics of antifungals is delivery of high drug – Prostaglandin inhibition concentrations, most often by direct – Matrix degradation administration, such as lock therapy for – Quorum sensing disruption Novel compounds an infected catheter [26,49–51]. Other inves– Natural products tigations have examined the impact of – Pharmaceutical targeting a variety of pathways involved in biofilm growth, including the stress responses, quorum sensing, extracellular matrix production and prostaglandin synthesis [45,47,48,52–54]. Disruption of these processes can greatly enhance the activity Figure 2. Anti-biofilm strategies. of antifungals during combination therapy. Lastly, screens of pharmaceutical and natural products have yielded promising compounds with excel- hydrazide, this compound forms a fungicidal gel. The product was well-tolerated in animals and has potential for future diverse lent anti-biofilm activity [55–57]. uses in the local treatment of Candida biofilm infection. Possible applications include the treatment of vascular catheter, bone, joint High-dose therapy with available antifungals While biofilms are capable of withstanding antifungals at the and abdominal infections involving Candida biofilms. No controlled trials have examined the efficacy of antifungal concentrations safely achievable in patients, they are susceptible to higher concentrations of a subset of these drugs, including lock therapy for patients with fungal biofilm infections and amphotericin B and the echinocandins [28]. The toxicity of data are limited to few case reports [51]. Lock therapy regimens amphotericin B formulations limits their use at higher doses. used in patients primarily have included high doses of amphoAlthough azoles are relatively well-tolerated, Candida biofilms tericin B formulations or ethanol. Although several case reports are tolerant to exceedingly high concentrations of these describe high salvage rates, there is a concern for the possibility drugs [40–43]. Of the available antifungals, the echinocandins of publication bias. A randomized controlled trial is needed to appear to have the most utility for biofilm treatment when determine if the lock therapies are effective, and if so, for administered systemically. Clinically, higher doses of these which patient population. As lock therapy is delivered to the drugs have been used for treatment of biofilm infection, such luminal surface only, biofilm infections involving the catheter as endocarditis [12]. However, even with the higher doses, bio- tip or outer surface would likely have a higher failure rate. films often persist and physical removal of biofilm is ultimately Also, if there is a concern for disseminated infection, concomitant systemic antifungal therapy may be required. In addition, required for cure [11,12]. there is the possibility that the use of an antifungal lock may promote development of resistance. Therefore, the use of antiAnti-infective lock therapy Lock therapy, the prolonged instillation of drug into the cathe- septics or non-antifungal agents is of interest. In vitro, EDTA, ter lumen, allows delivery of high-dose therapy while avoiding ethanol and high-dose minocycline (3 mg/ml) appear to be systemic toxicity. By directly administering drug to the infected promising lock agents for treatment of Candida biofilms [61–63]. site, local antifungal concentrations may safely reach 1000-fold higher than those achieved with systemic therapy [51]. Based on Exploiting combination therapy in vitro models, the antifungals anticipated to have the greatest One approach to increase the efficacy of antifungal therapies is anti-biofilm activity when administered in lock therapy are to combine synergistic agents. This strategy has been shown to drugs in the echinocandin class and two amphotericin B for- be successful for several drug classes for the treatment of mulations, liposomal amphotericin B and amphotericin B lipid Candida biofilms. Some of the most effective studies have complex [28,58]. In animal vascular catheter infection models, included agents targeting stress responses activated during biolock therapies with high doses of these agents (liposomal film growth [45,47]. Uppuluri et al. identified calcineurin as a key amphotericin B 10 mg/ml, liposomal amphotericin B and regulator of C. albicans biofilm drug resistance [47]. The addition amphotericin B lipid complex 5 mg/ml and caspofungin of inhibitors of calcineurin (cyclosporine A or tacrolimus) ren6.67 mg/ml) successfully eradicated C. albicans biofilms [26,49–51]. dered in vitro biofilms susceptible to fluconazole. This synergy However, consistent with in vitro data, azole therapy (flucona- was recapitulated in vivo using combination lock therapy in a zole 10 mg/ml) and low-dose echinocandin (0.25 mg/ml) were rat catheter biofilm infection model. Calcineurin inhibitors have much less effective as lock therapy for treatment of the animal since been shown to exhibit synergy with both an echinocandin catheter infections [26,59]. Hudson et al. described an innovative (caspofungin) and amphotericin B [48]. The clinical availability amphotericin B formulation, a dextran-aldehyde-amphotericin of calcineurin inhibitors makes this combination therapy appealB conjugate [60]. When mixed with carboxymethlcellulose- ing. However, the immunosuppressive effects of these drugs

informahealthcare.com

377

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

Review

Nett

would likely preclude systemic administration and may limit their utility to lock therapy. Combination therapy also looks promising for agents targeting Hsp90p, a molecular chaperone governing Candida biofilm resistance [45]. Combination therapy with an Hsp90 inhibitor, geldanamycin, potentiated the activity of fluconazole against C. albicans biofilms. The use of NSAIDs in combination with antifungal therapy shows potential for treatment of biofilm infections [52]. Medications in this drug class inhibit cyclooxygenases involved in the biosynthesis of mammalian prostaglandins. They are currently used for treatment of a variety of pain and inflammatory conditions. Interest for combination therapy arose from the discovery that several NSAIDs impair the ability of C. albicans to produce filaments and form biofilms in vitro [64,65]. One of the most potent inhibitors, aspirin, was even effective in eliminating established biofilms. This activity has been linked to inhibition of fungal prostaglandin E2 production [64]. Bink et al. explored the utility of NSAID-based combination therapy by testing the impact of combining an NSAID (diclofenac) with an echinocandin (caspofungin) [52]. In vitro, diclofenac acted synergistically with caspofungin and was most efficacious if administered during biofilm formation. In a subcutaneous device-associated biofilm model, treatment with diclofenac markedly enhanced the activity of caspofungin. This finding is exciting as NSAIDs are pharmaceutically available agents with established safety profiles. However, further studies are needed to determine the feasibility of this type of treatment regimen for patients. It is unclear if initiating the combination therapy in the setting of an established biofilm infection, as would be done clinically, would show this degree of efficacy. To identify novel agents effective against C. albicans biofilms, LaFleur et al. screened compounds for synergistic activity of azoles [55]. They discovered that 2-adamantanamine, the structural analog of the antiviral amantadine, potentiated the activity of the azole, miconazole. The specific fungal target of 2-adamantanamine is unknown, but it is thought to inhibit a component of the ergosterol pathway upstream of the azole target, a-14 lanosterol demethylase. The safety profile of 2-adamantanamine has not been established. If the profile is similar to the antiviral amantadine, therapeutic systemic levels may be achievable [55]. Disrupting quorum sensing

As Candida cells proliferate, they communicate through quorum sensing, a process of sensing cell density [66]. One of these secreted molecules, farnesol, is an isoprenoid that inhibits the yeast-to-hyphae transition as well as the initiation of biofilm. At physiologic concentrations, this quorum sensing molecule does not appear to impact cells that have already begun to filament or biofilms that have become established [67]. However, at farnesol concentrations exceeding physiologic conditions, biofilms are degraded [68]. This observation suggests a potential use of farnesol, or a compound targeting this pathway, in the treatment of biofilm infections. In vitro data suggest that farnesol treatment may even enhance the activity of azoles [53,54]. 378

In a non-biofilm murine candidiasis model, administration of farnesol was protective [69]. However, studies have not examined farnesol for treatment of biofilm infections in animal models. There is a concern that disruption of quorum sensing may provoke dispersion, the process of releasing cells from the biofilm, ultimately leading to disseminated disease [68]. Further studies of biofilms would be valuable to access the utility of targeting this quorum sensing pathway for Candida biofilm treatment. Targeting extracellular matrix

One unique characteristic of biofilms, the presence of an extracellular matrix, contributes greatly to the antifungal resistance. The matrix elements specifically linked to this resistance, eDNA and b-1,3 glucan, are potential targets for anti-biofilm therapies [40–43]. When these components are disrupted by targeted enzymatic digestion, the efficacy of antifungals is greatly enhanced. For example, digestion of b-1,3 glucan potentiated fluconazole activity while degradation of eDNA enhanced amphotericin B activity in vitro [25,35,42]. The utility of targeted enzyme degradation would likely be limited to local therapy. An example that supports the feasibility of this approach is dornase alfa (Pulmozyme), a clinically available inhaled enzymatic treatment that targets the eDNA of bacterial biofilms in patients with cystic fibrosis and chronic pulmonary infections [70]. Another potential approach would be to identify inhibitors of the pathways known to regulate biofilm matrix production and delivery [44,71,72]. Natural products

Many natural products are active against biofilms formed by a variety of Candida spp. [73]. Compounds in investigation include peptides, oils, plant extracts and polyphenols from teas [56,57,74–80]. Although the results of these studies appear promising, the toxicities of many of the compounds have not been established and the tolerable dose in patients is unknown. One of the more well-studied compounds is carbohydratederived fulvic acid (CHD-FA), a heat stable, cationic, colloidal material [56]. CHD-FA is active against Candida biofilms as well as bacterial biofilms formed by oral bacteria that often accompany Candida in polymicrobial oral biofilm infections [56,81]. CHD-FA appears to non-specifically disrupt the fungal cell membrane and retains activity against many drugresistant Candida strains. As it is relatively inexpensive and exhibits broad-spectrum anti-biofilm activity, it may be well suited for use as an oral antiseptic. The efficacy of CHD-FA has not been established in vivo, but the compound appears to be non-toxic in an epithelial cell line and a rat model [81]. To identify natural products with antifungal activity, Coleman et al. screened compounds using a high-throughput Caenorhabditis elegans assay [57]. This innovative approach concurrently assesses toxicity and treatment efficacy. With this screen, they identified 12 saponins with antifungal activity, several of which were active against C. albicans biofilms at concentrations without apparent toxicity. These compounds are proposed to target fungal ergosterol and form pores in the Expert Rev. Anti Infect. Ther. 12(3), (2014)

Future directions for anti-biofilm therapeutics targeting Candida

Candida membrane. With a mechanism of action distinct from available drug classes, these compounds have the potential to have activity against resistant organisms or act synergistically with other antifungals.

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

Expert commentary & five-year view

It is becoming increasingly clear that Candida cells of a biofilm vary greatly from their free-floating counterparts. Therapies that inhibit planktonic Candida often have little impact on biofilms. Given the high prevalence of biofilm infection, the development of anti-Candida therapies should focus on this mode of growth. Models of Candida biofilms are critical for discovery of new antifungals and for investigating their efficacy. The development of high-throughput models is necessary for economically testing large libraries of molecules for anti-biofilm activity. One such model is the biofilm chip, which consists of nano-biofilms on a high-density microarray platform [82]. A number of animal models with Candida biofilm infections closely mimicking patient infections will be of great value for examination of treatment efficacy [22,26,83–86]. To be the most useful medically, new antifungals would ideally be capable of completely eradicating a biofilm, as just a

Review

small nidus of infection may blossom to severe, recurrent infection. To achieve this degree of activity, several strategies may be considered for design of new drugs. One approach is to identify compounds with exquisite anti-Candida and antibiofilm activity, effective against even the most resistant subpopulation of biofilm cells. Another approach is to uncover agents that disrupt biofilm processes, allowing traditional antifungals to attack biofilms when used on combination. Still another approach is to identify drugs that disrupt biofilm in a manner that allows the immune system to take hold, ultimately completely clearing the infection. As our understanding of Candida biofilms continues to grow, it will be fascinating to see how this information is applied to discovery of anti-biofilm drugs. Financial & competing interests disclosure

The author’s research is supported by a Burroughs Wellcome Fund Career Award for Medical Scientists. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

Key issues • Candida, one of the most common fungal pathogens, frequently grows as a biofilm adherent to a medical device or other surface. • Candida biofilms exhibit increased resistance to antimicrobial therapies, including all available antifungal agents. • The drug resistance of Candida biofilms is multifactorial. Contributing mechanisms include the presence of an extracellular matrix, increased activity of efflux pumps, increased cell density and induction of stress responses. • Animal studies show that high doses of antifungals, such as liposomal amphotericin B and echinocandins, have anti-biofilm activity when delivered locally as catheter lock therapy. • Therapies targeting the fungal stress responses (calcineurin, heat shock protein 90) can augment the action of antifungal drugs. • Natural products, such as carbohydrate-derived fulvic acid and saponins, are promising compounds for treatment of Candida biofilms. • A combination of high-throughput screens and animal models of Candida biofilm infection will be important for identifying and testing novel antifungals.

References 1.

O’Toole GA. To build a biofilm. J Bacteriol 2003;185(9):2687-9

2.

Donlan RM. Biofilms and device-associated infections. Emerg Infect Dis 2001;7(2): 277-81

3.

Hawser SP, Baillie GS, Douglas LJ. Production of extracellular matrix by Candida albicans biofilms. J Med Microbiol 1998;47(3):253-6

4.

Passerini L, Lam K, Costerton JW, King EG. Biofilms on indwelling vascular catheters. Crit Care Med 1992;20(5):665-73

5.

Kojic EM, Darouiche RO. Candida infections of medical devices. Clin Microbiol Rev 2004;17(2):255-67

informahealthcare.com

6.

Douglas LJ. Medical importance of biofilms in Candida infections. Rev Iberoam Micol 2002;19(3):139-43

7.

Groeger JS, Lucas AB, Thaler HT, et al. Infectious morbidity associated with long-term use of venous access devices in patients with cancer. Ann Intern Med 1993; 119(12):1168-74

8.

9.

Richards MJ, Edwards JR, Culver DH, Gaynes RP. Nosocomial infections in medical intensive care units in the United States. National Nosocomial Infections Surveillance System. Crit Care Med 1999; 27(5):887-92 Edmond MB, Wallace SE, McClish DK, et al. Nosocomial bloodstream infections in United States hospitals: a three-year analysis. Clin Infect Dis 1999;29(2):239-44

10.

Pfaller MA, Diekema DJ. Epidemiology of invasive candidiasis: a persistent public health problem. Clin Microbiol Rev 2007; 20(1):133-63

11.

Andes DR, Safdar N, Baddley JW, et al. Impact of treatment strategy on outcomes in patients with candidemia and other forms of invasive candidiasis: a patient-level quantitative review of randomized trials. Clin Infect Dis 2012;54(8):1110-22

12.

Pappas PG, Kauffman CA, Andes D, et al. Clinical practice guidelines for the management of candidiasis: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis 2009;48(5):503-35

13.

Shin JH, Kee SJ, Shin MG, et al. Biofilm production by isolates of Candida species recovered from nonneutropenic patients:

379

Review

Nett

comparison of bloodstream isolates with isolates from other sources. J Clin Microbiol 2002;40(4):1244-8 14.

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

15.

Baillie GS, Douglas LJ. Effect of growth rate on resistance of Candida albicans biofilms to antifungal agents. Antimicrob Agents Chemother 1998;42(8):1900-5 Chandra J, Kuhn DM, Mukherjee PK, et al. Biofilm formation by the fungal pathogen Candida albicans: development, architecture, and drug resistance. J Bacteriol 2001;183(18):5385-94

16.

Hawser SP, Douglas LJ. Biofilm formation by Candida species on the surface of catheter materials in vitro. Infect Immun 1994;62(3):915-21

17.

Lewis RE, Kontoyiannis DP, Darouiche RO, et al. Antifungal activity of amphotericin B, fluconazole, and voriconazole in an in vitro model of Candida catheter-related bloodstream infection. Antimicrob Agents Chemother 2002;46(11):3499-505

18.

19.

Mukherjee PK, Chandra J, Kuhn DM, Ghannoum MA. Mechanism of fluconazole resistance in Candida albicans biofilms: phase-specific role of efflux pumps and membrane sterols. Infect Immun 2003; 71(8):4333-40 Ramage G, VandeWalle K, Bachmann SP, et al. In vitro pharmacodynamic properties of three antifungal agents against preformed Candida albicans biofilms determined by time-kill studies. Antimicrob Agents Chemother 2002;46(11):3634-6

26.

27.

28.

Schinabeck MK, Long LA, Hossain MA, et al. Rabbit model of Candida albicans biofilm infection: liposomal amphotericin B antifungal lock therapy. Antimicrob Agents Chemother 2004;48(5):1727-32 Ramage G, Vande Walle K, Wickes BL, Lopez-Ribot JL. Standardized method for in vitro antifungal susceptibility testing of Candida albicans biofilms. Antimicrob Agents Chemother 2001;45(9):2475-9 Kuhn DM, George T, Chandra J, et al. Antifungal susceptibility of Candida biofilms: unique efficacy of amphotericin B lipid formulations and echinocandins. Antimicrob Agents Chemother 2002;46(6): 1773-80

29.

Ramage G, Rajendran R, Sherry L, Williams C. Fungal biofilm resistance. Int J Microbiol 2012;2012:528521

30.

Fiori B, Posteraro B, Torelli R, et al. In vitro activities of anidulafungin and other antifungal agents against biofilms formed by clinical isolates of different Candida and Aspergillus species. Antimicrob Agents Chemother 2011;55(6):3031-5

31.

32.

Ramage G, Bachmann S, Patterson TF, et al. Investigation of multidrug efflux pumps in relation to fluconazole resistance in Candida albicans biofilms. J Antimicrob Chemother 2002;49(6):973-80 Kumamoto CA. A contact-activated kinase signals Candida albicans invasive growth and biofilm development. Proc Natl Acad Sci USA 2005;102(15):5576-81

20.

Dodds Ashley ES, Lewis R, Lewis JS, et al. Pharmacology of systemic antifungal agents. Clin Infect Dis 2006;43(S1):S28-39

21.

Hawser SP, Douglas LJ. Resistance of Candida albicans biofilms to antifungal agents in vitro. Antimicrob Agents Chemother 1995;39(9):2128-31

22.

Andes D, Nett J, Oschel P, et al. Development and characterization of an in vivo central venous catheter Candida albicans biofilm model. Infect Immun 2004; 72(10):6023-31

34.

LaFleur MD, Kumamoto CA, Lewis K. Candida albicans biofilms produce antifungal-tolerant persister cells. Antimicrob Agents Chemother 2006;50(11): 3839-46

23.

Ramage G, Vandewalle K, Wickes BL, Lopez-Ribot JL. Characteristics of biofilm formation by Candida albicans. Rev Iberoam Micol 2001;18(4):163-70

35.

24.

Chandra J, Mukherjee PK, Leidich SD, et al. Antifungal resistance of candidal biofilms formed on denture acrylic in vitro. J Dent Res 2001;80(3):903-8

Al-Fattani MA, Douglas LJ. Biofilm matrix of Candida albicans and Candida tropicalis: chemical composition and role in drug resistance. J Med Microbiol 2006;55(Pt 8): 999-1008

36.

Mitchell KF, Taff HT, Cuevas MA, et al. Role of matrix beta-1,3 glucan in antifungal resistance of non-albicans Candida Biofilms. Antimicrob Agents Chemother 2013;57(4): 1918-20

37.

Perumal P, Mekala S, Chaffin WL. Role for cell density in antifungal drug resistance in

25.

Nett J, Lincoln L, Marchillo K, et al. Putative role of beta-1,3 glucans in Candida albicans biofilm resistance. Antimicrob Agents Chemother 2007;51(2):510-20

380

33.

Khot PD, Suci PA, Miller RL, et al. A small subpopulation of blastospores in Candida albicans biofilms exhibit resistance to amphotericin B associated with differential regulation of ergosterol and beta-1,6-glucan pathway genes. Antimicrob Agents Chemother 2006;50(11):3708-16

Candida albicans biofilms. Antimicrob Agents Chemother 2007;51(7):2454-63 38.

Al-Fattani MA, Douglas LJ. Penetration of Candida biofilms by antifungal agents. Antimicrob Agents Chemother 2004;48(9): 3291-7

39.

Baillie GS, Douglas LJ. Matrix polymers of Candida biofilms and their possible role in biofilm resistance to antifungal agents. J Antimicrob Chemother 2000;46(3): 397-403

40.

Samaranayake YH, Ye J, Yau JY, et al. In vitro method to study antifungal perfusion in Candida biofilms. J Clin Microbiol 2005;43(2):818-25

41.

Nett JE, Crawford K, Marchillo K, Andes DR. Role of Fks1p and matrix glucan in Candida albicans biofilm resistance to an echinocandin, pyrimidine, and polyene. Antimicrob Agents Chemother 2010;54(8):3505-8

42.

Martins M, Henriques M, Lopez-Ribot JL, Oliveira R. Addition of DNase improves the in vitro activity of antifungal drugs against Candida albicans biofilms. Mycoses 2012;55(1):80-5

43.

Vediyappan G, Rossignol T, d’Enfert C. Interaction of Candida albicans biofilms with antifungals: transcriptional response and binding of antifungals to beta-glucans. Antimicrob Agents Chemother 2010;54(5): 2096-111

44.

Nett JE, Sanchez H, Cain MT, Andes DR. Genetic basis of Candida biofilm resistance due to drug-sequestering matrix glucan. J Infect Dis 2010;202(1):171-5

45.

Robbins N, Uppuluri P, Nett J, et al. Hsp90 governs dispersion and drug resistance of fungal biofilms. PLoS Pathog 2011;7(9):e1002257

46.

Chen YL, Brand A, Morrison EL, et al. Calcineurin controls drug tolerance, hyphal growth, and virulence in Candida dubliniensis. Eukaryot Cell 2011;10(6): 803-19

47.

Uppuluri P, Nett J, Heitman J, Andes D. Synergistic effect of calcineurin inhibitors and fluconazole against Candida albicans biofilms. Antimicrob Agents Chemother 2008;52(3):1127-32

48.

Shinde RB, Chauhan NM, Raut JS, Karuppayil SM. Sensitization of Candida albicans biofilms to various antifungal drugs by cyclosporine A. Ann Clin Microbiol Antimicrob 2012;11:27

49.

Mukherjee PK, Long L, Kim HG, Ghannoum MA. Amphotericin B lipid complex is efficacious in the treatment of Candida albicans biofilms using a model of

Expert Rev. Anti Infect. Ther. 12(3), (2014)

Future directions for anti-biofilm therapeutics targeting Candida

catheter-associated Candida biofilms. Int J Antimicrob Agents 2009;33(2):149-53

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

50.

Shuford JA, Rouse MS, Piper KE, et al. Evaluation of caspofungin and amphotericin B deoxycholate against Candida albicans biofilms in an experimental intravascular catheter infection model. J Infect Dis 2006; 194(5):710-13

51.

Walraven CJ, Lee SA. Antifungal lock therapy. Antimicrob Agents Chemother 2013;57(1):1-8

52.

Bink A, Kucharikova S, Neirinck B, et al. The nonsteroidal antiinflammatory drug diclofenac potentiates the in vivo activity of caspofungin against Candida albicans biofilms. J Infect Dis 2012;206(11):1790-7

53.

54.

55.

56.

57.

58.

59.

60.

61.

Yu LH, Wei X, Ma M, et al. Possible inhibitory molecular mechanism of farnesol on the development of fluconazole resistance in Candida albicans biofilm. Antimicrob Agents Chemother 2012;56(2):770-5 Sharma M, Prasad R. The quorum-sensing molecule farnesol is a modulator of drug efflux mediated by ABC multidrug transporters and synergizes with drugs in Candida albicans. Antimicrob Agents Chemother 2011;55(10):4834-43 Lafleur MD, Sun L, Lister I, et al. Potentiation of azole antifungals by 2-adamantanamine. Antimicrob Agents Chemother 2013;57(8):3585-92 Sherry L, Jose A, Murray C, et al. Carbohydrate Derived Fulvic Acid: an in vitro Investigation of a Novel Membrane Active Antiseptic Agent Against Candida albicans Biofilms. Front Microbiol 2012;3:116 Coleman JJ, Okoli I, Tegos GP, et al. Characterization of plant-derived saponin natural products against Candida albicans. ACS Chem Biol 2010;5(3):321-32 Bachmann SP, VandeWalle K, Ramage G, et al. In vitro activity of caspofungin against Candida albicans biofilms. Antimicrob Agents Chemother 2002;46(11):3591-6 Lazzell AL, Chaturvedi AK, Pierce CG, et al. Treatment and prevention of Candida albicans biofilms with caspofungin in a novel central venous catheter murine model of candidiasis. J Antimicrob Chemother 2009;64(3):567-70 Hudson SP, Langer R, Fink GR, Kohane DS. Injectable in situ cross-linking hydrogels for local antifungal therapy. Biomaterials 2010;31(6):1444-52 Raad I, Hanna H, Dvorak T, et al. Optimal antimicrobial catheter lock solution, using different combinations of minocycline, EDTA, and 25-percent ethanol, rapidly

informahealthcare.com

eradicates organisms embedded in biofilm. Antimicrob Agents Chemother 2007;51(1): 78-83 62.

Sherertz RJ, Boger MS, Collins CA, et al. Comparative in vitro efficacies of various catheter lock solutions. Antimicrob Agents Chemother 2006;50(5):1865-8

63.

Raad I, Chatzinikolaou I, Chaiban G, et al. In vitro and ex vivo activities of minocycline and EDTA against microorganisms embedded in biofilm on catheter surfaces. Antimicrob Agents Chemother 2003;47(11): 3580-5

64.

Alem MA, Douglas LJ. Prostaglandin production during growth of Candida albicans biofilms. J Med Microbiol 2005; 54(Pt 11):1001-5

65.

Ghalehnoo ZR, Rashki A, Najimi M, Dominguez A. The role of diclofenac sodium in the dimorphic transition in Candida albicans. Microb Pathog 2010; 48(3-4):110-15

66.

Hornby JM, Jensen EC, Lisec AD, et al. Quorum sensing in the dimorphic fungus Candida albicans is mediated by farnesol. Appl Environ Microbiol 2001;67(7): 2982-92

67.

Ramage G, Saville SP, Wickes BL, Lopez-Ribot JL. Inhibition of Candida albicans biofilm formation by farnesol, a quorum-sensing molecule. Appl Environ Microbiol 2002;68(11):5459-63

68.

69.

Uppuluri P, Chaturvedi AK, Srinivasan A, et al. Dispersion as an important step in the Candida albicans biofilm developmental cycle. PLoS Pathog 2010; 6(3):e1000828 Martins M, Lazzell AL, Lopez-Ribot JL, et al. Effect of exogenous administration of Candida albicans autoregulatory alcohols in a murine model of hematogenously disseminated candidiasis. J Basic Microbiol 2012;52(4):487-91

70.

Frederiksen B, Pressler T, Hansen A, et al. Effect of aerosolized rhDNase (Pulmozyme) on pulmonary colonization in patients with cystic fibrosis. Acta Paediatr 2006;95(9): 1070-4

71.

Taff HT, Nett JE, Zarnowski R, et al. A Candida biofilm-induced pathway for matrix glucan delivery: implications for drug resistance. PLoS Pathog 2012;8(8): e1002848

72.

Nobile CJ, Nett JE, Hernday AD, et al. Biofilm matrix regulation by Candida albicans Zap1. PLoS Biol 2009;7(6): e1000133

73.

Sardi JC, Scorzoni L, Bernardi T, et al. Candida species: current epidemiology,

Review

pathogenicity, biofilm formation, natural antifungal products and new therapeutic options. J Med Microbiol 2013;62(Pt 1): 10-24 74.

Rossignol T, Kelly B, Dobson C, d’Enfert C. Endocytosis-mediated vacuolar accumulation of the human ApoE apolipoprotein-derived ApoEdpL-W antimicrobial peptide contributes to its antifungal activity in Candida albicans. Antimicrob Agents Chemother 2011;55(10): 4670-81

75.

Mandal SM. A novel hydroxyproline rich glycopeptide from pericarp of Datura stramonium: proficiently eradicate the biofilm of antifungals resistant Candida albicans. Biopolymers 2012; 98(4):332-7

76.

Pires RH, Lucarini R, Mendes-Giannini MJ. Effect of usnic acid on Candida orthopsilosis and C. parapsilosis. Antimicrob Agents Chemother 2012;56(1):595-7

77.

Pires RH, Montanari LB, Martins CH, et al. Anticandidal efficacy of cinnamon oil against planktonic and biofilm cultures of Candida parapsilosis and Candida orthopsilosis. Mycopathologia 2011;172(6): 453-64

78.

Evensen NA, Braun PC. The effects of tea polyphenols on Candida albicans: inhibition of biofilm formation and proteasome inactivation. Can J Microbiol 2009;55(9): 1033-9

79.

Alviano WS, Mendonca-Filho RR, Alviano DS, et al. Antimicrobial activity of Croton cajucara Benth linalool-rich essential oil on artificial biofilms and planktonic microorganisms. Oral Microbiol Immunol 2005;20(2):101-5

80.

Shuford JA, Steckelberg JM, Patel R. Effects of fresh garlic extract on Candida albicans biofilms. Antimicrob Agents Chemother 2005;49(1):473

81.

Sherry L, Millhouse E, Lappin DF, et al. Investigating the biological properties of carbohydrate derived fulvic acid (CHD-FA) as a potential novel therapy for the management of oral biofilm infections. BMC Oral Health 2013;13:47

82.

Srinivasan A, Uppuluri P, Lopez-Ribot J, Ramasubramanian AK. Development of a high-throughput Candida albicans biofilm chip. PLoS ONE 2011;6(4):e19036

83.

Nett JE, Marchillo K, Spiegel CA, Andes DR. Development and validation of an in vivo Candida albicans biofilm denture model. Infect Immun 2010; 78(9):3650-9

381

Review

Johnson CC, Yu A, Lee H, et al. Development of a contemporary animal model of Candida albicans-associated denture stomatitis using a novel intraoral denture system. Infect Immun 2012;80(5): 1736-43

85.

Wang X, Fries BC. A murine model for catheter-associated candiduria. J Med Microbiol 2011;60(Pt 10):1523-9

86.

Vande Velde G, Kucharikova S, Schrevens S, et al. Towards non-invasive monitoring of

pathogen-host interactions during Candida albicans biofilm formation using in vivo bioluminescence. Cell Microbiol 2013. [Epub ahead of print]

Expert Review of Anti-infective Therapy Downloaded from informahealthcare.com by CDL-UC San Diego on 12/29/14 For personal use only.

84.

Nett

382

Expert Rev. Anti Infect. Ther. 12(3), (2014)

Future directions for anti-biofilm therapeutics targeting Candida.

While proliferating in its most common mode of growth, a biofilm, Candida spp. exhibit increased resistance to available antifungal agents. These adhe...
476KB Sizes 2 Downloads 0 Views