Context.—

Candida auris is an emerging yeast species that was first described in 2009. This ascomycetous yeast is notable for resistance to azole antifungal agents, for environmental persistence, and for its ability to contaminate health care environments, resulting in patient colonization and nosocomial infections.

Objective.—

To review the state of current knowledge addressing challenges in the accurate identification of C auris in the diagnostic microbiology laboratory, including application of phenotypic, proteomic, and genomic methodologies; characteristics that may predispose the human host to acquiring C auris; transmission; clinical presentations; treatment modalities; environmental decontamination; and infection prevention in health care settings.

Data Sources.—

The PubMed search engine was used to access peer-reviewed literature published from 2009 to 2019.

Conclusions.—

The rapid emergence of C auris has presented unique challenges for the areas of laboratory diagnostics and infection prevention and in options for antifungal treatment, which are limited. The current lack of established antifungal susceptibility test breakpoints complicates therapeutic decision making. Enhanced awareness of this pathogen is essential to monitor outbreaks and to reduce the risk of spread within health care environments.

Candida species are one of the leading causes of hospital-acquired bloodstream infections.1,2  The current belief about Candida species is that with good infection control practices they rarely cause outbreaks, and most Candida isolates are generally susceptible to available antifungal agents.3  In the past 20 years, epidemiology has shifted from a predominance of Candida albicans to a predominance of other Candida species.4,5  The recent introduction of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) to the clinical laboratory has facilitated the accurate identification of some rare Candida species.6  Although identification of a rare Candida species is not uncommon in an isolated laboratory setting, it is unprecedented to observe the rapid, simultaneous worldwide emergence of a newly identified Candida species. Yet that is exactly what is happening with Candida auris, which has changed the paradigm of Candida epidemiology. Candida auris causes outbreaks in health care settings, colonizes patients, and contaminates health care environments for extended periods of time. Antifungal resistance is the norm rather than the exception with this yeast species.

In 2009, Satoh and colleagues7  isolated a novel ascomycetous yeast in discharge from the ear canal of a hospitalized patient in Tokyo, Japan. Sequencing results of the internal transcribed spacer (ITS) and D1/D2 regions of the species ribosomal DNA showed only an 87.5% and 85.7% similarity to Candida haemulonii. In addition, sequencing of the D1/D2 regions revealed only an 81.4% and 83.0% similarity to Candida pseudohaemulonii. Subsequent analysis of isolate sequencing data, 42°C growth, and unique patterns of carbon source assimilation as compared with other closely related yeast species supported establishment of a new species type designated as C auris, the Latin term auris referring to the ear. Candida auris is considered to have recently emerged as a pathogen based on retrospective epidemiologic analyses of 15 271 isolates collected from 2004 through 2015. Interrogation of an international surveillance culture collection (SENTRY) indicated a total of 4 isolates confirmed as C auris collected in the years 2009, 2013, 2014, and 2015. A single isolate in this group had been misidentified as C haemulonii during initial analysis; the other 3 isolates were identified only as Candida species. The absence of C auris isolates in the SENTRY collection before 2009 further supported evidence of relatively recent emergence of this new Candida species.8  Less than a decade after the initial identification, C auris was identified on 5 continents.812 

In a study of global epidemiology of C auris, whole-genome sequencing allowed for geographic grouping of C auris into 4 distinct clades: East Asia, South Asia, Africa, and South America. Although isolates from different clades varied by tens of thousands of single-nucleotide polymorphisms, variation within a clade was minimal (0–70 single-nucleotide polymorphisms), suggesting independent clade evolution.8  Since the identification of these 4 clades, patients with C auris have been reported from more than 20 countries. The results of whole-genome sequencing of more than 1150 isolates from 15 countries performed at the Centers for Disease Control and Prevention (CDC) in the United States show that all of the isolates discovered worldwide originate from the previously described 4 clades (CDC, unpublished data, 2018). This indicates that after the simultaneous global emergence, C auris has been spreading from a few major epicenters, followed by regional clonal expansion of highly related isolates.8,13,14 

Culture and Morphologic Characteristics

Macroscopic characteristics of C auris may aid in identification but will not provide sufficient information for definitive differentiation of C auris from other species of Candida. On standard fungal media, such as Sabouraud dextrose agar, C auris appears as white to cream-colored, smooth, butyrous colonies (Figure 1). Growth of C auris can occur at 30°C; however, optimal development is noted at 37°C to 40°C. Growth of C auris at 42°C is slow and weak, and temperature cannot be used to differentiate this species from other closely related yeasts such as C haemulonii. Growth of C auris is inhibited in the presence of cycloheximide. Candida auris will ferment glucose, sucrose, and trehalose and will assimilate carbon sources such as glucose, sucrose, maltose, d-trehalose, d-raffinose, d-melezitose, d-mannitol, sorbitol, citrate, inulin, starch, ribitol, and galactitol.15 

Microscopically, C auris has 2 cellular morphologic features; some strains form aggregates of cells, whereas others have single cells.15  Budding yeast cells are approximately 2.0 to 3.0 × 2.5 to 5.0 μm, with an ovoid to elongate appearance. Similar to Candida glabrata, pseudohyphae are typically not formed on cornmeal agar or rice–Tween 80 agar, although rudimentary pseudohyphae and true hyphae have been reported under certain growth conditions (Figure 2).7,16  Although cellular morphology is traditionally used for the identification of many yeast species, C auris cannot be identified by morphology alone, and differentiation based only on phenotypic features should be discouraged.

Although useful for other species of Candida, chromogenic agar as a stand-alone method may not provide useful information for definitive identification of C auris. On chromogenic agar (eg, CHROMagar), C auris colonies appear beige or pink to red and may look similar to C glabrata (Figure 3).17,18  Difficulties in isolation and identification of C auris may be addressed in part by supplementation of CHROMagar media with Pal (sunflower seed extract) agar. This method has been reported to be useful and rapid in combination with temperature tolerance for differentiation of C auris from the C haemulonii complex (C haemulonii, Candida duobushaemulonii, and C haemulonii var vulnera).19 

Biochemical-Based Identification

Accurately identifying C auris using commercially available phenotypic methods is problematic, which has serious ramifications for treatment and infection control. Definitive identification of C auris and phenotypic differentiation from other closely related yeasts such as C haemulonii, C pseudohaemulonii, and C duobushaemulonii is challenging because of overlapping biochemical profiles and limitations in identification databases. Both nonautomated and automated biochemical methods commonly used in clinical microbiology laboratories have well-described performance issues that lead to inaccurate identification of C auris, primarily from lack of organism-specific biochemical profiles in system databases. This may result in a low confidence result, identification to Candida species only, or outright misidentification. The API 20C AUX clinical yeast system (bioMérieux Inc, Durham, North Carolina) misidentifies C auris as Candida sake, Candida famata, and Saccharomyces cerevisiae. In addition, this system may also misidentify C auris as Rhodotorula glutinis in isolates with a negative urease reaction that lack the characteristic pink-pigmented colonies of R glutinis. Automated identification systems also misidentify C auris as Candida catenulata and C haemulonii by BD Phoenix (Becton Dickinson and Company, Franklin Lakes, New Jersey); as C duobushaemulonii, C haemulonii, and C famata by Vitek 2 (bioMérieux); and as C catenulata, C famata, Candida guilliermondii, Candida lusitaniae, and Candida parapsilosis by MicroScan WalkAway Plus (Beckman Coulter, Inc, Brea, California).15,20  Informational tables summarizing limitations of US Food and Drug Administration (FDA)–approved contemporary commercial identification systems may be found at the CDC Web site21  and in Mizusawa et al.20  Laboratories with these organism identification/instrument combinations or that have a genus-only result of Candida should continue testing to determine if the isolate in question is C auris. Useful identification algorithms based on common identifications generated by commercial systems are available at the CDC Web site.21  Users of biochemical methods to identify yeasts should maintain awareness for updates in system databases that may allow for more accurate identification of C auris. Even if an updated database is used, identification of species commonly misidentified as C auris warrants additional investigation in order to verify results.

Use of MALDI-TOF MS

Proteomic methods such as MALDI-TOF MS can identify C auris rapidly and accurately with substantially better performance than phenotypic systems. The 2 MALDI-TOF platforms used most widely in clinical microbiology laboratories are the Bruker BioTyper (Bruker Daltonics, Billerica, Massachusetts) and the Vitek MS (bioMérieux). For MALDI-TOF MS, protein is first extracted from an isolate of interest in pure culture and applied to a target plate or slide. Then, MALDI-TOF MS generates a spectral profile of the protein and compares the profile with a reference database, identifying the isolate to the genus and species level. The method used to extract protein from the organism, coupled with the identification database used, affects the quality of identification. With respect to C auris, direct-on-target extraction methods have been shown to be successful with Vitek MS but may lead to a “no identification” result with the Bruker BioTyper. A tube-extraction method can be used with both systems but is slightly more time-consuming and labor-intensive.20  A modified direct-on-target method for protein extraction has been described that generated acceptable spectral scores for identification of C auris without the need for full-tube extraction.22 

As with biochemical methods, the quality of the reference database directly affects the identification accuracy of MALDI-TOF MS. Generally, both the Bruker and Vitek MS systems have the capability to accurately identify C auris using the research-use–only (RUO) spectral databases supplied with each instrument. Kathuria et al23  reported excellent identification of C auris and differentiation from C famata, C haemulonii, C haemulonii var vulnera, and C duobushaemulonii using MALDI-TOF MS. If an RUO library is used, users should confirm that the corresponding database version specifically contains reference spectra for C auris. In some cases, updated databases or software patches may be required to obtain high-confidence spectral scores.15,17  At this time, only the Bruker system contains C auris in the FDA-approved spectral database. Therefore, if a laboratory wants to use Vitek MS to identify C auris isolates, an RUO database must be used that includes reference spectra for C auris.17  It is also possible for a laboratory, using a manufacturer's protocols, to create its own custom database library.24 

In addition to these database options, the CDC, in collaboration with Bruker, offers an online tool to provide accurate classification of C auris to the species level (https://www.cdc.gov/microbenet/index.html).25  The tool contains expert curated information and access to CDC spectral libraries. Users may upload laboratory-generated MALDI-TOF MS information to a CDC database containing proteomic data for organism identification by in silico comparison. If a laboratory elects to identify C auris using MALDI-TOF MS, a review of the contents of instrument databases (RUO or FDA-approved) for C auris is strongly encouraged in order to achieve optimal performance and accuracy with these systems. In addition, well-characterized isolates of C auris are available to qualified laboratories as part of the CDC antibiotic resistance organism bank to assist with verification or validation of FDA-approved or RUO organism identification databases (https://www.cdc.gov/drugresistance/resistance-bank/index.html).

Molecular-Based Identification and Strain Typing

Clearly, identification of C auris based solely on phenotypic, biochemical, or proteomic characteristics is a challenge because of the inherent characteristics of the organism and limitations of commercial identification systems. Candida auris can also be identified through molecular methods, such as conventional and real-time polymerase chain reaction (PCR) assays, which are rapid to perform and highly accurate. Kordalewska et al26  developed an initial assay that specifically and accurately detected C auris. This group subsequently used a different set of PCR primers in an additional assay that enabled differentiation of C auris from other closely related species, such as C lusitaniae, C haemulonii, and C duobushaemulonii, by using SYBR Green (ThermoFisher Scientific, Waltham, Massachusetts) detection of amplicon and melting-point analysis. In an analysis of a 140-member proficiency panel, both PCR assays yielded an accuracy of 100% and complete concordance with sequencing results for ITS regions.26 

Definitive identification of C auris to species level may also be achieved through molecular sequencing of the 18S ITS regions or the D1/D2 regions of fungal ribosomal DNA.17,23  However, the financial investment and technical requirements needed for sequencing may preclude the routine use of these methods for identifying organisms in most clinical laboratories.25  A diagnostic modality using magnetic resonance for detection of specific Candida species directly from blood specimens is available, and an assay specific for identification of C auris using this technology is currently being developed.27  In addition, isolates may be submitted to regional Antibiotic Resistance Laboratory Network facilities (https://www.cdc.gov/drugresistance/laboratories/AR-lab-network-testing-details.html) or to many commercial reference laboratories for identification of C auris.

Traditional methods of strain typing, such as amplified fragment-length polymorphism or multilocus sequence typing, have been used in an attempt to type C auris.10  However, the limited variability within C auris clades may impede the utility of strain typing for epidemiologic studies of local transmission or infection control within health care facilities.28  Whole-genome sequencing remains state of the art for determination of clonality of C auris isolates.

Clinical Significance of C auris Infections

The clinical significance of C auris is considerable because of its rapid worldwide emergence, ability to cause invasive infections associated with high mortality, propensity to show resistance to multiple classes of antifungal agents, and ability to spread in health care settings and cause outbreaks. Patients most at risk for C auris infections have had extensive exposure to health care (eg, multiple admissions to long-term and acute care facilities) in the months preceding C auris diagnosis; indwelling devices such as tracheostomy tubes, percutaneous gastrostomy tubes, or central vascular access ports/catheters; or recent exposure to broad-spectrum antibiotics and antifungal agents. Nosocomial transmission has been reported to occur in both acute and long-term care settings, although long-term care facilities, especially skilled nursing facilities with patients who require ventilators, have generally had the highest prevalence of patients with C auris colonization.12,14  The ability of C auris to colonize in patients long-term and persist on both moist and dry surfaces for weeks is an additional factor considered highly contributory to transmission and recurrence within health care settings.15,29,30  Fungal virulence factors such as proteinases, phospholipases, and hemolysins have been shown to express diverse levels of activity in vitro, suggesting inherent variability of virulence among different strains of C auris.15  However, mouse and invertebrate experimental models of infection suggest the overall virulence of C auris may be comparable to that of C albicans.16,3133 

Candida auris has been implicated in a variety of superficial and invasive infections from multiple body sites. After the first reported case (2009) of C auris in a Japanese patient with otitis media,7  another 15 cases were reported from 5 hospitals in South Korea; all the patients also had chronic otitis media.34  Using the D1/D2 domain and ITS sequence analysis, all 15 isolates were determined to be a novel species (ultimately recognized as C auris), and 10 showed notable resistance to fluconazole (minimum inhibitory concentration [MIC] ≥32 μg/mL). Interestingly, all of these patients had a history of previous antibiotic treatment and manipulation of the ear canal.

The bloodstream is by far the most common site of isolation of C auris from clinical cultures. In a meta-analysis of reports from 2012 to 2017 describing 742 confirmed C auris isolates from 16 countries, 67% were isolated from blood.15  The bloodstream was the culture source for 5 of the first 7 patients infected with C auris in the United States (from 2013 to 2017); all of these patients had serious underlying medical conditions.35  Reports from various locations worldwide have noted an association between C auris fungemia with indwelling catheters (venous or urinary), use of broad-spectrum antibiotics, parenteral nutrition, surgery, and extended stay in an intensive care unit, in addition to underlying comorbid conditions, such as diabetes mellitus and malignancy.9,3638  Determining overall mortality attributable to C auris fungemia is difficult because of multiple confounding comorbid conditions in many patients.39  However, estimates of overall mortality from C auris fungemia range from 28% to 60%.8,9 

Although bloodstream infections are most common, C auris has also been isolated from diverse body sites, such as wounds; bone; urine; vagina; and cerebrospinal, peritoneal, pleural, and pericardial fluids.15  Isolates of C auris have also been found in tissues excised from gangrenous feet and postoperative wounds.40  Among US patients, approximately 46% of C auris infections have been identified from sites other than blood.60 

In a survey of cases with confirmed C auris isolated from bone, urine, peritoneal fluid, and cerebrospinal fluid, misidentification of the organism along with increased resistance of C auris to antifungal agents contributed to a 30-day mortality rate of 35.2%.37  A case report of fatal C auris pericarditis in a patient with chronic liver disease who was receiving empiric fluconazole therapy is notable for the concurrent recovery of C auris from urine, bronchoalveolar lavage, and blood.41  A recent report of donor-derived, transplant-transmitted C auris associated with a bilateral lung transplant suggests reconsideration of the clinical significance of yeast in donor lung surveillance cultures.42 

Regardless of the source of the infection, specimen, or underlying cause of disease, delayed diagnosis or misidentification of C auris will affect selection of appropriate antifungal therapy and impact infection control efforts, and may ultimately affect patient outcome. In the assessment of yeasts isolated from nonsterile sites (eg, skin, vaginal, upper respiratory), many laboratories will report “yeast, not Cryptococcus” with no additional speciation. At this time, there are no guidelines supporting routine assessment of nonsterile site specimens for the presence of C auris. However, in areas where C auris has been reported, both clinicians and laboratorians should be cognizant of this possibility and request additional identification studies, as appropriate.

Emerging evidence suggests that immune system suppression attributable to malignancy or immunosuppressive agents (eg, corticosteroids) is a risk factor for C auris infection and may contribute to dissemination of infection in the compromised host. In a 2013 report of C auris isolated from the bloodstream in 12 patients in India, 11 patients were considered immunosuppressed because of a wide variety of conditions, such as diabetes mellitus, chronic kidney disease, HIV, cancer chemotherapy, or a stay in the intensive care unit.10  A survey of the first C auris cases in the United States also showed that infection developed in association with serious underlying conditions, such as malignancy and corticosteroid use, that resulted in immunosuppression.35 

One of the most concerning features of C auris is its reduced susceptibility to azoles, polyenes, and, in some instances, echinocandins. This feature severely limits the antifungal treatment options for infected patients, especially given the diminished health status of many of these patients. Triazoles, including fluconazole, are an important therapy for Candida infections. Early susceptibility reports for C auris suggested the organism might be intrinsically resistant to fluconazole, as nearly all early isolates evaluated displayed elevated fluconazole MICs.8  For instance, a study of 350 Indian isolates showed 315 (90%) with fluconazole MICs greater than 16 μg/mL.38  Further study of C auris showed that isolates from other geographic locations had fluconazole MICs of 2 to 8 μg/mL, similar to those for C glabrata, suggesting that azole resistance in C auris is acquired and not intrinsic.25  The azoles prevent Candida cell growth by inhibiting the cell's sterol pathway, specifically the 14-α demethylation of ergosterol precursors. This enzyme is encoded by the Erg11 gene. Ergosterol is an essential component of the fungal cell membrane, and inhibition of ergosterol biosynthesis leads to accumulation of toxic methylated sterols, arresting cell growth. Substitutions to the Erg11p azole target have been described for C auris, leading to an altered protein structure, reduced azole binding affinity, and increased azole MICs. The specific mutations to Erg11p observed to date in C auris are associated with geographic clades, such that isolates from South Africa and Venezuela share F126L alterations, whereas Indian isolates harbor Y132F or K143R substitutions.8  Candida auris can exhibit reduced susceptibility to other triazole antifungals, including voriconazole, posaconazole, itraconazole, and isavuconazole, but most isolates have relatively low MIC values.43 

Variable susceptibility is exhibited by C auris isolates to the polyene amphotericin B. Again, susceptibility to this antifungal agent is defined geographically and may be affected when cases are imported from multiple geographic locations.44  Most isolates from India have MICs less than 2 μg/mL,43  but up to 64% of isolates in a Colombian study45  had amphotericin B MICs greater than 1 μg/mL. The mechanisms that define amphotericin B resistance in C auris are not yet defined and are poorly defined for other Candida species; therefore, readers of case reports must be mindful of selection bias, which tends to present a picture of increased amphotericin B resistance.

Echinocandins are used as first-line therapy for C auris, pending antifungal susceptibility test results, unless the patient is an infant less than 2 months of age, in which case amphotericin B deoxycholate is recommended. Development of echinocandin resistance (ie, MICs >1 μg/mL for caspofungin) has been documented in C auris isolates recovered from patients in multiple geographic areas who were initially treated with an echinocandin. Echinocandins act by targeting the fungal-specific β-glucan synthase, which synthesizes a major cell wall polymer. Mutation to the Fks subunits of the glucan synthase in hot-spot regions decreases susceptibility of fungi to echinocandins. Isolates of C auris with elevated echinocandin MICs harbor the mutation S639F in the position equivalent to the well-characterized S645 FKS1 hot-spot 1 region.43  General guidance on treatment recommendations may be found at https://www.cdc.gov/fungal/candida-auris/c-auris-treatment.html. It should be noted that for cases of C auris, treatment is indicated only if clinical disease is present or if the organism is isolated from a sterile site. Treatment is not recommended when C auris is identified in a noninvasive infection or if the patient exhibits no clinical signs of infection.

Interpretation of reduced susceptibility to azoles, polyenes, and echinocandins is challenging because no antifungal breakpoints for C auris have been defined by the Clinical and Laboratory Standards Institute (CLSI) or the European Committee on Antimicrobial Susceptibility Testing. However, tentative epidemiologic cutoff values (ECVs) have been established for both the CLSI and European Committee on Antimicrobial Susceptibility Testing methods, which differ slightly.40  It should be cautioned that these ECVs are tentative, as the study used to derive them was not performed according to European Committee on Antimicrobial Susceptibility Testing or CLSI specifications for establishing ECVs. Furthermore, ECVs are not a clinical breakpoint, and isolates with MICs below the ECV should not be reported as susceptible, nor should isolates with MICs above the ECV be considered resistant. Insufficient pharmacodynamic and clinical data are available to establish clinical breakpoints for this species. The CLSI guidance on interpreting ECVs can be found in the CLSI M59 standard.46  On the basis of ECVs and identified molecular mechanisms of resistance, the CDC has established tentative breakpoints. These breakpoints are informational only and have not been approved by any regulatory agency or standardization committee (Table).21 

Analytical challenges can make evaluating antifungal susceptibility test results for C auris a daunting task. Kordalewska et al26  documented a caspofungin Eagle effect for isolates of C auris that lacked the FKS1 mutation associated with echinocandin resistance. The Eagle effect, also known as a paradoxical effect, occurs when growth is observed at higher but not lower concentrations of drug, possibly because of dose-dependent echinocandin tolerance in response to cell-wall damage.47  The isolates showed in vivo susceptibility to caspofungin at human therapeutic doses in a murine model of invasive candidiasis. Mixed results have been obtained when evaluating C auris susceptibility by commercial antifungal susceptibility test systems. Kathuria et al23  noted that the Vitek 2 generated higher amphotericin B MICs than did the CLSI broth microdilution reference method for a collection of 90 Indian C auris isolates, with MIC50 of 8 μg/mL and 1 μg/mL, respectively. In contrast, Etest (bioMérieux) was comparable to broth microdilution for this drug, with an MIC50 of 0.5 μg/mL. Caspofungin and voriconazole MICs were similar among the 3 methods. Magobo et al11  reported falsely high caspofungin MICs by using the Vitek 2, which is particularly concerning because caspofungin is recommended as first-line therapy for C auris infections.

Because resistance of C auris to antifungal agents has been shown to emerge while a patient is receiving therapy, repeated susceptibility testing should be performed if a laboratory isolates C auris on subsequent cultures. In the United States, isolates should also be referred to the local public health laboratory for confirmatory identification and antifungal susceptibility testing. As there are no CLSI- or FDA-approved specific interpretative breakpoints for C auris susceptibility testing, MIC-only results should not be reported.

Patients with C auris infections often become colonized with C auris at multiple body sites, which can include the axilla, groin, nares, ears, mouth, rectum, and vagina, but most often C auris is detected in the axilla and the groin.12,14,48  A contaminated patient room or intensive care unit can lead to transmission of C auris and subsequent colonization or infection, or both, of other patients. In one hospital, more than 50% of C auris infections were preceded by colonization of the patient, which emphasizes the need to prevent colonization and to identify those who are colonized.48 

Both culture-based and molecular-based techniques for the detection of C auris colonization have been developed.29,49  The culture technique is based on the ability of C auris to grow at 10% NaCl and 40°C and to use dulcitol as a carbon source to enrich for C auris over other Candida species that colonize the skin.29  This technique has been successfully used during multiple surveys of health care facilities to detect patients colonized with C auris.14,35  The molecular assay is a TaqMan-based real-time PCR assay that targets the ITS region of the ribosomal cistron.49  Although it only has 89% sensitivity for detection of C auris from swabs compared with culture, it reduces the overall time to positivity from days to hours, allowing the rapid implementation of infection control procedures. There is an additional SYBR Green detection real-time PCR assay that has been developed, but this assay has not yet been optimized for use with specimens collected on swabs.26 

It is unlikely that persistently colonized health care personnel serve as a major source of transmission. During outbreaks in both England and Spain, health care personnel were screened for C auris colonization. Multiple sites of 258 health care workers in England were screened; sites included the hands, nares, axilla, and groin. Only a single positive nares swab was found, and colonization in that person was transient.12  In Spain, the hands and ears of 101 personnel who worked in a surgical intensive care unit were screened, and none were positive.48  However, it is possible that health care personnel who are transiently colonized moving from a colonized or infected patient to a subsequent patient or from a contaminated environmental surface to a patient could transmit C auris in health care settings.

Decolonizing skin has proven to be an additional challenge. Chlorhexidine wipes (2%) with isopropyl alcohol are effective in eliminating C auris from surfaces, as is povidone iodine. However, their efficacy for skin has not been proven.50,51  In vitro, 2% and 4% chlorhexidine have been shown to be effective against C auris, but the effect on decolonization of patients is not known. Even if the amount of C auris on the skin can be reduced through the application of these antiseptics, it is unclear what effect this will have on transmission because patients may continue to harbor reservoirs of C auris in the ears, nares, gastrointestinal, or genitourinary tracts.

In health care settings, C auris has been recovered from mattresses, beds, chairs, tables, floors, walls, carts, windowsills, equipment monitors and keypads, and countertops.12,35,48  Several reports have shown that C auris can persist for 7 to 14 days on surfaces such as steel, ceramic, and plastic.29,52  There is some evidence that C auris may enter a viable, nonculturable state for up to 4 weeks on some surfaces.29  Quaternary ammonium compounds alone, which are the most common disinfectants used in health care settings, might not achieve the desired log reduction against C auris and are therefore not recommended for use. Many chlorine-based and peroxygen-based disinfectants appear to be effective against C auris and have shown substantial log reductions on surfaces and in solution (≥4 log10).51,53,54  Other disinfectant products that have been shown to achieve substantial reductions in C auris include some quaternary ammonium compounds with alcohol.29,50,53,54  Mobile, short-wavelength ultraviolet light devices are also effective in reducing the bioburden of C auris substantially, and exposure times are less than those required for Clostridioides (Clostridium) difficile.55 

Candida species have not previously been considered organisms for which contact precautions were necessary. However, there is ample evidence that C auris can be transmitted from person to person in health care facilities.12,14,48  Therefore, it is imperative that C auris is identified accurately by the laboratory so that infection control procedures can be put in place. The CDC has published guidance on infection control measures for C auris (https://www.cdc.gov/fungal/candida-auris/c-auris-infection-control.html).56  Patients infected or colonized with C auris should be placed in a single-patient room and contact precautions instigated. Meticulous hand hygiene with an alcohol-based hand rub or soap and water is required for health care personnel caring for patients with C auris. Cleaning and disinfection of the patient room should take place daily, and then a terminal cleaning should be done before other patients use the room. Currently, the CDC recommends the use of an Environmental Protection Agency–registered disinfectant that is effective against C difficile spores (List K [https://www.epa.gov/pesticide-registration/list-k-epas-registered-antimicrobial-products-effective-against-clostridium])57  until Environmental Protection Agency–registered products specifically designated for C auris are available. Lastly, close contacts of newly identified patients with C auris should be screened for colonization by swabbing of the axilla and groin.58 

Because of the clinical and epidemiologic implications of C auris infection, diagnostic laboratories are advised to identify to species level all Candida organisms isolated from sterile body sites, Candida isolates with unusual antifungal resistance profiles, isolates from patients with possible colonization based on known exposure or risk factors, or isolates from patients with prior C auris exposure.59,60  If C auris is suspected, it is critical for clinical laboratories to partner with institutional infection preventionists to complete the clinical and epidemiologic details pertinent to the case in order to reduce the risk of nosocomial transmission. Isolates may be referred to reference laboratories, state public health laboratories, or the CDC for assistance with definitive identification. Recommendations for C auris identification may be found at https://www.cdc.gov/fungal/diseases/candidiasis/recommendations.html#identify. If a clinical laboratory has the capacity to confirm identification of suspect isolates, every report of C auris should be immediately conveyed to local and state health officials in addition to the CDC via candidaauris@cdc.gov.

In the clinical microbiology laboratory, identification of C auris in an accurate and timely fashion is challenging but essential for patient care and infection prevention. Clinical laboratories must assess their ability to identify C auris based on the methodology they use. Antifungal susceptibility profiles and interpretations in addition to treatment recommendations for patients with C auris must be understood for appropriate clinical management. Because of the ability of C auris to spread in health care facilities and cause outbreaks, rapid communication of suspected cases with institutional infection prevention providers and local, state, and national surveillance networks is essential to enhance awareness of this newly emerging yeast species.

We gratefully acknowledge the contributions of the College of American Pathologists Microbiology Committee for concept development, the Mycotic Diseases Branch at the Centers for Disease Control and Prevention for technical expertise, and Megan Bentz, BS, MS, for laboratory photography. We also gratefully acknowledge Diana M. Meza Villegas, MS, SM(ASCP)CM, and Miranda E. Diaz, BS, SM(ASCP)CM, for data collection and photography, and the Mayo Clinic Clinical Microbiology Laboratory in Jacksonville, Florida, for laboratory support.

1
Magill
SS
,
Edwards
JR
,
Bamberg
W
, et al.
Multistate point-prevalence survey of health care-associated infections
.
N Engl J Med
.
2014
;
370
(
13
):
1198
1208
.
2
Vincent
JL
,
Rello
J
,
Marshall
J
, et al.
International study of the prevalence and outcomes of infection in intensive care units
.
JAMA
.
2009
;
302
(
21
):
2323
2329
.
3
Lamoth
F
,
Lockhart
SR
,
Berkow
EL
,
Calandra
T.
Changes in the epidemiological landscape of invasive candidiasis
.
J Antimicrob Chemother
.
2018
;
73
(
suppl 1
):
i4
i13
.
4
Lockhart
SR
,
Iqbal
N
,
Cleveland
AA
, et al.
Species identification and antifungal susceptibility testing of Candida bloodstream isolates from population-based surveillance studies in two U.S. cities from 2008 to 2011
.
J Clin Microbiol
.
2012
;
50
(
11
):
3435
3442
.
5
Pfaller
MA
,
Jones
RN
,
Castanheira
M.
Regional data analysis of Candida non-albicans strains collected in United States medical sites over a 6-year period, 2006–2011
.
Mycoses
.
2014
;
57
(
10
):
602
611
.
6
Wang
H
,
Fan
YY
,
Kudinha
T
, et al.
A comprehensive evaluation of the Bruker Biotyper MS and Vitek MS matrix-assisted laser desorption ionization–time of flight mass spectrometry systems for identification of yeasts, part of the National China Hospital Invasive Fungal Surveillance Net (CHIF-NET) study, 2012 to 2013
.
J Clin Microbiol
.
2016
;
54
(
5
):
1376
1380
.
7
Satoh
K
,
Makimura
K
,
Hasumi
Y
,
Nishiyama
Y
,
Uchida
K
,
Yamaguchi
H.
Candida auris sp. nov., a novel ascomycetous yeast isolated from the external ear canal of an inpatient in a Japanese hospital
.
Microbiol Immunol
.
2009
;
53
(
1
):
41
44
.
8
Lockhart
SR
,
Etienne
KA
,
Vallabhaneni
S
, et al.
Simultaneous emergence of multidrug-resistant Candida auris on 3 continents confirmed by whole-genome sequencing and epidemiological analyses
.
Clin Infect Dis
.
2017
;
64
(
2
):
134
140
.
9
Calvo
B
,
Melo
AS
,
Perozo-Mena
A
, et al.
First report of Candida auris in America: clinical and microbiological aspects of 18 episodes of candidemia
.
J Infect
.
2016
;
73
(
4
):
369
374
.
10
Chowdhary
A
,
Sharma
C
,
Duggal
S
, et al.
New clonal strain of Candida auris, Delhi, India
.
Emerg Infect Dis
.
2013
;
19
(
10
):
1670
1673
.
11
Magobo
RE
,
Corcoran
C
,
Seetharam
S
,
Govender
NP
.
Candida auris-associated candidemia, South Africa
.
Emerg Infect Dis
.
2014
;
20
(
7
):
1250
1251
.
12
Schelenz
S
,
Hagen
F
,
Rhodes
JL
, et al.
First hospital outbreak of the globally emerging Candida auris in a European hospital
.
Antimicrob Resist Infect Control
.
2016
;
5
:
35
.
13
Rhodes
J
,
Abdolrasouli
A
,
Farrer
RA
, et al.
Genomic epidemiology of the UK outbreak of the emerging human fungal pathogen Candida auris
.
Emerg Microbes Infect
.
2018
;
7
(
1
):
43
.
14
Tsay
S
,
Welsh
RM
,
Adams
EH
, et al.
Notes from the field: ongoing transmission of Candida auris in health care facilities—United States, June 2016–May 2017
.
MMWR Morb Mortal Wkly Rep
.
2017
;
66
(
19
):
514
515
.
15
Osei Sekyere
J.
Candida auris: a systematic review and meta-analysis of current updates on an emerging multidrug-resistant pathogen
.
Microbiologyopen
.
2018
;
7
(
4
):
e00578
.
16
Yue
H
,
Bing
J
,
Zheng
Q
, et al.
Filamentation in Candida auris, an emerging fungal pathogen of humans: passage through the mammalian body induces a heritable phenotypic switch
.
Emerg Microbes Infect
.
2018
;
7
(
1
):
188
.
17
Spivak
ES
,
Hanson
KE
.
Candida auris: an emerging fungal pathogen
.
J Clin Microbiol
.
2018
;
56
(
2
):
1
10
.
18
Bentz
ML
,
Sexton
DJ
,
Welsh
RM
,
Litvintseva
AP
.
Phenotypic switching in newly emerged multidrug-resistant pathogen Candida auris
[
published online
October
16,
2018
].
Med Mycol
. doi:
19
Kumar
A
,
Sachu
A
,
Mohan
K
,
Vinod
V
,
Dinesh
K
,
Karim
S.
Simple low cost differentiation of Candida auris from Candida haemulonii complex using CHROMagar Candida medium supplemented with Pal's medium
.
Rev Iberoam Micol
.
2017
;
34
(
2
):
109
111
.
20
Mizusawa
M
,
Miller
H
,
Green
R
, et al.
Can multidrug-resistant Candida auris be reliably identified in clinical microbiology laboratories?
J Clin Microbiol
.
2017
;
55
(
2
):
638
640
.
21
Centers for Disease Control and Prevention
.
Identification of Candida auris
.
https://www.cdc.gov/fungal/candida-auris/recommendations.html. Published 2018. Accessed February 20, 2019
.
22
Fraser
M
,
Brown
Z
,
Houldsworth
M
,
Borman
AM
,
Johnson
EM
.
Rapid identification of 6328 isolates of pathogenic yeasts using MALDI-ToF MS and a simplified, rapid extraction procedure that is compatible with the Bruker Biotyper platform and database
.
Med Mycol
.
2016
;
54
(
1
):
80
88
.
23
Kathuria
S
,
Singh
PK
,
Sharma
C
, et al.
Multidrug-resistant Candida auris misidentified as Candida haemulonii: characterization by matrix-assisted laser desorption ionization–time of flight mass spectrometry and DNA sequencing and its antifungal susceptibility profile variability by Vitek 2, CLSI broth microdilution, and Etest method
.
J Clin Microbiol
.
2015
;
53
(
6
):
1823
1830
.
24
Bao
JR
,
Master
RN
,
Azad
KN
, et al.
Rapid, accurate identification of Candida auris by using a novel matrix-assisted laser desorption ionization–time of flight mass spectrometry (MALDI-TOF MS) database (library)
.
J Clin Microbiol
.
2018
;
56
(
4
):
1
3
.
25
Lockhart
SR
,
Berkow
EL
,
Chow
N
,
Welsh
RM
.
Candida auris for the clinical microbiology laboratory: not your grandfather's Candida species
.
Clin Microbiol Newsl
.
2017
;
39
(
13
):
99
103
.
26
Kordalewska
M
,
Zhao
Y
,
Lockhart
SR
,
Chowdhary
A
,
Berrio
I
,
Perlin
DS
.
Rapid and accurate molecular identification of the emerging multidrug-resistant pathogen Candida auris
.
J Clin Microbiol
.
2017
;
55
(
8
):
2445
2452
.
27
Sexton
DJ
,
Bentz
ML
,
Welsh
RM
,
Litvintseva
AP
.
Evaluation of a new T2 magnetic resonance assay for rapid detection of emergent fungal pathogen Candida auris on clinical skin swab samples
.
Mycoses
.
2018
;
61
(
10
):
786
790
.
28
Lesho
EP
,
Bronstein
MZ
,
McGann
P
, et al.
Importation, mitigation, and genomic epidemiology of Candida auris at a large teaching hospital
.
Infect Control Hosp Epidemiol
.
2018
;
39
(
1
):
53
57
.
29
Welsh
RM
,
Bentz
ML
,
Shams
A
, et al.
Survival, persistence, and isolation of the emerging multidrug-resistant pathogenic yeast Candida auris on a plastic health care surface
.
J Clin Microbiol
.
2017
;
55
(
10
):
2996
3005
.
30
Adams
E
,
Quinn
M
,
Tsay
S
, et al.
Candida auris in healthcare facilities, New York, USA, 2013–2017
.
Emerg Infect Dis
.
2018
;
24
(
10
):
1816
1824
.
31
Lamoth
F
,
Kontoyiannis
DP
.
The Candida auris alert: facts and perspectives
.
J Infect Dis
.
2018
;
217
(
4
):
516
520
.
32
Borman
AM
,
Szekely
A
,
Johnson
EM
.
Comparative pathogenicity of United Kingdom isolates of the emerging pathogen Candida auris and other key pathogenic Candida species
.
mSphere
.
2016
;
1
(
4
):
1
8
.
33
Ben-Ami
R
,
Berman
J
,
Novikov
A
, et al.
Multidrug-resistant Candida haemulonii and C. auris, Tel Aviv, Israel
.
Emerg Infect Dis
.
2017
;
23
(
1
):
195
203
.
34
Kim
MN
,
Shin
JH
,
Sung
H
, et al.
Candida haemulonii and closely related species at 5 university hospitals in Korea: identification, antifungal susceptibility, and clinical features
.
Clin Infect Dis
.
2009
;
48
(
6
):
e57
e61
.
35
Vallabhaneni
S
,
Kallen
A
,
Tsay
S
, et al.
Investigation of the first seven reported cases of Candida auris, a globally emerging invasive, multidrug-resistant fungus—United States, May 2013–August 2016
.
MMWR Morb Mortal Wkly Rep
.
2016
;
65
(
44
):
1234
1237
.
36
Lee
WG
,
Shin
JH
,
Uh
Y
, et al.
First three reported cases of nosocomial fungemia caused by Candida auris
.
J Clin Microbiol
.
2011
;
49
(
9
):
3139
3142
.
37
Morales-Lopez
SE
,
Parra-Giraldo
CM
,
Ceballos-Garzon
A
, et al.
Invasive infections with multidrug-resistant yeast Candida auris, Colombia
.
Emerg Infect Dis
.
2017
;
23
(
1
):
162
164
.
38
Rudramurthy
SM
,
Chakrabarti
A
,
Paul
RA
, et al.
Candida auris candidaemia in Indian ICUs: analysis of risk factors
.
J Antimicrob Chemother
.
2017
;
72
(
6
):
1794
1801
.
39
Jeffery-Smith
A
,
Taori
SK
,
Schelenz
S
, et al.
Candida auris: a review of the literature
.
Clin Microbiol Rev
.
2018
;
31
(
1
):
1
18
.
40
Chowdhary
A
,
Anil Kumar
V
,
Sharma
C
, et al.
Multidrug-resistant endemic clonal strain of Candida auris in India
.
Eur J Clin Microbiol Infect Dis
.
2014
;
33
(
6
):
919
926
.
41
Khillan
V
,
Rathore
N
,
Kathuria
S
,
Chowdhary
A.
A rare case of breakthrough fungal pericarditis due to fluconazole-resistant Candida auris in a patient with chronic liver disease
.
JMM Case Rep
.
2014
;
1
(
3
):
1
5
.
42
Azar
MM
,
Turbett
SE
,
Fishman
JA
,
Pierce
VM
.
Donor-derived transmission of Candida auris during lung transplantation
.
Clin Infect Dis
.
2017
;
65
(
6
):
1040
1042
.
43
Chowdhary
A
,
Prakash
A
,
Sharma
C
, et al.
A multicentre study of antifungal susceptibility patterns among 350 Candida auris isolates (2009–17) in India: role of the ERG11 and FKS1 genes in azole and echinocandin resistance
.
J Antimicrob Chemother
.
2018
;
73
(
4
):
891
899
.
44
Chow
NA
,
Gade
L
,
Tsay
SV
, et al.
Multiple introductions and subsequent transmission of multidrug-resistant Candida auris in the USA: a molecular epidemiological survey
.
Lancet Infect Dis
.
2018
;
18
(
12
):
1377
1384
.
45
Escandon
P
,
Chow
NA
,
Caceres
DH
, et al.
Molecular epidemiology of Candida auris in Colombia reveals a highly related, countrywide colonization with regional patterns in amphotericin B resistance
.
Clin Infect Dis
.
2019
;
68
(
1
):
15
21
.
46
Clinical and Laboratory Standards Institute
.
M59: Epidemiological Cutoff Values for Antifungal Susceptibility Testing. 2nd ed
.
Wayne, PA
:
Clinical and Laboratory Standards Institute;
2018
:
28
.
47
Steinbach
WJ
,
Lamoth
F
,
Juvvadi
PR
.
Potential microbiological effects of higher dosing of echinocandins
.
Clin Infect Dis
.
2015
;
61
(
suppl 6
):
S669
S677
.
48
Ruiz-Gaitan
A
,
Moret
AM
,
Tasias-Pitarch
M
, et al.
An outbreak due to Candida auris with prolonged colonisation and candidaemia in a tertiary care European hospital
.
Mycoses
.
2018
;
61
(
7
):
498
505
.
49
Leach
L
,
Zhu
Y
,
Chaturvedi
S.
Development and validation of a real-time PCR assay for rapid detection of Candida auris from surveillance samples
.
J Clin Microbiol
.
2018
;
56
(
2
):
1
7
.
50
Abdolrasouli
A
,
Armstrong-James
D
,
Ryan
L
,
Schelenz
S.
In vitro efficacy of disinfectants utilised for skin decolonisation and environmental decontamination during a hospital outbreak with Candida auris
.
Mycoses
.
2017
;
60
(
11
):
758
763
.
51
Moore
G
,
Schelenz
S
,
Borman
AM
,
Johnson
EM
,
Brown
CS
.
Yeasticidal activity of chemical disinfectants and antiseptics against Candida auris
.
J Hosp Infect
.
2017
;
97
(
4
):
371
375
.
52
Piedrahita
CT
,
Cadnum
JL
,
Jencson
AL
,
Shaikh
AA
,
Ghannoum
MA
,
Donskey
CJ
.
Environmental surfaces in healthcare facilities are a potential source for transmission of Candida auris and other Candida species
.
Infect Control Hosp Epidemiol
.
2017
;
38
(
9
):
1107
1109
.
53
Cadnum
JL
,
Shaikh
AA
,
Piedrahita
CT
, et al.
Effectiveness of disinfectants against Candida auris and other Candida species
.
Infect Control Hosp Epidemiol
.
2017
;
38
(
10
):
1240
1243
.
54
Kean
R
,
Sherry
L
,
Townsend
E
, et al.
Surface disinfection challenges for Candida auris: an in-vitro study
.
J Hosp Infect
.
2018
;
98
(
4
):
433
436
.
55
Cadnum
JL
,
Shaikh
AA
,
Piedrahita
CT
, et al.
Relative resistance of the emerging fungal pathogen Candida auris and other Candida species to killing by ultraviolet light
.
Infect Control Hosp Epidemiol
.
2018
;
39
(
1
):
94
96
.
56
Centers for Disease Control and Prevention
.
Infection prevention and control for Candida auris
. .
57
United States Environmental Protection Agency
.
LIST K: EPA's registered antimicrobial products effective against Clostridium difficile spores
. .
58
Sexton
DJ
,
Kordalewska
M
,
Bentz
ML
,
Welsh
RM
,
Perlin
DS
,
Litvintseva
AP
.
Direct detection of emergent fungal pathogen Candida auris in clinical skin swabs by SYBR Green-based quantitative PCR assay
.
J Clin Microbiol
.
2018
;
56
(
12
):
1
6
.
59
Lockhart
SR
,
Jackson
BR
,
Vallabhaneni
S
,
Ostrosky-Zeichner
L
,
Pappas
PG
,
Chiller
T.
Thinking beyond the common Candida species: need for species-level identification of Candida due to the emergence of multidrug-resistant Candida auris
.
J Clin Microbiol
.
2017
;
55
(
12
):
3324
3327
.
60
Centers for Disease Control and Prevention
.
Candida auris clinical update—September 2017
. .
61
Centers for Disease Control and Prevention
.
Antifungal susceptibility testing and interpretation
.
https://www.cdc.gov/fungal/candida-auris/c-auris-antifungal.html. Published 2018. Accessed February 4, 2019
.

Author notes

The authors have no relevant financial interest in the products or companies described in this article.

Competing Interests

The findings and conclusions in this report are those of the authors and do not necessarily represent the official position of the Centers for Disease Control.