Next Article in Journal
Study of Prescription-Indication of Outpatient Systemic Anti-Fungals in a Colombian Population. A Cross-Sectional Study
Next Article in Special Issue
Impact of Antibiotic Prophylaxis on Surgical Site Infections in Cardiac Surgery
Previous Article in Journal
Expression Profiles of Zebrafish (Danio rerio) Lysozymes and Preparation of c-Type Lysozyme with High Bacteriolytic Activity against Vibrio vulnificus
Previous Article in Special Issue
Evolution of the Concept of Sepsis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

How to Identify Invasive Candidemia in ICU—A Narrative Review

1
Infectious Diseases Department, Hospital de Braga, R. Comunidades Lusíadas 133, 4710-357 Braga, Portugal
2
Microbiology Department Laboratory, Fundació Puigvert, C. de Cartagena, 340, 350, 08025 Barcelona, Spain
3
Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
4
Clinical Research in Pneumonia & Sepsis (CRIPS), Vall d’Hebron Institute of Research (VHIR), Pg. de la Vall d’Hebron, 129, 08035 Barcelona, Spain
5
Clinical Research, CHU Nîmes, Rue du Professeur Robert Debré 4, 30900 Nîmes, France
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(12), 1804; https://doi.org/10.3390/antibiotics11121804
Submission received: 1 November 2022 / Revised: 5 December 2022 / Accepted: 9 December 2022 / Published: 12 December 2022

Abstract

:
The incidence of invasive fungal infection in ICUs has increased over time, and Candida spp. is the most common cause. Critical care patients are a particular set of patients with a higher risk of invasive fungal infections; this population is characterized by extensive use of medical devices such as central venous lines, arterial lines, bladder catheters, hemodialysis and mechanical intubation. Blood cultures are the gold standard diagnosis; still, they are not an early diagnostic technique. Mannan, anti-mannan antibody, 1,3-β-D-glucan, Candida albicans germ tube antibody, Vitek 2, PNA-FISH, MALDI-TOF, PCR and T2Candida panel are diagnostic promising microbiological assays. Scoring systems are tools to distinguish patients with low and high risk of infection. They can be combined with diagnostic tests to select patients for pre-emptive treatment or antifungal discontinuation. Candidemia is the focus of this narrative review, an approach to contributing factors and diagnosis, with an emphasis on critical care patients.

1. Introduction

Patients admitted to intensive care units (ICUs) have the highest risk of healthcare-associated infection, 19.2% compared to 5.2% on the 2018 European point-prevalence survey [1]; The incidence of invasive fungal infection in ICUs has increased over time, and Candida is the most common cause [2,3]. The most frequent invasive fungal diseases in ICU are invasive candidiasis and invasive aspergillosis (among other molds). Invasive candidiasis is mostly manifested as candidaemia [4]. Candida spp. is a leading cause of bloodstream infections (BSIs) [5,6,7], and mortality associated with invasive Candida infections remains high. Crude mortality can reach up to 50% [5,6,8,9]. Candidemia prolongs hospital stays and increases the costs associated with patient management [6,9].
Among human pathogenic Candida spp., Candida albicans, Candida glabrata, Candida parapsilosis, Candida tropicalis and Candida krusei account for the majority of infections [10]. C. albicans remains the most common species causing candidemia, yet non-albicans Candida has been rising [10]. This epidemiological change may be partially explained by the use of antifungals. Other risk factors for infection include previous Candida colonization, exposure to broad-spectrum antibiotics, malignancy, surgery or use of intravascular catheters, among others [10,11].
Despite improvements in diagnosis, it remains a challenge in intensive care units. Early diagnosis, source control and timely antifungal therapy are the cornerstone. Scoring systems are tools to distinguish patients at low and high risk of infection in an ICU setting. Scoring systems can be combined with diagnostic tests for optimal utilization. Prompt diagnoses can be made with non-culture diagnostic tools, yet they do not substitute blood cultures. The gold standard for candidaemia diagnosis is Candida identification in blood cultures. There are several diagnostic methods used for the rapid identification of Candida spp. based on biochemical characteristics or molecular amplification, each one with limitations.
A worrying emergence of resistance in Candida spp. in critically ill patients threatens appropriate antifungal therapy [12]. Antifungal stewardship (AFS) is a component of antimicrobial stewardship and has received increasing relevance to optimize the use of antifungal therapy.
Candidemia is the focus of this narrative review, an approach to contributing factors and diagnosis, with an emphasis on critical care patients.

2. Candidemia Risk Factors

Advances in medicine allowed us to decrease mortality and prolong life; still, the growing number of immunocompromised patients and associated risk factors explain the increased frequency of candidemia (Table 1).
Candida spp. are yeasts, and there are more than 150 species, but only a few causes disease in humans. Candida spp. are normal human commensals and can be isolated on the skin, gastrointestinal tract, expectorated sputum or respiratory specimens in intubated patients, female genital tract and in the urine of patients with indwelling catheters. Candida spp. becomes pathogenic after immune system defects, either iatrogenic or idiopathic.
Critical care patients are a particular set of patients with a higher risk of invasive fungal infections; this population is characterized by extensive use of medical devices such as central venous lines, arterial lines, bladder catheters, hemodialysis and mechanical intubation [10,11]. All these devices are gateways for colonization and further infection.

2.1. Colonization and Infection

Prior Candida spp. colonization is an independent risk factor, particularly in patients with multifocal fungal colonization [13]. According to León et al. [13], mortality rate was higher in patients with multifocal colonization, with 50.9% against 26.5% mortality rate in patients with unifocal colonization. Multifocal colonization was defined when Candida spp. were simultaneously isolated from various non-contiguous foci [13]. A previous study did not associate colonization with infection risk [14], yet only rectal and/or urine isolates were collected. Pittet et al. [15] demonstrated a higher risk of fungal infection depending on the intensity of colonization—patients colonized at more than two sites.
Etiology has seen major shifts through time towards non-albicans Candida, and different Candida spp. identification are related to different contributing factors. Candidemia by C. glabrata is described in patients with solid organ transplants and with previous antifungal therapy [10,16,17]. C. parapsilosis is identified in patients with recent surgery, patients with intravascular devices or parental nutrition [10,18]. C. parapsilosis has a particular affinity to intravascular devices due to their adherence ability and biofilm formation [19].
C. tropicalis and C. krusei were mainly isolated in patients with hematologic malignancies [10,16,17]. Patients on dialysis or with HIV infection were prone to Candida dubliniensis infection [10]. Candida guilliermondii cases had prior antifungal exposure [10]. Candida lusitaniae had solid tumor history and recent surgery [10]. Candida auris is an emerging multidrug-resistant yeast in patients with surgery or intravascular devices and with previous antifungal therapy [20] and is prone to cause nosocomial outbreaks [21]. C. albicans infection has a lower mortality risk when compared to non-albicans Candida [12,22].
The emergence of resistance in Candida spp. has raised concern in critically ill patients and threatens appropriate antifungal therapy. C. glabrata is the most commonly resistant identified species [12]. The reduced susceptibility to azoles has modified antifungal prescription practices for echinocandins; as a result, selection pressure increased resistance to echinocandins [23,24,25].

2.2. Malignancy

The base of the immune response to invasive candidiasis is a neutrophil function, and neutropenia is a well-recognized risk factor for invasive fungal infection. Monocytes/macrophages also play an important role in protection. Cancer patients have immune defects and a disruption in intestinal mucosal integrity that allows local Candida spp. overgrowth and access to the bloodstream. Patients with candidemia have a 30 to 50% cancer prevalence [10,26]. There are a number of cancer diagnoses related to candidemia, such as acute leukemia, lymphoma or myelodysplastic syndrome, allogeneic hematopoietic cell transplantation and graft versus host disease [26,27,28].
Among solid tumors, the risk correlates with Candida colonization sites. Patients with gastrointestinal cancer have the majority of cases of invasive fungal infections, followed by genito-urinary cancer [26,27].

2.3. Surgery

Bloodstream infections caused by Candida spp. are mainly caused through the intestinal barrier, particularly relevant in patients after surgery due to mucosal damage.
The incidence of candidaemia is higher on surgical ICUs when compared to medical ICUs [14,22]. Surgery is a major risk factor and is well-proven when involving the gastrointestinal tract [14,26]. This association is explained by Candida spp. colonization in this site. Patients submitted to upper gastrointestinal tract surgery or the presence of gastroesophageal junction leakage is a risk factor for Candida infections [29]. Patients under thoracic surgery also had a risk for candidaemia, while trauma and neurosurgical cases had a lower risk [14].
Surgical patients usually recover with hyperalimentation fluids, a relevant risk for candidemia. Total parental nutrition has been associated with a higher risk for candidaemia than peripheral parental nutrition (OR 26.8 vs. 20.0) [30].

2.4. Catheter-Related Bloodstream Infection

Catheter-related Bloodstream Infection (CRBSI) is defined as occurring 48 h before or after catheter removal and positive culture with the same microorganism of either quantitative CVC culture ≥ 103 CFU/mL or semi-quantitative CVC culture > 15 CFU or BSI occurring with or without catheter removal, and quantitative blood culture ratio CVC blood sample/peripheral blood sample > 5 or differential delay of positivity of blood cultures (CVC blood sample culture positive two hours or more before peripheral blood culture) or positive culture with the same microorganism from pus from insertion site [31].
Skin colonization is the first step for invasive candidiasis. Devices disrupt the physical barrier of the skin and mucous membranes allowing the fungus to access the blood. Cardiovascular invasive procedures and the presence of intravascular catheters are common risk factors [10,32].
Patients admitted to intensive care units have the highest risk of healthcare-associated infection (HAI), 19.2% compared to 5.2% on the 2018 European point-prevalence survey (PPS) [1]. Bloodstream infections were the fourth most frequently reported HAI, 10.8% in PPS [1]. Candida spp. was one of the 10 most frequently isolated microorganisms [7]. In ICU, BSIs are the third most common site of infection and the highest infection-associated mortality [3]. Among patients with positive microbiological cultures, 16% had a fungal microorganism [3].
Nosocomial BSIs often are related to the presence of a catheter; therefore, ICUs have higher rates of catheter-related BSI.

2.5. Sepsis

Septic shock in the setting of candidemia was believed to occur less than bacteremia septic shock, but in a 2016 published EUROBACT study [32], 39.6% of patients admitted with fungemia presented with septic shock against 21.6% of bacteremia patients. Candida septic shock carries a high crude mortality, reported being in the range of 36 to 61% [3,33,34,35]. Risk factors associated with higher mortality in ICU are failure of source control, delay in antifungal therapy, and increasing APACHE score [35]. Patients with vasopressors who underwent renal replacement therapy or positive ventilatory support present an increase in the volume of distribution and are exposed to antifungal underdosing, and therapeutic drug measurement (TDM) is advised.

2.6. Broad-Spectrum Antibiotics

Antibiotics are responsible for changes in endogenous microbial flora, which allows fungal overgrowth on site. The use of broad-spectrum antibiotics is believed to be a risk factor for Candida spp. infection [36], particularly when more than two drugs are used [11]. In a recent retrospective study on catheter-related C. parapsilosis BSI, patients with prior use of more than three antibiotics had seven times greater risk of candidemia [37]. Quinolones and third generation cephalosporins were the mainly used antibiotics associated with Candida BSI in intensive care units [38].
Antibiotic consumption has decreased between 2019 and 2020 (consequences of the SARS-CoV-2 pandemic), yet antibiotic consumption is higher in critical care compared to infirmary patients. On the other hand, antifungal consumption has increased, possibly due to an increase in fungal co-infection in patients with COVID-19 and corticosteroid use [39]. Previous use of antimicrobial therapy must be evaluated in all patients with suspected Candida spp. infection.

3. Diagnostic Approach

Invasive candidiasis (IC) includes candidaemia and deep-seated tissue candidiasis. Candidaemia is the most common identity in ICU versus deep-seated candidiasis [4]. Diagnosis of candidaemia is challenging due to its similarity with other infections. The selection of patients with Candida isolates (colonization) who have the risk of invasive infection is a decisive step. Specimens often can be colonized with Candida species in respiratory samples (sometimes representing oral flora contamination), urine (genital contamination), surgical drainages (external skin contamination) or indwelling catheters, and they must not be interpreted as true infection. Diagnostic misinterpretations can lead to unnecessary antifungal use, antifungal resistance, ignoring the true pathogenic organisms and increased healthcare costs. Candidaemia is defined by the presence of Candida species in the blood. Diagnosis is established by blood culture, ideally three different sets of two bottles with 10 mL in each bottle (total of 60 mL) [40].
Signs of invasive candidiasis might be scarce, and the diagnosis is usually late in the course of the ICU stay. The median time between the onset of infection and antifungal therapy can be up to eight days [34]. Early diagnosis is challenging, and a high index of suspicion is the baseline approach to the patient with risk factors for invasive fungal infection.
A probable diagnosis of IC requires the presence of a risk factor, clinical criteria and mycological evidence [41]. Mycological criteria are based on cytology, direct microscopy or culture in a sterile site, or detection of B-1,3-D-glucan (BDG) detected in at least two consecutive serum samples or a Candida spp. identification with the T2Candida panel [41,42]. Possible invasive candidiasis is defined as a patient with a risk factor and clinical syndrome without mycological criteria [41].
Bassetti et al. [4] proposed a definition of probable IC for critically ill patients where some host factors were adjusted to ICU: impaired gut wall integrity (patients with recent abdominal surgery, recurrent intestinal perforations), impaired cutaneous barriers to bloodstream infection (presence of central vascular access device, ECMO cannula or hemodialysis) and Candida colonization (recovery of Candida spp. in cultures from two or more sites).

3.1. Culture

The standard of care for definite diagnosis is the isolation of Candida in blood cultures (BC), sill it is not an early diagnostic technique. Sensitivity of BC to detect Candida ranges from 50 to 71%. Still, it can be lower in neutropenic patients [43,44]. Candida isolation can take between two and three (some until eight) days to grow [45]. Time to positivity is different between species. C. glabrata grows slower than C. albicans [46]. Blood cultures can be negative in patients with antifungal drug exposure, and sensitivity can be increased when the volume of the complete set of blood cultures is 60 mL. In patients with probable invasive candidiasis, the recommended frequency of blood culture collection is daily [44]. Non-culture diagnostic tools do not substitute blood cultures. They can only be combined for earlier intervention. After organism identification in BC, an antifungal susceptibility test is required to guide the management of candidaemia and oral azole de-escalation due to the emergence of resistance to azoles and echinocandins, significant in C. glabrata.

3.2. Serum Biomarkers

Biomarkers are essential tools for early diagnosis; still, despite extensive research, they are not validated to distinguish colonized patients from patients with fungal infections. Available biomarkers, such as mannan, anti-mannan antibody and 1,3-β-D-glucan (BDG), have been developed to improve and anticipate the detection of invasive disease prior to microbiological confirmation.
Mannans are a main cell wall component of Candida spp. and are used to detect Candida infections [47]. The combined detection of mannan and anti-mannan antibody increases the sensitivity from 58% to 83% and specificity from 59% to 86% [48,49]. In the ICU setting, they have a high negative predictive value, which is particularly useful in excluding invasive Candida infections, especially after five days of unnecessary antifungal therapy [33]. These biomarkers can predict infection prior to blood cultures. They are an important tool in reducing the diagnosis time yield or reducing the use of antifungal agents. For non-albicans bloodstream infections, such as C. parapsilosis and C. krusei, antigen and antibody detection have lower sensitivity [50].
BDG is a pan-fungal diagnostic test. BDG is a cell wall component of Candida and other fungi (such as Aspergillus, Pneumocystis jiroveci and others) with a high diagnostic sensitivity of 75–80% and specificity of around 80% [49] Odabasi et al. [51] reported positive BDG result up to 10 days before clinical diagnosis in patients with proven or probable invasive fungal infection.
To optimize BDG performance, two consecutive positive results are required. A meta-analysis to evaluate the accuracy of BDG on ICU patients by Haydour et al. [52] reported 80% of sensitivity but low specificity (only 60%). Patients with albumin, renal replacement therapies with cellulose membranes, intravenous immunoglobulin and concomitant BSI, may have false positive BDG [43]. BDG can be used to withdraw unneeded antifungals due to high negative predictive value (NPV) in ICU [53,54].
Antibodies against C. albicans germ tubes (CAGTA) is an immunofluorescence assay that detects responses against a hyphal protein expressed during tissue invasion and biofilm formation [49]. Sensitivity and specificity are variable and higher in candidemia [55,56,57]. Still, false positives can occur in the presence of concomitant bacterial infection [58]. Serology tests usually take time to be positive, normally more than 15 days after the first encounter of the microorganism antigen with the host. Some researchers have started to study the kinetics of these antibodies [59]. Nevertheless, more knowledge about the time to positivity after the initiation of candidemia of this promising test is needed to decide if it is useful at the beginning of the clinical episode or only after some days. It might be more useful to rule out candidemia in already treated patients and withdraw unnecessary antifungal empirical treatment [33].
A positive biomarker report does not provide a diagnosis yet raises the probability of infection. Simultaneous use of different biomarkers can improve the negative predictive value or avoid false positive results. The combination of CAGTA and BDG improved the NPV to 95 to 97% for proven invasive candidiasis in some ICUs [48,60].
Biomarkers can be expensive and time-consuming. They should probably not be used widely for the potential risk of extra antifungal consumption. Additionally, the use of a biomarker-based strategy in the ICU demonstrated increased early discontinuation of empirical antifungal therapy without a negative impact on outcomes [48,61].

3.3. Molecular Biology

Polymerase Chain Reaction (PCR) performed in blood samples have the highest sensitivity, 90–95%, and specificity, 90–92%. PCR shortens the time to a diagnosis, yet the interpretation is heterogenic, and colonized patients may have a positive PCR [55,62]. The need to follow a strict aseptic technique to obtain the blood for this test is as important, if not more, than for the routine blood cultures because PCR detects very small quantities of the genetic material of either viable or non-viable microorganisms.
The detection limit of PCR is under 10 CFU/mL; still, if the number of Candida CFU/mL in the blood is under the threshold, like in an early set of the disease, the test might be negative. Pfeiffer et al. [45] reported CFU/mL of ≤1 on half of Candida spp. blood cultures, particularly in patients with candidemia by C. glabrata. Low organism burden was associated with neutropenic patients, recent major surgery, end-stage live disease, renal replacement therapy, interrupted gastrointestinal tract and candidemia from the abdominal site [45]. The five most common pathogenic Candida spp., such as C. albicans, C. glabrata, C. parapsilosis, C. tropicalis and C. krusei, can be targeted by commercial multiplex PCR kits. Specificity is superior in molecular amplification techniques over BDG and CAGTA, and, as a consequence, better positive predictive values can be achieved [55].
The BioFireFilmArray BCID assay identifies 24 organisms (19 bacteria and 5 most common Candida species) by multiplex PCR from positive blood cultures [63], with a sensitivity of 100% and results in one hour [64]. An updated version of the panel—BCID2 identifies 33 species, including 7 fungi, 6 Candida spp. (C. albicans, C. glabrata, C. parapsilosis, C. tropicalis, C. krusei and C. auris) [65].
A novel nanodiagnostic test, T2Candida Panel, is a PCR-based assay that detects Candida within whole blood through mechanical lyses of cells and DNA amplification later detected by amplicon-induced agglomeration of super magnetic particles and T2 magnetic resonance measurement. Sensitivity and specificity are 89–91% and 98–99% [66,67]. The limit of detection is 1 CFU/mL [67], and the limit of blood cultures is 1 CFU/60 mL of blood, usually obtained in a routine set of three 20 mL samples. An important feature is the availability of the result in 3–4 h directedly from whole blood. The T2Candida Panel reports the five most common Candida spp. as a positive or negative result. Results are reported based on susceptibility to fluconazole and divided as C. albicans/C. tropicalis, C. parapsilosis, and C. krusei/C. glabrata. An important benefit of this novel test is the higher sensitivity on follow-up analysis versus blood cultures, demonstrated in already Candida BSI patients either with neutropenia or in patients receiving prior antifungal therapy [68], probably because it is a genetic amplification assay that detects non-viable yeasts. Positive blood cultures and positive T2Candida in candidemia follow-up samples had higher mortality (42% vs. 5% when they were negative) [68].
All these molecular tests do not discriminate colonization or past infection from ongoing true infection, and diagnostic stewardship is advised to avoid over-diagnosis of true active fungal infections.

3.4. Other Methods

Conventional methods of identification are based on morphological and biochemical Candida characteristics. They have low identification accuracy and are time-consuming [69]. Biochemical methods based on automated systems improve etiology diagnosis. VITEK 2 is an automated identification system capable of antifungal susceptibility testing after positive blood cultures. VITEK 2 has sensitivity and specificity above 95% for common Candida spp. [70]. Misidentification of uncommon Candida spp. [69,71,72] were reported, particularly C. guilliermondii. Peptide Nucleic Acid in Situ Hybridisation Yeast Traffic Light system (PNA-FISH YTL) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) rapidly identifies Candida spp., still only after positive blood cultures.
Although high sensitivity (96 and 99%), PNA-FISH YTL cannot distinguish between C. albicans and C. parapsilosis or between C. glabrata and C. krusei [73,74]. MALDI-TOF MS has a 56 to 73% sensitivity and differentiates distinct and related species [75,76]. C. auris, often misdiagnosed, can be identified on MALDI-TOF MS [77]. MALDI-TOF MS requires pure growth of the organism on artificial media. After the isolation of the organism, MALDI-TOF takes 10 to 15 min to identify. MALDI-TOF MS can be used for antifungal susceptibility tests. This assay reports fluconazole/caspofungin/anidulafungin resistance for C. albicans and C. glabrata [76].
The Accelerate PhenoTestTM BC Kit detects C. albicans and C. glabrata (in addition to identification and rapid antimicrobial susceptibility testing of gram-positive and gram-negative bacteria in positive blood cultures). This assay had 100% and 99% sensitivity and specificity for C. albicans, while C. glabrata had 80% and 97%, respectively [78].

4. Scoring Systems

Since the delay in antifungal therapy is associated with increased mortality in patients with candidemia [79], the selection of the right patient at risk of invasive fungal infection is essential. Antifungal therapy should be considered in intra-abdominal postoperative ICU patients with risk factors for invasive Candida infection who present fever of unknown cause (or persistent clinical signs of sepsis) and positive Candida serum biomarkers [80]. Early pre-emptive or prophylaxis therapy has been suggested based on scoring systems to select high-risk patients and through the existing diagnostic techniques (Figure 1). Clinical scores are essential tools to distinguish patients at low and high risk of infection and may reduce costs and unnecessary use of antifungals.
Since colonization status is an independent risk factor and multifocal colonization predictor of a higher risk of invasive Candida infection, a colonization index was suggested first by Pittet et al. [15] and then evaluated in a prospective cohort [35] with surgical patients admitted to ICU. Patients with a corrected colonization index (CCI) above 0.4 received preemptive antifungal therapy, and a significant decrease in the acquisition of proven candidiasis was demonstrated. No patients with a CCI under 0.4 developed proven candidiasis.
Ostrosky-Zeichner et al. [81] described three predictive rules (one major criterium and two minor criteria) for candidaemia in an intensive care setting with any antibiotic use, presence of CVC, any surgery, immunosuppressive therapy, pancreatitis, total parental nutrition, dialysis and steroid use. Patients selected by rule 3 had a higher rate of infection, yet only one-third of patients with candidaemia were identified. Another study reported higher sensitivity [82].
Candida Score [13] is a simple bedside scoring system. It was developed based on previous colonization status (multifocal) and clinical predisposition: surgery, total parental nutrition and severe sepsis. This score is validated for critically ill patients admitted to an ICU and had 81% sensitivity and 74% specificity. A score above 2.5 in an intra-abdominal postoperative ICU patient was suggested for consideration of starting pre-emptive antifungal agents, and under 2.5 strongly decreases the chance of infection.
Another scoring system was evaluated in intra-abdominal surgical patients: Dupont et al. [83] demonstrated an 84% sensitivity and 50% specificity with a score based on the female gender, the upper gastrointestinal origin of peritonitis, cardiovascular failure and use of antibiotics. Basseti et al. [84] proposed an algorithm considering abdominal surgical patients versus non-abdominal surgery/medical, colonization status and Candida score and biomarkers to address the need for antifungals; the efficacy and mortality should be validated.
These scores are targeted to abdominal surgical patients admitted to ICU; these scores are not validated for medical patients admitted to ICU or patients with malignancy. The optimal utilization of risk factors or serum biomarkers or score systems is yet to be known. Its efficacy and influence on mortality call for randomized controlled trials.

5. Antifungal Stewardship

Accompanying the rise of invasive fungal infections, there is a worldwide rise of resistance of Candida to azoles and echinocandins [24]. The resistance to azoles has increased the use of echinocandins to treat Candida infections; as a consequence, exposure to echinocandins has led to reduced susceptibility [24]. The emergence of fungal resistance has an impact on patient outcomes. There is a call for active stewardship to ensure responsible use and minimize the development of resistance.
Antimicrobial stewardship refers to “a coherent set of actions which promote using antimicrobials responsibly” [85], which includes optimizing antibiotics, antivirals and antifungals. Antifungal stewardship improves antifungal use, patient care and outcomes [85,86,87,88,89]; AFS interventions reduce the time to antifungal prescription [87]. AFS should be included as part of the AMS program, depending on the ICU setting. In an ICU where the patient population already has a degree of complexity, when fungal infections are frequent and the use of antifungal agents is increasing, AFS is required.
Diagnostic stewardship is a fundamental step in stewardship programs. There are three important issues in diagnosis stewardship improving time to diagnosis, appropriate antifungal based on timely antifungal susceptibility tests and antifungal discontinuation in selected patients in intensive care units with negative biomarkers or negative PCR.
A recent core intervention for Antifungal Stewardship was published [90] with the recommendation to use both fungal culture and non-culture-based tests. Biomarkers and molecular amplification techniques reduce the time yield for diagnosis with the highest sensitivity [48,61,90]. However, they do not represent confirmation of an active infection, and the interpretation is not equivalent to the antifungal requirement.
Empirical antifungal therapy based only on clinical signs and risk factors leads to the unnecessary use of antifungals [87]. Antifungal discontinuation can be based on the high negative predictive value that can be achieved by using non-culture-based tests but nevertheless still requires an individualized patient approach.
Rautemaa-Richardson et al. [91] evaluated the use of BDG testing on antifungal discontinuation. This strategy reduced echinocandin consumption by 39%. Ito-Takeichi et al. [92] implemented an antifungal stewardship intervention based on BDG and reported a significant reduction of antifungals and better outcomes in patients with proven candidaemia.
Appropriate prescription of antifungal drugs depends on timely Candida isolation and susceptibility testing; AFS strategies reduce the time to start antifungal therapy [87]. There are several different diagnostic methods used for the rapid identification of Candida spp. MALDI-TOF MS, PNA-FISH and Multiplex PCR provide results within minutes to a few hours, only they are blood-culture-based methods. The T2Candida panel is not a blood-culture-based test, reducing the time yield for diagnosis [67,93,94]. Gill et al. [94] evaluated the discontinuation of antifungals, and only 3% had the development of proven candidemia after discontinuation.
Among different AFS interventions to improve patient outcomes with candidaemia, diagnostic stewardship for prompt diagnosis is fundamental for the appropriate and cost-effective treatment of patients at risk of invasive candidiasis. Antifungal therapy for prophylaxis of cannula or indwelling catheter insertion is not recommended.

6. Conclusions

Critical care patients are a particular set of patients with a higher risk of invasive fungal infections. This population is characterized by extensive use of medical devices such as central venous lines, arterial lines, bladder catheters, ECMO and renal replacement therapy cannulas and tracheal intubation. Septic shock in the setting of candidemia is a reality in critically ill patients with high crude mortality. Available biomarkers, such as mannan, anti-mannan antibody, 1,3-β-D-glucan and Candida albicans germ tube antibody, have been developed to improve and anticipate the detection of invasive disease or withdraw unnecessary empirical antifungal treatment. The use of a biomarker-based strategy in the ICU demonstrated increased early discontinuation of empirical antifungal therapy without a negative impact on outcomes. Still, biomarkers tests should probably not be used widely for the potential risk of extra antifungal consumption. Recent advances in molecular biology shorten diagnostic time yield with high sensitivity and specificity. There are a number of commercial multiplex PCR tests to target the five most common pathogenic Candida species after positive blood cultures and a novel nanodiagnostic panel, the T2Candida panel, that detects Candida directly from a whole blood sample. Scoring systems are tools to distinguish patients at low and high risk of infection; these scores are targeted to abdominal surgical patients admitted to ICU. Scoring systems can be combined with diagnostic tests to select patients for pre-emptive treatment. Still, the optimal utilization of risk factors and score tools or serum biomarkers is yet to be known. Its efficacy and influence on mortality call for randomized controlled trials.
Antifungal stewardship is fundamental to optimize antifungal therapy and, consequently, patient care and outcomes improvement; diagnostic stewardship is the core strategy to reduce time yield to diagnosis and timely antifungal susceptibility test. Antifungal discontinuation based on non-culture-based tests is reserved for the right clinical setting, such as the intensive care unit.
In summary, appropriate antifungal therapy is a determinant of survival in critically ill patients with susceptible Candida infections. Safe implementation requires a smart strategy to avoid both delays in starting antifungal therapy and avoid over-prescription (for colonization or contaminated specimens). Renal function, the daily dose administered, and the site of infection are determinants of the right prescription. Optimized drug dosing and diagnosis should be considered core priorities for improving clinical outcomes for critically ill patients with fungal infections.

Author Contributions

The first author (J.A.) undertook the literature search and first draft manuscript preparation and is the guarantor for this article. J.R. and C.A.-T. were responsible for manuscript revision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

J.R. served in advisory boards/speaker bureau for Pfizer and MSD. CAT is a member of Sysmex advisory Board on Urinalysis. J.A. does not disclose conflicts of interest.

References

  1. Suetens, C.; Latour, K.; Kärki, T.; Ricchizzi, E.; Kinross, P.; Moro, M.L.; Jans, B.; Hopkins, S.; Hansen, S.; Lyytikäinen, O.; et al. Prevalence of healthcare-associated infections, estimated incidence and composite antimicrobial resistance index in acute care hospitals and long-term care facilities: Results from two European point prevalence surveys, 2016 to 2017. Eurosurveillance 2018, 23, 1800516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Vincent, J.L.; Rello, J.; Marshall, J.; Silva, E.; Anzueto, A.; Martin, C.D.; Moreno, R.; Lipman, J.; Gomersall, C.; Sakr, Y.; et al. International Study of the Prevalence and Outcomes of Infection in Intensive Care Units. JAMA 2009, 302, 2323–2329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Vincent, J.-L.; Sakr, Y.; Singer, M.; Martin-Loeches, I.; Machado, F.R.; Marshall, J.C.; Finfer, S.; Pelosi, P.; Brazzi, L.; Aditianingsih, D.; et al. Prevalence and Outcomes of Infection Among Patients in Intensive Care Units in 2017. JAMA 2020, 323, 1478–1487. [Google Scholar] [CrossRef] [PubMed]
  4. Bassetti, M.; Azoulay, E.; Kullberg, B.-J.; Ruhnke, M.; Shoham, S.; Vazquez, J.; Giacobbe, D.R.; Calandra, T. EORTC/MSGERC Definitions of Invasive Fungal Diseases: Summary of Activities of the Intensive Care Unit Working Group. Clin. Infect. Dis. 2021, 72, S121–S127. [Google Scholar] [CrossRef] [PubMed]
  5. Wisplinghoff, H.; Bischoff, T.; Tallent, S.M.; Seifert, H.; Wenzel, R.P.; Edmond, M.B. Nosocomial Bloodstream Infections in US Hospitals: Analysis of 24,179 Cases from a Prospective Nationwide Surveillance Study. Clin. Infect. Dis. 2004, 39, 309–317. [Google Scholar] [CrossRef] [Green Version]
  6. Gudlaugsson, O.; Gillespie, S.; Lee, K.; Vande Berg, J.; Hu, J.; Messer, S.; Herwaldt, L.; Pfaller, M.; Diekema, D. Attributable Mortality of Nosocomial Candidemia, Revisited. Clin. Infect. Dis. 2003, 37, 1172–1177. [Google Scholar] [CrossRef]
  7. Rosenthal, V.D.; Bat-Erdene, I.; Gupta, D.; Belkebir, S.; Rajhans, P.; Zand, F.; Myatra, S.N.; Afeef, M.; Tanzi, V.L.; Muralidharan, S.; et al. Six-year multicenter study on short-term peripheral venous catheters-related bloodstream infection rates in 727 intensive care units of 268 hospitals in 141 cities of 42 countries of Africa, the Americas, Eastern Mediterranean, Europe, South East Asia, and Western Pacific Regions: International Nosocomial Infection Control Consortium (INICC) findings. Infect. Control Hosp. Epidemiol. 2020, 41, 553–563. [Google Scholar] [CrossRef]
  8. Falagas, M.E.; Apostolou, K.E.; Pappas, V.D. Attributable mortality of candidemia: A systematic review of matched cohort and case-control studies. Eur. J. Clin. Microbiol. Infect. Dis. 2006, 25, 419–425. [Google Scholar] [CrossRef]
  9. Cornely, F.B.; Cornely, O.A.; Salmanton-García, J.; Koehler, F.C.; Koehler, P.; Seifert, H.; Wingen-Heimann, S.; Mellinghoff, S.C. Attributable mortality of candidemia after introduction of echinocandins. Mycoses 2020, 63, 1373–1381. [Google Scholar] [CrossRef]
  10. Pfaller, M.; Neofytos, D.; Diekema, D.; Azie, N.; Meier-Kriesche, H.-U.; Quan, S.-P.; Horn, D. Epidemiology and outcomes of candidemia in 3648 patients: Data from the Prospective Antifungal Therapy (PATH Alliance®) registry, 2004–2008. Diagn. Microbiol. Infect. Dis. 2012, 74, 323–331. [Google Scholar] [CrossRef]
  11. Wang, H.; Liu, N.; Yin, M.; Han, H.; Yue, J.; Zhang, F.; Shan, T.; Guo, H.; Wu, D. The epidemiology, antifungal use and risk factors of death in elderly patients with candidemia: A multicentre retrospective study. BMC Infect. Dis. 2014, 14, 609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Pristov, K.E.; Ghannoum, M.A. Resistance of Candida to azoles and echinocandins worldwide. Clin. Microbiol. Infect. 2019, 25, 792–798. [Google Scholar] [CrossRef]
  13. León, C.; Ruiz-Santana, S.; Saavedra, P.; Almirante, B.; Nolla-Salas, J.; Álvarez-Lerma, F.; Garnacho-Montero, J.; León, M.A.; EPCAN Study Group. A bedside scoring system (“Candida score”) for early antifungal treatment in nonneutropenic critically ill patients with Candida colonization. Crit. Care Med. 2006, 34, 730–737. [Google Scholar] [CrossRef] [PubMed]
  14. Blumberg, H.M.; Jarvis, W.R.; Soucie, J.M.; Edwards, J.E.; Patterson, J.E.; Pfaller, M.A.; Rangel-Frausto, M.S.; Rinaldi, M.G.; Saiman, L.; Wiblin, R.; et al. Risk Factors for Candidal Bloodstream Infections in Surgical Intensive Care Unit Patients: The NEMIS Prospective Multicenter Study. Clin. Infect. Dis. 2001, 33, 177–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Pittet, D.; Monod, M.; Suter, P.M.; Frenk, E.; Auckenthaler, R. Candida Colonization and Subsequent Infections in Critically III Surgical Patients. Ann. Surg. 1994, 220, 751–758. [Google Scholar] [CrossRef]
  16. Lortholary, O.; Desnos-Ollivier, M.; Sitbon, K.; Fontanet, A.; Bretagne, S.; Dromer, F. Recent Exposure to Caspofungin or Fluconazole Influences the Epidemiology of Candidemia: A Prospective Multicenter Study Involving 2441 Patients. Antimicrob. Agents Chemother. 2011, 55, 532–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hachem, R.; Hanna, H.; Kontoyiannis, D.P.; Jiang, Y.; Raad, I. The changing epidemiology of invasive candidiasis. Cancer 2008, 112, 2493–2499. [Google Scholar] [CrossRef]
  18. Almirante, B.; Rodríguez, D.; Cuenca-Estrella, M.; Almela, M.; Sanchez, F.; Ayats, J.; Alonso-Tarres, C.; Rodriguez-Tudela, J.L.; Pahissa, A. Epidemiology, Risk Factors, and Prognosis of Candida parapsilosis Bloodstream Infections: Case-Control Population-Based Surveillance Study of Patients in Barcelona, Spain, from 2002 to 2003. J. Clin. Microbiol. 2006, 44, 1681–1685. [Google Scholar] [CrossRef] [Green Version]
  19. Trofa, D.; Gácser, A.; Nosanchuk, J.D. Candida parapsilosis, an Emerging Fungal Pathogen. Clin. Microbiol. Rev. 2008, 21, 606–625. [Google Scholar] [CrossRef] [Green Version]
  20. Lockhart, S.R.; Etienne, K.A.; Vallabhaneni, S.; Farooqi, J.; Chowdhary, A.; Govender, N.P.; Colombo, A.L.; Calvo, B.; Cuomo, C.A.; Desjardins, C.A.; 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, 134–140. [Google Scholar] [CrossRef]
  21. Bougnoux, M.-E.; Kac, G.; Aegerter, P.; D’Enfert, C.; Fagon, J.-Y. CandiRea Study Group Candidemia and candiduria in critically ill patients admitted to intensive care units in France: Incidence, molecular diversity, management and outcome. Intensiv. Care Med. 2007, 34, 292–299. [Google Scholar] [CrossRef] [PubMed]
  22. Gangneux, J.-P.; Cornet, M.; Bailly, S.; Fradin, C.; Féger, C.; Timsit, J.-F.; Leroy, O.; Sendid, B.; Bougnoux, M.-E. Clinical Impact of Antifungal Susceptibility, Biofilm Formation and Mannoside Expression of Candida Yeasts on the Outcome of Invasive Candidiasis in ICU: An Ancillary Study on the Prospective AmarCAND2 Cohort. Front. Microbiol. 2018, 9, 2907. [Google Scholar] [CrossRef] [PubMed]
  23. Walker, L.A.; Gow, N.; Munro, C.A. Fungal echinocandin resistance. Fungal Genet. Biol. 2010, 47, 117–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Frías-De-León, M.G.; Hernández-Castro, R.; Conde-Cuevas, E.; García-Coronel, I.H.; Vázquez-Aceituno, V.A.; Soriano-Ursúa, M.A. Candida glabrata Antifungal Resistance and Virulence Factors, a Perfect Pathogenic Combination. Pharmaceutics 2021, 13, 1529. [Google Scholar] [CrossRef]
  25. Lortholary, O.; The French Mycoses Study Group; Renaudat, C.; Sitbon, K.; Desnos-Ollivier, M.; Bretagne, S.; Dromer, F. The risk and clinical outcome of candidemia depending on underlying malignancy. Intensiv. Care Med. 2017, 43, 652–662. [Google Scholar] [CrossRef] [Green Version]
  26. Cornely, O.A.; Gachot, B.; Akan, H.; Bassetti, M.; Uzun, O.; Kibbler, C.; Marchetti, O.; de Burghgraeve, P.; Ramadan, S.; Pylkkanen, L.; et al. Epidemiology and Outcome of Fungemia in a Cancer Cohort of the Infectious Diseases Group (IDG) of the European Organization for Research and Treatment of Cancer (EORTC 65031). Clin. Infect. Dis. 2015, 61, 324–331. [Google Scholar] [CrossRef]
  27. Gamaletsou, M.N.; Walsh, T.J.; Zaoutis, T.; Pagoni, M.; Kotsopoulou, M.; Voulgarelis, M.; Panayiotidis, P.; Vassilakopoulos, T.; Angelopoulou, M.K.; Marangos, M.; et al. A prospective, cohort, multicentre study of candidaemia in hospitalized adult patients with haematological malignancies. Clin. Microbiol. Infect. 2014, 20, O50–O57. [Google Scholar] [CrossRef] [Green Version]
  28. Brotfain, E.; Sebbag, G.; Friger, M.; Kirshtein, B.; Borer, A.; Koyfman, L.; Frank, D.; Bichovsky, Y.; Peiser, J.G.; Klein, M. Invasive Candida Infection after Upper Gastrointestinal Tract Surgery for Gastric Cancer. Int. J. Surg. Oncol. 2017, 2017, 1–7. [Google Scholar] [CrossRef] [Green Version]
  29. Luzzati, R.; Cavinato, S.; Giangreco, M.; Granà, G.; Centonze, S.; Deiana, M.L.; Biolo, G.; Barbone, F. Peripheral and total parenteral nutrition as the strongest risk factors for nosocomial candidemia in elderly patients: A matched case-control study. Mycoses 2013, 56, 664–671. [Google Scholar] [CrossRef]
  30. European Centre for Disease Prevention and Control. Point Prevalence Survey of Healthcare-Associated Infections and Antimicrobial Use in European Acute Care Hospitals; Protocol Version 6.1; European Centre for Disease Prevention and Control: Stockholm, Sweden, 2022. [Google Scholar]
  31. Puig-Asensio, M.; Padilla, B.; Garnacho-Montero, J.; Zaragoza, O.; Aguado, J.M.; Zaragoza, R.; Montejo, M.; Muñoz, P.; Ruiz-Camps, I.; Cuenca-Estrella, M.; et al. Epidemiology and predictive factors for early and late mortality in Candida bloodstream infections: A population-based surveillance in Spain. Clin. Infect. Dis. 2014, 20, O245–O254. [Google Scholar] [CrossRef]
  32. Paiva, J.-A.; Pereira, J.M.; Tabah, A.; Mikstacki, A.; de Carvalho, F.B.; Koulenti, D.; Ruckly, S.; Çakar, N.; Misset, B.; Dimopoulos, G.; et al. Characteristics and risk factors for 28-day mortality of hospital acquired fungemias in ICUs: Data from the EUROBACT study. Crit. Care 2016, 20, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bassetti, M.; Righi, E.; Ansaldi, F.; Merelli, M.; Cecilia, T.; de Pascale, G.; Diaz-Martin, A.; Luzzati, R.; Rosin, C.; Lagunes, L.; et al. A multicenter study of septic shock due to candidemia: Outcomes and predictors of mortality. Intensive Care Med. 2014, 40, 839–845. [Google Scholar] [CrossRef] [PubMed]
  34. Kollef, M.H.; Micek, S.T.; Hampton, N.; Doherty, A.J.; Kumar, A. Septic Shock Attributed to Candida Infection: Importance of Empiric Therapy and Source Control. Clin. Infect. Dis. 2012, 54, 1739–1746. [Google Scholar] [CrossRef] [PubMed]
  35. Ostrosky-Zeichner, L.; Sable, C.; Sobel, J.; Alexander, B.D.; Donowitz, G.; Kan, V.; Kauffman, C.A.; Kett, D.; Larsen, R.A.; Morrison, V.; et al. Multicenter retrospective development and validation of a clinical prediction rule for nosocomial invasive candidiasis in the intensive care setting. Eur. J. Clin. Microbiol. Infect. Dis. 2007, 26, 271–276. [Google Scholar] [CrossRef]
  36. Yamin, D.H.; Husin, A.; Harun, A. Risk Factors of Candida parapsilosis Catheter-Related Bloodstream Infection. Front. Public Health 2021, 9, 631865. [Google Scholar] [CrossRef]
  37. Peng, S.; Lu, Y. Clinical epidemiology of central venous catheter–related bloodstream infections in an intensive care unit in China. J. Crit. Care 2013, 28, 277–283. [Google Scholar] [CrossRef]
  38. Peng, J.; Wang, Q.; Mei, H.; Zheng, H.; Liang, G.; She, X.; Liu, W. Fungal co-infection in COVID-19 patients: Evidence from a systematic review and meta-analysis. Aging 2021, 13, 7745–7757. [Google Scholar] [CrossRef]
  39. Leber, A.L. (Ed.) Clinical Microbiology Procedures Handbook; ASM Press: Washington, DC, USA, 2016. [Google Scholar]
  40. De Pauw, B.; Walsh, T.J.; Donnelly, J.P.; Stevens, D.A.; Edwards, J.E.; Calandra, T. Faculty Opinions recommendation of Revised definitions of invasive fungal disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin. Infect. Dis. 2008, 46, 1813–1821. [Google Scholar] [CrossRef] [Green Version]
  41. Donnelly, J.P.; Chen, S.C.; Kauffman, C.A.; Steinbach, W.J.; Baddley, J.W.; Verweij, P.E.; Clancy, C.J.; Wingard, J.R.; Lockhart, S.R.; Groll, A.H.; et al. Revision and Update of the Consensus Definitions of Invasive Fungal Disease From the European Organization for Research and Treatment of Cancer and the Mycoses Study Group Education and Research Consortium. Clin. Infect. Dis. 2020, 71, 1367–1376. [Google Scholar] [CrossRef] [Green Version]
  42. Clancy, C.J.; Nguyen, M.H. Finding the “Missing 50%” of Invasive Candidiasis: How Nonculture Diagnostics Will Improve Understanding of Disease Spectrum and Transform Patient Care. Clin. Infect. Dis. 2013, 56, 1284–1292. [Google Scholar] [CrossRef]
  43. Cuenca-Estrella, M.; Verweij, P.e.; Arendrup, M.C.; Arikan-Akdagli, S.; Bille, J.; Donnelly, J.P.; Jensen, H.E.; Lass-Flörl, C.; Richardson, M.d.; Akova, M.; et al. ESCMID* guideline for the diagnosis and management of Candida diseases 2012: Diagnostic procedures. Clin. Microbiol. Infect. 2012, 18, 9–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Pfeiffer, C.D.; Samsa, G.P.; Schell, W.A.; Reller, L.B.; Perfect, J.R.; Alexander, B.D. Quantitation of Candida CFU in Initial Positive Blood Cultures. J. Clin. Microbiol. 2011, 49, 2879–2883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. McCarty, T.P.; White, C.M.; Pappas, P.G. Candidemia and Invasive Candidiasis. Infect. Dis. Clin. N. Am. 2021, 35, 389–413. [Google Scholar] [CrossRef] [PubMed]
  46. Chumpitazi, B.F.F.; Lebeau, B.; Faure-Cognet, O.; Hamidfar-Roy, R.; Timsit, J.-F.; Pavese, P.; Thiebaut-Bertrand, A.; Quesada, J.-L.; Pelloux, H.; Pinel, C. Characteristic and clinical relevance of Candida mannan test in the diagnosis of probable invasive candidiasis. Med. Mycol. 2014, 52, 462–471. [Google Scholar] [CrossRef] [Green Version]
  47. Martínez-Jiménez, M.C.; Muñoz, P.; Valerio, M.; Vena, A.; Guinea, J.; Bouza, E. Combination of Candida biomarkers in patients receiving empirical antifungal therapy in a Spanish tertiary hospital: A potential role in reducing the duration of treatment. J. Antimicrob. Chemother. 2016, 71, 2679. [Google Scholar] [CrossRef] [Green Version]
  48. Clancy, C.J.; Nguyen, M.H. Non-Culture Diagnostics for Invasive Candidiasis: Promise and Unintended Consequences. J. Fungi 2018, 4, 27. [Google Scholar] [CrossRef] [Green Version]
  49. Mikulska, M.; Calandra, T.; Sanguinetti, M.; Poulain, D.; Viscoli, C.; The Third European Conference on Infections in Leukemia Group. The use of mannan antigen and anti-mannan antibodies in the diagnosis of invasive candidiasis: Recommendations from the Third European Conference on Infections in Leukemia. Crit. Care 2010, 14, R222. [Google Scholar] [CrossRef] [Green Version]
  50. Odabasi, Z.; Mattiuzzi, G.N.; Estey, E.; Kantarjian, H.M.; Saeki, F.; Ridge, R.J.; Ketchum, P.A.; Finkelman, M.A.; Rex, J.; Ostrosky-Zeichner, L. β-D-Glucan as a Diagnostic Adjunct for Invasive Fungal Infections: Validation, Cutoff Development, and Performance in Patients with Acute Myelogenous Leukemia and Myelodysplastic Syndrome. Clin. Infect. Dis. 2004, 39, 199–205. [Google Scholar] [CrossRef]
  51. Haydour, Q.; Hage, C.A.; Carmona-Porquera, E.M.; Epelbaum, O.; Evans, S.E.; Gabe, L.M.; Knox, K.S.; Kolls, J.K.; Wengenack, N.L.; Prokop, L.J.; et al. Diagnosis of Fungal Infections. A Systematic Review and Meta-Analysis Supporting American Thoracic Society Practice Guideline. Ann. Am. Thorac. Soc. 2019, 16, 1179–1188. [Google Scholar] [CrossRef]
  52. Prattes, J.; Hoenigl, M.; Rabensteiner, J.; Raggam, R.B.; Prueller, F.; Zollner-Schwetz, I. Serum 1,3-beta-d-glucan for antifungal treatment stratification at the intensive care unit and the influence of surgery. Mycoses 2014, 57, 679–686. [Google Scholar] [CrossRef]
  53. Posteraro, B.; Tumbarello, M.; De Pascale, G.; Liberto, E.; Vallecoccia, M.S.; De Carolis, E.; Di Gravio, V.; Trecarichi, E.M.; Sanguinetti, M.; Antonelli, M. (1,3)-β-d-Glucan-based antifungal treatment in critically ill adults at high risk of candidaemia: An observational study. J. Antimicrob. Chemother. 2016, 71, 2262–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Fortun, J.; Meije, Y.; Buitrago, M.J.; Gago, S.; Bernal-Martinez, L.; Pemán, J.; Perez, M.; Pedrosa, E.G.-G.; Madrid, N.; Pintado, V.; et al. Clinical validation of a multiplex real-time PCR assay for detection of invasive candidiasis in intensive care unit patients. J. Antimicrob. Chemother. 2014, 69, 3134–3141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. León, C.; Ruiz-Santana, S.; Saavedra, P.; Castro, C.; Loza, A.; Zakariya, I.; Úbeda, A.; Parra, M.; Macías, D.; Tomás, J.I.; et al. Contribution of Candida biomarkers and DNA detection for the diagnosis of invasive candidiasis in ICU patients with severe abdominal conditions. Crit Care 2016, 20, 149. [Google Scholar] [CrossRef] [Green Version]
  56. Parra-Sánchez, M.; The CAVA Trem Study Group; Breval, I.Z.-Y.; Méndez, C.C.; García-Rey, S.; Vazquez, A.L.; Iglesias, A.; Guerrero, D.M.; Mejías, A.R.; Gil, C.L.; et al. Candida albicans Germ-Tube Antibody: Evaluation of a New Automatic Assay for Diagnosing Invasive Candidiasis in ICU Patients. Mycopathologia 2017, 182, 645–652. [Google Scholar] [CrossRef] [PubMed]
  57. Martínez-Jiménez, M.C.; Muñoz, P.; Valerio, M.; Alonso, R.; Martos, C.; Guinea, J.; Bouza, E. Candida biomarkers in patients with candidaemia and bacteraemia. J. Antimicrob. Chemother. 2015, 70, 2354–2361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Trovato, L.; Astuto, M.; Castiglione, G.; Scalia, G.; Oliveri, S. Diagnostic surveillance by Candida albicans germ tube antibody in intensive care unit patients. J. Microbiol. Immunol. Infect. 2019, 53, 778–784. [Google Scholar] [CrossRef] [PubMed]
  59. Pini, P.; Colombari, B.; Marchi, E.; Castagnoli, A.; Venturelli, C.; Sarti, M.; Blasi, E. Performance of Candida albicans germ tube antibodies (CAGTA) and its association with (1 → 3)-β-D-glucan (BDG) for diagnosis daof invasive candidiasis (IC). Diagn. Microbiol. Infect. Dis. 2018, 93, 39–43. [Google Scholar] [CrossRef] [PubMed]
  60. Rouze, A.; The S-TAFE Study Group; Loridant, S.; Poissy, J.; Dervaux, B.; Sendid, B.; Cornu, M.; Nseir, S. Biomarker-based strategy for early discontinuation of empirical antifungal treatment in critically ill patients: A randomized controlled trial. Intensiv. Care Med. 2017, 43, 1668–1677. [Google Scholar] [CrossRef]
  61. Avni, T.; Leibovici, L.; Paul, M. PCR Diagnosis of Invasive Candidiasis: Systematic Review and Meta-Analysis. J. Clin. Microbiol. 2011, 49, 665–670. [Google Scholar] [CrossRef] [Green Version]
  62. Salimnia, H.; Fairfax, M.R.; Lephart, P.R.; Schreckenberger, P.; DesJarlais, S.M.; Johnson, J.K.; Robinson, G.; Carroll, K.C.; Greer, A.; Morgan, M.; et al. Evaluation of the FilmArray Blood Culture Identification Panel: Results of a Multicenter Controlled Trial. J. Clin. Microbiol. 2016, 54, 687–698. [Google Scholar] [CrossRef]
  63. Simor, A.E.; Porter, V.; Mubareka, S.; Chouinard, M.; Katz, K.; Vermeiren, C.; Fattouh, R.; Matukas, L.M.; Tadros, M.; Mazzulli, T.; et al. Rapid Identification of Candida Species from Positive Blood Cultures by Use of the FilmArray Blood Culture Identification Panel. J. Clin. Microbiol. 2018, 56, e01387-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sparks, R.; Balgahom, R.; Janto, C.; Polkinghorne, A.; Branley, J. Evaluation of the BioFire Blood Culture Identification 2 panel and impact on patient management and antimicrobial stewardship. Pathology 2021, 53, 889–895. [Google Scholar] [CrossRef]
  65. Mylonakis, E.; Clancy, C.J.; Ostrosky-Zeichner, L.; Garey, K.W.; Alangaden, G.J.; Vazquez, J.A.; Groeger, J.S.; Judson, M.A.; Vinagre, Y.-M.; Heard, S.O.; et al. T2 Magnetic Resonance Assay for the Rapid Diagnosis of Candidemia in Whole Blood: A Clinical Trial. Clin. Infect. Dis. 2015, 60, 892–899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Clancy, C.J.; Pappas, P.G.; Vazquez, J.; Judson, M.A.; Kontoyiannis, D.P.; Thompson, G.R.; Garey, K.W.; Reboli, A.; Greenberg, R.N.; Apewokin, S.; et al. Detecting Infections Rapidly and Easily for Candidemia Trial, Part 2 (DIRECT2): A Prospective, Multicenter Study of the T2Candida Panel. Clin. Infect. Dis. 2018, 66, 1678–1686. [Google Scholar] [CrossRef] [PubMed]
  67. Monday, L.; Acosta, T.P.; Alangaden, G. T2Candida for the Diagnosis and Management of Invasive Candida Infections. J. Fungi 2021, 7, 178. [Google Scholar] [CrossRef] [PubMed]
  68. Huang, Y.-S.; Wang, F.-D.; Chen, Y.-C.; Hsieh, M.-H.; Hii, I.-M.; Lee, Y.-L.; Ho, M.-W.; Liu, C.-E.; Chen, Y.-H.; Liu, W.-L. High rates of misidentification of uncommon Candida species causing bloodstream infections using conventional phenotypic methods. J. Formos. Med. Assoc. 2020, 120, 1179–1187. [Google Scholar] [CrossRef] [PubMed]
  69. Sanguinetti, M.; Porta, R.; Sali, M.; La Sorda, M.; Pecorini, G.; Fadda, G.; Posteraro, B. Evaluation of VITEK 2 and RapID Yeast Plus Systems for Yeast Species Identification: Experience at a Large Clinical Microbiology Laboratory. J. Clin. Microbiol. 2007, 45, 1343–1346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Kim, S.H.; Shin, J.H.; Mok, J.H.; Kim, S.Y.; Song, S.A.; Kim, H.R.; Kook, J.; Chang, Y.; Bae, I.K.; Lee, K. Misidentification of Candida guilliermondii as C. famata among Strains Isolated from Blood Cultures by the VITEK 2 System. Biomed. Res. Int. 2014, 2014, 250408. [Google Scholar] [CrossRef] [Green Version]
  71. Cheng, J.-W.; Yu, S.-Y.; Xiao, M.; Wang, H.; Kudinha, T.; Kong, F.; Xu, Y.-C. Identification and Antifungal Susceptibility Profile of Candida guilliermondii and Candida fermentati from a Multicenter Study in China. J. Clin. Microbiol. 2016, 54, 2187–2189. [Google Scholar] [CrossRef] [Green Version]
  72. Radic, M.; Goic-Barisic, I.; Novak, A.; Rubic, Z.; Tonkic, M. Evaluation of PNA FISH ® Yeast Traffic Light in identification of Candida species from blood and non-blood culture specimens. Med. Mycol. 2016, 54, 654–658. [Google Scholar] [CrossRef]
  73. Klingspor, L.; Lindbäck, E.; Ullberg, M.; Özenci, V. Seven years of clinical experience with the Yeast Traffic Light PNA FISH: Assay performance and possible implications on antifungal therapy. Mycoses 2017, 61, 179–185. [Google Scholar] [CrossRef] [PubMed]
  74. Gorton, R.L.; Ramnarain, P.; Barker, K.; Stone, N.; Rattenbury, S.; McHugh, T.D.; Kibbler, C.C. Comparative analysis of Gram’s stain, PNA-FISH and Sepsityper with MALDI-TOF MS for the identification of yeast direct from positive blood cultures. Mycoses 2014, 57, 592–601. [Google Scholar] [CrossRef] [PubMed]
  75. Delavy, M.; Dos Santos, A.R.; Heiman, C.M.; Coste, A.T. Investigating Antifungal Susceptibility in Candida Species with MALDI-TOF MS-Based Assays. Front. Cell. Infect. Microbiol. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  76. Jeffery-Smith, A.; Taori, S.K.; Schelenz, S.; Jeffery, K.; Johnson, E.M.; Borman, A.; Manuel, R.; Brown, C.S. Candida auris: A review of the literature. Clin. Microbiol. Rev. 2018, 31, e00029-17. [Google Scholar] [CrossRef] [Green Version]
  77. Burnham, J.P.; Wallace, M.A.; Fuller, B.M.; Shupe, A.; Burnham, C.-A.D.; Kollef, M.H. Clinical Effect of Expedited Pathogen Identification and Susceptibility Testing for Gram-Negative Bacteremia and Candidemia by Use of the Accelerate PhenoTM System. J. Appl. Lab. Med. 2019, 3, 569–579. [Google Scholar] [CrossRef] [Green Version]
  78. Garey, K.W.; Rege, M.; Pai, M.P.; Mingo, D.E.; Suda, K.J.; Turpin, R.S.; Bearden, D. Time to Initiation of Fluconazole Therapy Impacts Mortality in Patients with Candidemia: A Multi-Institutional Study. Clin. Infect. Dis. 2006, 43, 25–31. [Google Scholar] [CrossRef] [Green Version]
  79. Pappas, P.G.; Kauffman, C.A.; Andes, D.R.; Clancy, C.J.; Marr, K.A.; Ostrosky-Zeichner, L.; Reboli, A.C.; Schuster, M.G.; Vazquez, J.A.; Walsh, T.J.; et al. Clinical Practice Guideline for the Management of Candidiasis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 62, e1–e50. [Google Scholar] [CrossRef] [Green Version]
  80. Piarroux, R.; Grenouillet, F.; Balvay, P.; Tran, V.; Blasco, G.; Millon, L.; Boillot, A. Assessment of preemptive treatment to prevent severe candidiasis in critically ill surgical patients(*). Crit. Care Med. 2004, 32, 2443–2449. [Google Scholar] [CrossRef]
  81. Hermsen, E.D.; Zapapas, M.K.; Maiefski, M.E.; Rupp, M.; Freifeld, A.G.; Kalil, A.C. Validation and comparison of clinical prediction rules for invasive candidiasis in intensive care unit patients: A matched case-control study. Crit. Care 2011, 15, R198. [Google Scholar] [CrossRef] [Green Version]
  82. Dupont, H.; Bourichon, A.; Paugam-Burtz, C.; Mantz, J.; Desmonts, J.-M. Can yeast isolation in peritoneal fluid be predicted in intensive care unit patients with peritonitis? Crit. Care Med. 2003, 31, 752–757. [Google Scholar] [CrossRef]
  83. Bassetti, M.; Garnacho-Montero, J.; Calandra, T.; Kullberg, B.; Dimopoulos, G.; Azoulay, E.; Chakrabarti, A.; Kett, D.; Leon, C.; Ostrosky-Zeichner, L.; et al. Intensive care medicine research agenda on invasive fungal infection in critically ill patients. Intensiv. Care Med. 2017, 43, 1225–1238. [Google Scholar] [CrossRef] [Green Version]
  84. Dyar, O.J.; Huttner, B.; Schouten, J.; Pulcini, C. What is antimicrobial stewardship? Clin. Microbiol. Infect. 2017, 23, 793–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Rac, H.; Wagner, J.L.; King, S.T.; Barber, K.E.; Stover, K.R. Impact of an antifungal stewardship intervention on optimization of candidemia management. Ther. Adv. Infect. Dis. 2017, 5, 3–10. [Google Scholar] [CrossRef] [Green Version]
  86. Capoor, M.R.; Subudhi, C.P.; Collier, A.; Bal, A.M. Antifungal stewardship with an emphasis on candidaemia. J. Glob. Antimicrob. Resist. 2019, 19, 262–268. [Google Scholar] [CrossRef] [PubMed]
  87. Johnson, M.D.; Lewis, E.R.; Ashley, E.S.D.; Ostrosky-Zeichner, L.; Zaoutis, T.; Thompson, G.R.; Andes, D.R.; Walsh, T.J.; Pappas, P.G.; A Cornely, O.; et al. Core Recommendations for Antifungal Stewardship: A Statement of the Mycoses Study Group Education and Research Consortium. J. Infect. Dis. 2020, 222, S175–S198. [Google Scholar] [CrossRef] [PubMed]
  88. Tascini, C.; Bertolino, G.; Sozio, E.; Sbrana, F.; Ripoli, A.; Carmignani, C. Antifungal Stewardship Programs and candidemia. Clin. Infect. Dis. 2019, 70, 1522–1523. [Google Scholar] [CrossRef]
  89. Menichetti, F.; Bertolino, G.; Sozio, E.; Carmignani, C.; Del Turco, E.R.; Tagliaferri, E.; Sbrana, F.; Ripoli, A.; Barnini, S.; Desideri, I.; et al. Impact of infectious diseases consultation as a part of an antifungal stewardship programme on candidemia outcome in an Italian tertiary-care, University hospital. J. Chemother. 2018, 30, 304–309. [Google Scholar] [CrossRef] [PubMed]
  90. Rautemaa-Richardson, R.; Rautemaa, V.; Al-Wathiqi, F.; Moore, C.B.; Craig, L.; Felton, T.W.; Muldoon, E.G. Impact of a diagnostics-driven antifungal stewardship programme in a UK tertiary referral teaching hospital. J. Antimicrob. Chemother. 2018, 73, 3488–3495. [Google Scholar] [CrossRef] [Green Version]
  91. Ito-Takeichi, S.; Niwa, T.; Fujibayashi, A.; Suzuki, K.; Ohta, H.; Niwa, A. The impact of implementing an antifungal stewardship with monitoring of 1-3, β-D-glucan values on antifungal consumption and clinical outcomes. J. Clin. Pharm. Ther. 2019, 44, 454–462. [Google Scholar] [CrossRef]
  92. Aitken, S.; Beyda, N.D.; Shah, D.N.; Palmer, H.R.; Lasco, T.M.; Koo, H.; Garey, K.W. Clinical Practice Patterns in Hospitalized Patients at Risk for Invasive Candidiasis. Ann. Pharmacother. 2014, 48, 683–690. [Google Scholar] [CrossRef]
  93. Bomkamp, J.P.; Sulaiman, R.; Hartwell, J.L.; Desai, A.; Winn, V.C.; Wrin, J.; Kussin, M.L.; Hiles, J.J. Evaluation of a Rapid Fungal Detection Panel for Identification of Candidemia at an Academic Medical Center. J. Clin. Microbiol. 2020, 58, e01408-19. [Google Scholar] [CrossRef] [PubMed]
  94. Gill, C.M.; Kenney, R.M.; Hencken, L.; Mlynarek, M.E.; Alangaden, G.J.; Samuel, L.P.; Davis, S.L. T2 Candida versus beta-D-glucan to facilitate antifungal discontinuation in the intensive care unit. Diagn. Microbiol. Infect. Dis. 2019, 95, 162–165. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proposed Algorithm for pre-emptive antifungal therapy on ICU patients who underwent intra-abdominal surgery.
Figure 1. Proposed Algorithm for pre-emptive antifungal therapy on ICU patients who underwent intra-abdominal surgery.
Antibiotics 11 01804 g001
Table 1. Risk Factors for Invasive Candidemia.
Table 1. Risk Factors for Invasive Candidemia.
Risk Factors
1.
Major Risk Factors
Intravascular devices
Recent surgery (particularly abdominal surgery)
Broad-spectrum antibiotics/antifungals
Immunosuppressive therapy (corticosteroids and chemotherapy)
Malignancies (solid tumors and hematologic)
Diabetes mellitus
2.
Other Risk Factors
Hyperalimentation fluids
Previous ICU stay
Mechanical ventilation
Urinary catheterization
Prior Candida colonization/infection
Concomitant bacterial infections
Solid organ transplant patients
Hemodialysis
HIV-associated low CD4+ T cell counts
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alves, J.; Alonso-Tarrés, C.; Rello, J. How to Identify Invasive Candidemia in ICU—A Narrative Review. Antibiotics 2022, 11, 1804. https://doi.org/10.3390/antibiotics11121804

AMA Style

Alves J, Alonso-Tarrés C, Rello J. How to Identify Invasive Candidemia in ICU—A Narrative Review. Antibiotics. 2022; 11(12):1804. https://doi.org/10.3390/antibiotics11121804

Chicago/Turabian Style

Alves, Joana, Carles Alonso-Tarrés, and Jordi Rello. 2022. "How to Identify Invasive Candidemia in ICU—A Narrative Review" Antibiotics 11, no. 12: 1804. https://doi.org/10.3390/antibiotics11121804

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop