356
Views
0
CrossRef citations to date
0
Altmetric
REVIEW

Updated Perspectives on the Diagnosis and Management of Neonatal Invasive Candidiasis

ORCID Icon, , &
Pages 45-63 | Received 14 Aug 2023, Accepted 25 Oct 2023, Published online: 09 Nov 2023

Abstract

Invasive candidiasis can cause severe illness in immunocompromised hosts, such as premature infants. Clinical presentation in neonates is variable and often characterized by non-specific signs with potential to involve several organ systems. Awareness of risk factors for Candida infections in the neonatal intensive care unit (NICU) can aid in screening infants with signs and symptoms of generalized illness. Cultures of blood, urine, and cerebrospinal fluid are the main diagnostic tools available in this population of infants, but several biomarkers and alternate identification methodologies such as 1,3-β-D-glucan, serum mannan or anti-mannan, and T2 magnetic resonance testing are being studied in the neonatal population. Prompt diagnosis of Candida infection, in conjunction with a comprehensive assessment of disease progression and organ involvement, is critical for optimizing treatment and patient outcomes. Supportive care and systemic antifungal medications remain the mainstay of treatment, and the efficacy and safety of newer therapeutic agents continue to be evaluated in neonates. Disease prevention strategies must be thoughtfully implemented and customized to each individual NICU based on local incidence of Candida infection, practice patterns, and risk factors, and may include prophylactic antifungal therapy. This review summarizes the evidence for current approaches to diagnosis and management of neonatal invasive candidiasis and provides an overview of the newer diagnostic tools and therapeutic agents on the horizon.

Introduction

Invasive candidiasis is an important contributor to neonatal morbidity and mortality. Although this fungal organism colonizes human skin and mucosa and may cause only limited mucocutaneous disease in immunocompetent hosts,Citation1,Citation2 immunocompromised hosts, such as premature infants, with invasive candidiasis can suffer from severe and life-threatening illness.

The incidence of invasive candidiasis in extremely low birth weight infants is approximately 9%, although this incidence varies by neonatal intensive care unit (NICU) from 2% to 28%.Citation3 Candida can spread to premature infants via vertical and horizontal transmission.Citation4 Vertical transmission to newborns can occur from the maternal genitourinary tract during labor.Citation5 A significantly higher incidence of colonization is seen after vaginal delivery compared to infants delivered via caesarean section.Citation6 Alternatively, horizontal transmission may occur from exposures within the NICU environment.Citation5 Once acquired, Candida invasion can lead to severe disease in nearly any organ system of an infant, including the eyes, brain, heart, liver, spleen, genitourinary system, bones, and joints.Citation7,Citation8

Most cases of neonatal invasive candidiasis are caused by Candida albicans and Candida parapsilosis.Citation7 Candida glabrata, Candida tropicalis, and Candida krusei are also pathogens in infants,Citation9 and Candida auris has recently emerged as a significant threat to neonates as it can cause severe systemic disease and is resistant to many first-line antifungal agents.Citation10 In low- and middle-income countries, a higher proportion of infections are caused by non-albicans Candida species as compared to high-income countries.Citation11 Species identification is an important step in diagnosis of invasive candidiasis as it can predict mortality risk and guide treatment decisions.Citation12

Risk factors for invasive candidiasis are commonly present in the NICU patient population (). While prematurity and extremely low birth weight are commonly cited predisposing factors, other important considerations include delayed enteral feeding; use of broad-spectrum antibiotics, H2 blockers, or systemic steroids; Candida colonization of skin or gastrointestinal (GI) mucosa; invasive instrumentation via central venous catheters (CVC), urinary catheters, or mechanical ventilation; hospitalization longer than 7 days; presence of sepsis or shock; and coagulation derangements such as disseminated intravascular coagulation or thrombocytopenia.Citation3,Citation4,Citation7,Citation13 Factors such as translocation across the GI tract after colonization are major sources of invasive infection but not easily modifiable.Citation14 Use of central lines, on the other hand, is a potentially modifiable risk factor and a potential source of intervention to mitigate the risk of invasive infection.Citation8 Of note, these risk factors are most predictive of invasive candidiasis infections for neonates in high-income countries; invasive candidiasis infections occur more frequently in neonates born at later gestational ages and higher birth weights in low- and middle-income countries.Citation15

Table 1 Risk Factors for Neonatal Invasive Candidiasis

Although the incidence of invasive neonatal candidiasis has decreased over the past few decades, it continues to be a significant source of neonatal morbidity and can cause life-threatening illness.Citation16 The Centers for Disease Control and Prevention’s population-based surveillance demonstrates that rates of invasive candidiasis in infants from the United States have dropped dramatically from 2009 to 2012. Since then, rates continue to slowly decline, with an estimated incidence of 9.6% in 2019 for children less than 1 year old.Citation17 Incidence is lower in other high-income countries, such as Italy, the United Kingdom, Canada, and Australia.Citation18–21 Incidence of invasive candidiasis in pediatric intensive care units is significantly higher in low- and middle-income countries when compared to high-income countries; however, the incidence in the neonatal population has not been well characterized.Citation11,Citation15

Over 20% of infants with invasive candidiasis die in spite of systemic antifungal treatment, and those who survive are at high risk for neurodevelopmental impairment.Citation12,Citation22,Citation23 Other long-term consequences of this illness include risk of moderate or severe cerebral palsy, blindness, deafness, and increased healthcare costs.Citation7,Citation12 Early identification and prompt initiation of appropriate treatment can optimize both short-term and long-term outcomes for premature infants receiving care in the NICU.Citation24 This review summarizes the evidence for current approaches to diagnosis and management of neonatal invasive candidiasis and provides an overview of the newer diagnostic tools and therapeutic agents on the horizon.

Diagnosis

Manifestation of Disease

Neonates with invasive candidiasis often present with nonspecific signs similar to that of sepsis, including lethargy, apnea, feeding difficulties, and hemodynamic instability.Citation7,Citation8,Citation25 However, nearly every organ system can be affected. Neurologic invasion is more commonly characterized by meningoencephalitis, which may present as seizures or intraventricular hemorrhage,Citation7,Citation26 and less commonly by obstructive hydrocephalus with ventriculitis or cerebral abscesses.Citation27 The most common ophthalmologic manifestation is endophthalmitis, an infection of the internal ocular spaces that occurs through hematogenous spreadCitation28,Citation29 and is characterized by chorioretinitis and/or vitreal lesions.Citation26 The cardiovascular system is rarely involved, presenting as endocarditis or pericarditis.Citation7,Citation10 Hepatosplenic abscesses and hepatosplenic candidiasis should be considered when there is disseminated candidiasis, persistent fever, and abdominal symptoms.Citation26,Citation27 Intestinal perforation has also been associated with candidemia.Citation30 Genitourinary system involvement takes many forms, including fungal bezoars leading to obstructive nephropathy and renal parenchymal abnormalities.Citation26,Citation31 Bones and joints can be involved through osteoarticular lesions.Citation26 Skin involvement is a hallmark of congenitally acquired disease and consists of a widespread, erythematous, vesiculopapular rash that can progress to severe illness.Citation25

Testing and Imaging Considerations

The diagnostic approach to invasive candidiasis in neonates should be comprehensive, including physical examination, laboratory testing, and evaluation for multiorgan involvement (). The most common sites of infection are blood, urine, and cerebrospinal fluid (CSF);Citation1 therefore, cultures of these sites are recommended as an initial step in diagnosis. It is important to note that routine blood cultures are sufficient for recovering all varieties of Candida species, and the use of fungal blood cultures, intended to recover intracellular or more fastidious fungi, does not improve recovery rates.Citation32–35 Other sites that raise suspicion for infection, such as peritoneal fluid and tracheal aspirate, can also be collected and cultured to evaluate for Candida infection.Citation36 The major advantage of microbiologic diagnosis via cultures is species identification and susceptibility testing, especially in the setting of drug-resistant strains of C. glabrata, C. parapsilosis, and C. auris.Citation37 Newer microbiologic methods for rapid identification of species such as the multiplex polymerase chain reaction (PCR) FilmArray blood culture identification panel (bioMéieux, Inc.) may facilitate more prompt initiation of optimal antifungal therapy.Citation38

Table 2 Tools for Diagnosis of Neonatal Invasive Candidiasis: Strengths and Limitations

While cultures remain the gold standard for diagnosis, they have important limitations (). A study of 74 infants with invasive candidiasis calculated a median of 36 hours between blood culture draw and Candida detection for infants not on antifungal therapy and 42 hours for infants on empiric antifungal therapy.Citation39 Additionally, the sensitivity of routine blood cultures incubated on conventional automated blood culture systems in diagnosing any form of invasive candidiasis in any age group is approximately 50% with an average detection time ranging from 14 to 38 hours.Citation40,Citation45 A study of 150 NICUs demonstrated that of 19 infants with culture-positive Candida meningoencephalitis, only 37% had positive blood cultures.Citation46 The sensitivity of blood cultures to detect Candida may be related to total disease burden. In one small study of autopsies on infants with invasive candidiasis, blood cultures were positive in 28% of patients with one infected organ, whereas blood cultures were positive in 78% of patients with greater than 3 infected organs.Citation47 Furthermore, cultures can be persistently positive despite systemic antifungal treatment, as demonstrated by a prospective study in 2006 where 10% of neonates had candidemia for greater than or equal to 14 days after starting therapy.Citation12 Negative cultures do not preclude a diagnosis of invasive candidiasis; neonates can have invasive candidiasis in the absence of proven candidemia and therefore are at risk for mortality and lasting neurodevelopmental delay if infection is not identified promptly.Citation37,Citation48

A barrier to optimizing the yield of blood cultures in premature infants is the blood volume required for detection.Citation37 One must balance obtaining adequate volume for diagnosis while not exceeding the threshold that results in hemodynamic and cardiorespiratory instability.Citation49,Citation50 In adults, a blood culture volume of approximately 10 mL is standard;Citation51 in infants, recommendations vary. Existing pediatric guidelines recommend volume limits between 1% and 5% of total blood volume over 1 day and up to 10% of total blood volume over 8 weeks.Citation52 The 2018 update by the Infectious Diseases Society of America (IDSA) and American Society for Microbiology recommends collecting 4% of total blood volume in a single aerobic blood culture in patients weighing less than 2 kg and 3% in patients weighing 2.1–12.7 kg.Citation51 A 2015 in vitro study on banked adult whole blood demonstrated that at low concentrations (1–10 CFU/mL), Candida albicans and Candida parapsilosis were detected in volumes as low as 0.5 mL.Citation53 At very low concentrations (<1 CFU/mL), a volume of 3 mL was required for detection of Candida albicans and Candida parapsilosis.Citation53 However, obtaining these volumes is not always feasible. A study of pediatric blood cultures at Royal Children’s Hospital Melbourne determined that 39% of blood samples drawn in infants younger than 1 month of age had negligible volume for culture.Citation54

In the setting of positive blood or urine cultures, the 2016 IDSA clinical practice guidelines for the management of candidiasis recommend a lumbar puncture and dilated retinal examination to assess for meningoencephalitis and endophthalmitis, respectively.Citation7 The IDSA strongly recommends a computerized tomography scan or ultrasound of the genitourinary tract, liver, and spleen when blood cultures are persistently positive to identify other potential sites of infection.Citation7 Tissue biopsy can also be performed to detect invasive candidiasis via microscopy and histologic examination, especially in cases of suspected hepatosplenic candidiasis; however, the rates of detection are low at approximately 50%.Citation55

In addition to the dilated retinal examination recommended by the IDSA, new imaging technologies to diagnose Candida endophthalmitis are in development. The currently recommended evaluation tool, a dilated fundoscopic exam, may show retinal lesions, which would correspond to hyperfluorescent lesions with ill-defined margins and irregular edges on fluorescein angiography.Citation56 However, B-scan ultrasound and handheld spectral-domain optical coherence technology (HH-SD OCT) may identify pathology beyond retinal lesions. B-scan ultrasound has been used in neonates and can show infiltrates in the vitreous cavity, subretinal fluid, and/or retinal detachment.Citation29 HH-SD OCT, the newest of these technologies, has been used in neonates for retinopathy of prematurity evaluations but can also demonstrate vitreal lesions in addition to retinal lesions.Citation56

Serum Biomarkers

Because of the aforementioned limitations of diagnosing candidiasis with blood cultures, other surrogate markers of invasive fungal infection in blood and serum can be employed to make a more timely diagnosis, leading to faster initiation of treatment (). Laboratory derangements frequently associated with invasive candidiasis include thrombocytopenia, hyperglycemia, and leukocytosis. Neonates with sepsis from Candida are more likely to have thrombocytopenia and hyperglycemia than infants with bacterial sepsis.Citation3,Citation57 However, there is conflicting evidence on the significance of thrombocytopenia as a marker for fungal sepsis.Citation8,Citation58–60 Leukocytosis is not a reliable marker as up to 40% of infants with fungal sepsis have a normal white blood cell count.Citation61

Three additional markers targeting specific Candida fungal components are 1,3-β-D-glucan, serum mannan or anti-mannan, and T2 magnetic resonance testing (T2Candida). 1,3-β-D-glucan is a constituent of the cell wall of many fungi and can be detected in the serum during invasive fungal infection. Its concentration is significantly higher in infants with invasive candidiasis as compared to those without disease.Citation8,Citation31 This molecule can be detected via a variety of widely available assays, including the Fungitell Assay (Associates of Cape Cod, Inc., Massachusetts, USA) and the Wako β-Glucan test (Fujifilm Wako Pure Chemical Corp, Tokyo, Japan). A 2013 meta-analysis of neonatal studies demonstrated that the serum tests to detect this polysaccharide have a sensitivity of 89% and specificity of 60% in diagnosis of neonatal candidiasis, although there was considerable variability between studies.Citation62 The average baseline 1,3-β-D-glucan level in healthy children and infants is higher than in adults, which should be considered when interpreting these test results;Citation63 when using a higher positivity threshold, sensitivity and specificity were 81% and 80%, respectively.Citation62 1,3-β-D-glucan levels may also be used to monitor response to treatment, as they decrease over the course of appropriate antifungal therapy.Citation31 A major limitation of 1,3-β-D-glucan testing is the high false-positive rate.Citation64,Citation65 False positives can occur with the use of various beta-lactam antibiotics, the administration of blood products (such as albumin or intravenous immune globulin), bacterial infections, the use of surgical gauze and other related materials, the use of leukocyte-removing filters or extracorporeal membrane treatments, and sample collection from contaminated central venous catheters – many factors frequently seen in hospitalized neonates.Citation66–68 Further investigation is still needed to better understand this marker’s ability to accurately identify disease and monitor progression.

Mannan is an abundant constituent of the Candida cell wall. Therefore, detection of mannan antigen and serum anti-mannan antibodies in the serum or plasma represent potential targets for the diagnosis of invasive candidiasis. The best performance using this combined mannan/anti-mannan antibody testing approach has been observed with the commercial PLATELIA™ Candida Ag Plus system (Bio-Rad Laboratories, Marnes-la-Coquette, France), which has been studied predominantly in immunosuppressed hosts with neutropenia and/or impairment in cell-mediated immunity. While evaluation of the performing characteristics of this test in neonates is very limited, a preliminary study of 70 neonates reported a sensitivity of 94.4% and specificity of 94.2% with a positive predictive value of 85% and negative predictive value of 98% based on a prevalence of 6.5%.Citation41 However, similar investigation in adults demonstrated high specificity but low sensitivity, particularly for C. parapsilosis and C. krusei infection, thought to be due to differences in mannose epitopes across species.Citation8,Citation41 Ultimately, utilizing this test in combination with blood cultures may improve the diagnosis of early invasive candidiasis.Citation41 This testing is more commonly used in European centers and rarely employed for diagnosis in North America.Citation40

T2Candida (T2 Biosystems, Inc., Wilmington, MA, USA) is an FDA-approved test that combines amplification steps and magnetic resonance imaging to rapidly identify the 5 most common Candida species in blood samples in an average of 4.4 hours.Citation43 In a 2022 retrospective single-center study of 106 pediatric patients, including neonates, the T2Candida assay was 100% sensitive, 94.1% specific, and provided definitive organism identification in 3.7 hours on average; however, only 4 patients with positive blood cultures for Candida were included in the study.Citation42 In another small pediatric study, blood T2Candida correctly identified more candidemia diagnoses than blood cultures.Citation69 Like 1,3-β-D-glucan testing, T2Candida outperformed blood cultures for monitoring candidemia and therefore could be useful for following treatment response.Citation70 While multicenter studies have demonstrated high sensitivity and specificity for targeted Candida species, the sensitivity in clinical practice is low, ranging from 33% to 45% for deep-seated candidiasis and 65–83% for candidemia.Citation43,Citation71 Limitations of this test include the need for implementation in high pre-test probability settings, such as critical care settings with risk factors for invasive candidiasis, and the need for onsite testing to yield short turnaround times. However, evaluations indicate that its use is cost effective, including in resource-limited settings.Citation72,Citation73

Identification Methods

Matrix-assisted laser deposition/ionization time-of-flight (MALDI-TOF) is a common method used to identify organisms, including a variety of Candida species, isolated from positive microbiological cultures. Traditionally, clinical microbiological identification often required growth from subcultures, which may take 24 to 72 hours; however, more recent studies have demonstrated that direct identification can be done from positive blood culture specimens, decreasing time to diagnosis.Citation74–76 This test can identify over 200 species of bacteria and yeasts with just 1 minute of hands-on time and 5 minutes of turn-around time.Citation48 In a pre-post quasi-experimental study of adults with bacterial and fungal bloodstream infections, including Candida, use of MALDI-TOF in conjunction with antibiotic stewardship decreased time to organism identification and time to antifungal therapy, which was associated with a decrease in mortality and ICU length of stay.Citation77

Fluorescent in-situ hybridization (FISH), a technique that uses fluorescent microscopy to identify hybridization of probes to organism-specific rRNA, has also been studied to rapidly identify Candida species from positive blood cultures.Citation78 Like MALDI-TOF, this test is fast, requiring only 5 minutes of hands-on time and 90 minutes of turn-around time after culture growth and positivity.Citation48 Another unique aspect of FISH testing is its ability to detect Candida in peritoneal fluid in addition to blood.Citation78 A 2018 analysis of the Peptide Nucleic Acid FISH Yeast Traffic Light System (AdvanDx, Woburn, MA) demonstrated that this test could quickly and accurately diagnose candidemia, as it identified Candida in 96.4% of patients not yet on antifungal therapy.Citation79 However, drawbacks of this test include false positive and negative results related to polymicrobial infections and that few clinical laboratories currently employ this technique.

Panfungal PCR tests can identify several common Candida species that cause bloodstream infections in NICU patients.Citation44 The main advantages of PCR tests are their speed and sensitivity; positive PCR results are available on average 2.2 days before positive blood culture results.Citation80,Citation81 PCR has also demonstrated greater sensitivity than blood cultures for detecting neonatal invasive candidiasis in very low birth weight infants, for whom obtaining adequate volume for blood culture is especially difficult. A 2017 multicenter, observational case–control study in very low birth weight infants reported panfugal PCR testing sensitivity of 87.5% and specificity of 81.6%; PCR targets were identified in 17.4% of septic patients with negative blood cultures.Citation44 In low prevalence settings, panfungal PCR has a high negative predictive value, which can be useful when deciding to start and/or stop antifungal therapy, thereby limiting potential drug toxicity.Citation37 This test has several disadvantages, including its lack of standardization and approval for use in the United States, limited study in neonates, and low positive predictive value in low prevalence cohorts.Citation37,Citation44,Citation82 One subtype of PCR-based testing is cationic conjugated polymer fluorescence resonance energy transfer. This system, composed of a cationic conjugated polymer fluorescent probe and pathogen-specific DNA labeled with fluorescent dyes, was developed to diagnose neonatal invasive fungal infection quickly and accurately with a detection limit that is one-tenth that of real-time PCR testing.Citation83,Citation84 The system’s major disadvantage is the need for optimization and judicious primer selection.Citation83

Metagenomic next-generation sequencing is a newer testing methodology that simultaneously sequences billions of nucleic acid fragments in parallel, allowing for the rapid identification of bacteria, viruses, and fungi.Citation85 This method has been used in pediatric populations in the evaluation of sepsis, meningitis, and encephalitis and shown promise in positively impacting clinical care.Citation86,Citation87 A 2023 systematic review on next-generation sequencing determined that this technology improves etiologic identification of neonatal and pediatric fungal sepsis, but neonatal data is limited and requires further characterization.Citation87 The major advantage of this technology is its unbiased approach to identifying all potential pathogens within the same test without the use of specific primers or probes.Citation85 Disadvantages of this method include its inability to distinguish organisms that are pathogenic versus those which are colonizers, its high cost, and long turnaround time. Only one commercially available laboratory can perform this testing at this time (Karius, Redwood City).Citation85 While next-generation sequencing has been applied to the diagnosis of invasive mold infections in immunocompromised hosts, there is a paucity of evidence in invasive Candida infections, particularly in the neonatal population.Citation88,Citation89

Management

Once invasive candidiasis has been diagnosed, expedient and effective management is critical to minimize risk of long-term morbidity. A combination of supportive care and antifungal therapeutics is often employed. Supportive care mostly centers on device management; indwelling materials are potential reservoirs for continued infection despite treatment with systemic antifungal therapy. Infection risk increases with duration of CVC use, as indwelling catheters provide a means for Candida to enter the bloodstream and create a surface for biofilm formation.Citation8 The most recent IDSA guidelines strongly recommend removal of CVCs and central nervous system (CNS) devices in the setting of invasive candidiasis.Citation7 Prompt removal of CVCs in infants with invasive candidiasis is critical; it is associated with better immediate and long-term outcomes as well as lower rates of neurodevelopmental impairment.Citation12

Therapeutics

The mainstay of treatment for neonatal invasive candidiasis is systemic antifungal therapy (). Prompt administration of antifungal therapeutics is essential; neonatal mortality increases as the duration between onset of symptoms and initiation of antifungal therapy increases.Citation24 Extrapolation of adult data regarding therapy selection and dosing warrants caution due to important differences in immune response, disease pathology, and drug metabolism in neonates.Citation90 However, neonatal treatment guidelines are mostly based on small, single-center studies and a limited number of multicenter cohort studies.Citation7

Table 3 Overview of Antifungal Therapy Used in the Treatment of Neonatal Invasive Candidiasis

Amphotericin B is a first-line therapy for the treatment of invasive candidiasis of neonates, as recommended by the IDSA,Citation7 European Society for Clinical Microbiology and Infectious Diseases (ESCMID),Citation91 and the joint German Speaking Mycological Society (DMYG) and Paul Ehrlich Society for Chemotherapy (PEG) guidelines ().Citation9,Citation55 It is most commonly administered in the amphotericin B deoxycholate form in neonates; the lipid formulations are rarely used in infants due to poor urinary tract penetration.Citation7,Citation90 Alternatively, fluconazole is another first-line therapy supported by the IDSA, ESCMID, and DMYG-PEG.Citation7,Citation55,Citation91 When treating with fluconazole, the ESCMID recommends a loading dose. Because of its prolonged half-life and large volume of distribution in neonates, a loading dose of fluconazole may be required to quickly achieve adequate antifungal activity in neonates.Citation92–94 Alternative agents, including micafungin and caspofungin, have poor penetration in the urinary tract and CSF.Citation12,Citation55,Citation91 Additionally, in the setting of neuroinvasive disease, 5-flucytosine can be added to the treatment regimen of an infant who does not respond to amphotericin B alone, and fluconazole is recommended as step-down rather than first-line therapy by the IDSA.Citation7

Table 4 Guidelines for the Treatment of Invasive Candidiasis in Neonates by Infectious Disease Society

Recommended treatment duration depends on extent of disease spread. In candidemia, treatment should be continued for 2 weeks after clearance of blood cultures and until resolution of signs and symptoms of invasive candidiasis.Citation7 For CNS infection, treatment should be continued for at least 3 weeks and until all signs, symptoms, CSF abnormalities, and imaging abnormalities have resolved.Citation7,Citation95 For urinary tract infection, therapy for 10 to 14 days is recommended.Citation95 For invasive candidiasis associated with focal infections, such as endocarditis or renal fungal masses, prolonged therapy for several weeks is needed, in addition to surgical removal of focal infection, until all clinical signs, blood culture, and imaging findings have normalized.Citation95

Amphotericin B

Amphotericin B is recommended by all three societies that have published recent guidelines on the treatment of neonatal invasive candidiasis. This compound in the polyene drug class interacts with ergosterol, the main sterol in the cell membrane and cell walls of many fungi, including Candida.Citation36 Interaction with ergosterol leads to the formation of pores in the cell membrane through which electrolytes and proteins can move freely, ultimately leading to cell death.Citation36 Advantages of this fungicidal drug include its broad spectrum of activity and good tissue penetration, including into the CNS.Citation7,Citation96 Additionally, this drug is well tolerated in infants and has little risk of nephrotoxicity as compared to adults and older children.Citation7 However, it lacks adequate oral absorption and has high rates of Candida auris resistance (up to 30–40%).Citation97,Citation98

There are many formulations of amphotericin B, including amphotericin B deoxycholate, liposomal amphotericin B, amphotericin B lipid complex (ABLC), and amphotericin B colloidal dispersion (ABCD). Amphotericin B deoxycholate is the most recommended formulation, administered at a dose of 1 mg/kg/day in neonates.Citation7,Citation90 It circulates in plasma highly bound to protein, is taken up by reticulo-endothelial organs, and is eventually excreted in urine and feces.Citation99 The pharmacokinetics of amphotericin B in children are characterized by a lower volume of distribution and faster clearance than adults, but neonate-specific activity is highly variable.Citation100–105 This formulation also penetrates the CSF well in neonates; amphotericin B CSF concentrations reach 40–90% of simultaneous serum concentrations.Citation100 Documented side effects include infusion-related reactions, hypokalemia, hepatotoxicity, and renal toxicity; however, in general, neonates tolerate this drug very well.Citation9,Citation106 The incidence of renal toxicity attributable to amphotericin B deoxycholate, especially in infants, is questionable, as studies yield contradictory conclusions.Citation90 For instance, a 1990 study of 36 infants determined that 54% of patients who received a total of more than 5 mg/kg had laboratory abnormalities consistent with renal dysfunction.Citation107 In contrast, a 2003 study of 56 infants demonstrated that no infants on any formulation of amphotericin B had a deterioration in renal function.Citation108 On the other hand, a 2009 study of 92 infants on amphotericin B concluded that 16% experienced nephrotoxicity, defined as a serum creatinine rise of at least 0.4 mg/dl at any time during antifungal therapy, and this abnormality resolved in all but 1 infant by the end of therapy.Citation109 One retrospective study of 25 extremely low birth weight infants with fungal sepsis suggested that at least 4 mEq/kg/day sodium intake and adequate hydration while on amphotericin B deoxycholate therapy may adequately reduce the risk of renal injury.Citation110 Concern for nephrotoxicity should not prevent administration of amphotericin B deoxycholate in neonates.

Although amphotericin B deoxycholate is the most appropriate formulation choice for neonates, lipid formulations of amphotericin B also exist. Drugs within this class include liposomal amphotericin B, ABLC, and ABCD. These larger molecule formulations are attractive due to their side effect profile but cost more than amphotericin B deoxycholate and may be less effective in treating urinary tract disease due to reduced renal excretion.Citation7,Citation82,Citation90 Additionally, CSF penetration is not well characterized. As CSF and urine are common sites of neonatal invasive candidiasis, these drugs should be used with caution.

An oral formulation of amphotericin B, encochleated amphotericin B, is currently under investigation. Unlike the other forms of amphotericin B, this drug’s composition evades degradation by the GI tract, allowing for oral administration with reduced toxicity.Citation100 It has demonstrated comparable activity to amphotericin B deoxycholate in vitro and in mouse models, and Phase 1 and 2 trials are ongoing.Citation111

Triazoles

Triazoles inhibit the cytochrome P (CYP) 450 enzyme that synthesizes ergosterol.Citation36,Citation90 First-generation triazoles include fluconazole and itraconazole, and second-generation triazoles include voriconazole and posaconazole. Newer triazoles include isavuconazole and opelconazole, and oteseconazole is a tetrazole currently under investigation. Common pharmacokinetic and pharmacodynamic characteristics of all triazoles include their time-dependent, rather than concentration-dependent, fungistatic activity; their interactions with CYP 450 enzymes; and their relatively safe side effect profiles.

Fluconazole 12 mg/kg/day is the triazole most often recommended for neonatal invasive candidiasis treatment.Citation7,Citation55,Citation91 Pharmacokinetic and pharmacodynamic characteristics of note include low protein binding, leading to high penetration in tissues, urine and CSF; renal elimination; and higher median 24 hour area under the curve in infants born at less than 30 weeks’ gestation compared to infants born at greater than 30 weeks’ gestation, necessitating dose adjustment based on gestational and postnatal age.Citation90,Citation112 A unique feature of fluconazole treatment is the use of a 25 mg/kg loading dose.Citation91 A prospective, single-center, open-label pharmacokinetics and safety trial in 10 infants <60 days old demonstrated that a 25 mg/kg fluconazole loading dose was effective and well tolerated with few adverse events, with all infants reaching 24-hour trough concentrations of >8 µg/mL following the loading dose and the majority of infants reaching the therapeutic target area under the curve.Citation94 Benefits of this drug include its efficacy, activity against the most common Candida strains,Citation112 availability in IV and oral formulations, high bioavailability, and tolerability.Citation90 Of note, strains of C. glabrata and C. krusei have demonstrated resistance to fluconazole.Citation90 There are disadvantages to fluconazole use. Fluconazole is a strong CYP2C9 and CYP3A4 inhibitor, which raises concern for potential drug–drug interactions.Citation90 It requires dose adjustment for renal insufficiency, but specific dose adjustments for renal function in neonates have not been well studied.Citation112 Side effects are minimal and most commonly include gastrointestinal symptoms or rash but rarely include hepatotoxicity and QT interval prolongation.Citation90,Citation96 Ultimately, while fluconazole has demonstrated activity against Candida, it is generally recommended as first-line therapy only in cases of uncomplicated disease without prior triazole exposure. In the setting of invasive candidiasis, fluconazole is more frequently used as a step-down therapy.Citation7,Citation55,Citation91

Itraconazole, another first-generation triazole with fungistatic activity against yeast-like fungi and molds, is well tolerated and available in an oral formulation.Citation90 Limited safety and efficacy data are available for itraconazole use in infancy. A review of 32 studies in neonates and infants concluded that a dose of 10 mg/kg daily was effective and safe for systemic fungal infections.Citation113 Specifically, one double-blinded randomized control trial of 43 pediatric patients, including infants, demonstrated that 10 mg/kg/day itraconazole cleared candidemia at a similar rate as fluconazole with very few adverse effects.Citation114 Like fluconazole, itraconazole exhibits CYP3A4 inhibition, and GI symptoms are the most common side effect.Citation90 Unlike fluconazole, itraconazole is highly protein bound and undergoes extensive hepatic metabolism before excretion by the liver and kidneys.Citation90 It additionally exhibits variable, dose-dependent oral bioavailability, which makes predicting its efficacy more difficult.Citation90 Its oral bioavailability is also determined by stomach pH and gastric retention time; for optimal absorption, the drug should be administered on an empty stomach, which is difficult to accomplish in the neonatal population due to their feeding schedules.Citation115

Voriconazole has moderate protein binding, resulting in good penetration into tissues and CSF and extensive metabolism by CYP2C19, but interpatient bioavailability is highly variable.Citation90,Citation116 Voriconazole is also more potent than other azoles, especially in infections caused by C. glabrata and C. krusei,Citation9 and has demonstrated activity against the highly resistant C. auris.Citation10 Voriconazole has been studied in neonates and infants, and children younger than 3 years old require higher daily doses as compared to older children, with dosing recommendations ranging from 2 mg/kg twice daily to 6 mg/kg three times daily.Citation117–120 Intra-vitreal voriconazole has been used for neonatal Candida endophthalmitis with gradual improvement in imaging findings over several weeks.Citation121 Unlike fluconazole, voriconazole has activity against Aspergillus species.Citation96 However, there is minimal excretion of the active drug into the urine, preventing effective treatment of urinary tract infection.Citation116 This drug is available in both IV and oral formulations, but the IV formulation should be used with caution in the setting of renal insufficiency.Citation122 Potential side effects include elevated transaminases, skin photosensitization, and abnormal vision.Citation90,Citation116 Ultimately, voriconazole is approved by the US Food and Drug Administration (FDA) for children aged 2 years or older.

Like voriconazole, posaconazole is an expanded-spectrum triazole with activity against some first-generation triazole-resistant Candida species.Citation90 This drug is available in IV and oral formulations, but the IV formulation is more commonly used in pediatrics because the highly bioavailable oral formulation is packaged in tablet form. In the body, posaconazole circulates highly bound to protein before being eliminated mainly in the feces but also by the kidneys.Citation90 The most commonly noted side effect is GI upset.Citation90 Posaconazole is most notably used in adults as antifungal prophylaxis;Citation123 there are limited data on the neonatal use of posaconazole, and the drug is not currently FDA approved for use in infants. Drug monitoring is critical if using voriconazole or posaconazole in children and neonates due to the drugs’ variable bioavailability and potential for toxicity.Citation116,Citation124

Recently developed triazoles include isavuconazole and opelconazole; a tetrazole, oteseconazole, is also under investigation. All three drugs have demonstrated fungicidal activity against Candida species, including fluconazole-resistant strains.Citation90,Citation125 Opelconazole is uniquely available in an inhaled formulation, allowing administration directly to the lungs.Citation126 Initial studies of isavuconazole and oteseconazole in adults demonstrate that they are well tolerated, with common side effects of headaches, rhinitis, and GI symptoms.Citation90,Citation126 Isavuconazole was approved by the FDA in 2015 for the treatment of invasive aspergillosis and invasive mucormycosis in the form of isavuconazonium sulfate, and oteseconazole has been approved to reduce the incidence of vulvovaginal candidiasis in adults. Further studies in neonates are still needed.

Echinocandins

Echinocandins are an option for salvage therapy of invasive candidiasis in the setting of resistance to or toxicity from other first-line agents.Citation112 They block cell wall synthesis by inhibiting 1,3-β-D-glucan synthase.Citation36 This drug class has several advantageous features. Echinocandins have activity against many triazole-resistant species, including certain strains of C. albicans, C. parapsilosis, C. glabrata, C. tropicalis, C. krusei, and C. auris.Citation90,Citation127 They are fungicidal against Candida and fungistatic against Aspergillus species.Citation96 They are well tolerated with minimal side effects or drug interactions due to their fungus-specific mechanism of action. However, they have relatively poor penetration in the CSF and urine, necessitating high drug concentrations for therapeutic effect.Citation12 Additional limitations include poor oral availability, requiring IV administration, and elevated minimal inhibitory concentrations against C. parapsilosis.Citation90,Citation128

Micafungin is the most studied echinocandin in neonates. Important pharmacokinetic and pharmacodynamic characteristics include concentration-dependent fungicidal activity, high protein binding, elimination via the biliary system, and weight-dependent clearance.Citation90 Micafungin exhibits dose-dependent penetration of the CNS; higher doses should be used for suspected or proven CNS involvement.Citation7 Additionally, higher doses are required to achieve adequate micafungin exposure as body weight decreases;Citation129 therefore, young infants with very high clearance require high micafungin doses of 9–15 mg/kg/day to achieve a similar mean area under the curve compared to an adult dose of 150 mg or a pediatric dose of 2 mg/kg/day.Citation55,Citation90,Citation91,Citation130 Micafungin has demonstrated efficacy against invasive candidiasis in the limited neonatal data that exists and is licensed by regulatory agencies, but knowledge gaps on its use in neonates remain. A Phase 3, randomized, double-blind, multicenter, parallel-group, noninferiority trial performed on infants with invasive candidiasis between 2 and 120 days old showed fungal-free survival in 60% of infants treated with micafungin versus 70% of infants treated with amphotericin B deoxycholate; however, the study ended early due to low recruitment.Citation131 Another sub-study of a randomized double-blind trial comparing micafungin and liposomal amphotericin B in premature infants and children found that micafungin was similarly effective and better tolerated than liposomal amphotericin B.Citation132 Micafungin is a relatively safe drug; its most common side effects include transient transaminitis and hypokalemia.Citation133 Further studies are needed to fully characterize this drug’s activity and safety in neonates.

Other echinocandins of note include caspofungin, anidulafungin, and rezafungin. Important features of caspofungin include its concentration-dependent fungicidal activity, hepatic metabolism, and dosing based on body surface area rather than weight with a recommended dose of 25 mg/m2/day.Citation7,Citation55,Citation90,Citation134 This medication is generally well tolerated with notable side effects of fever, rash, hypokalemia, and elevated transaminases.Citation90 However, caspofungin is not FDA approved for neonates younger than 3 months of age. Important features of anidulafungin include its concentration-dependent fungicidal activity, near complete protein binding, and unique metabolism, characterized by slow, non-enzymatic degradation to inactive metabolites.Citation90 A 2011 pharmacokinetic study of 15 infants and neonates administered 1.5 mg/kg/day IV anidulafungin found similar exposure levels in neonates as compared to children receiving similar weight-based dosing and adults receiving 100 mg/day; no serious adverse effects were reported.Citation135 A 2020 study of 19 patients between 1 month and 2 years of age receiving anidulafungin for 5–35 days found a 68.8% global response success rate to therapy.Citation136 Despite its tolerability with no reported serious adverse effects, there are few trials studying the efficacy of anidulafungin in neonates, and it is FDA approved for infants with candidemia starting at 1-month old.Citation90 A unique feature of rezafungin is its exceptionally long half-life, which allows for once-weekly dosing.Citation137 A 2023 multicenter phase 3 clinical trial in adults with candidemia or invasive candidiasis demonstrated that the 30-day all-cause mortality of patients treated with rezafungin was non-inferior to caspofungin.Citation138 Rezafungin has been approved by the FDA for the treatment of invasive candidiasis in adults with limited or no alternative treatment options, but more studies in neonates are still needed.Citation139

5-Flucytosine

5-Flucytosine 25 mg/kg four times daily is an alternative fungistatic agent recommended by the IDSA for CNS invasive candidiasis that does not respond to amphotericin B.Citation7,Citation96 This antimetabolite drug causes RNA miscoding, which interferes with DNA synthesis.Citation90 Important pharmacokinetic and pharmacodynamic properties include excellent oral bioavailability, very little protein binding, and high tissue penetration, including in the eye and CNS.Citation37 This drug is cleared by the kidneys and therefore requires renal adjustment, especially in premature infants, who can quickly accumulate high plasma concentrations as a result of low weight and renal immaturity.Citation37 Advantages of 5-flucytosine include broad antifungal activity against Candida species, with the exception of C. krusei,Citation37 and its aforementioned high eye and CNS penetration. In addition to required renal adjustment, a disadvantage of 5-flucytosine is its high rate of resistance if used alone; therefore, 5-flucytosine should be used in combination with other agents.Citation37 Another disadvantage is the drug’s availability only in an oral formulation. Common side effects include gastrointestinal symptoms, although bone marrow suppression and hepatotoxicity have been noted in adults taking high doses of 5-flucytosine.Citation140

Prevention

The most effective method for reducing the morbidity and mortality of invasive candidiasis in neonates is prevention of infection. The main prevention strategies include mitigation of well-known risk factors and thoughtful use of prophylactic antifungal regimens.

Mitigating Risk Factors

Risk factors for neonatal invasive candidiasis infections include prematurity, low birth weight, vaginal birth, skin or GI colonization at delivery, low Apgar scores, hospitalization longer than 7 days, compromise of GI mucosal integrity, presence of sepsis or shock, disseminated intravascular coagulation, and thrombocytopenia ().Citation1, While many of these are not easily modifiable, factors such as central line placement and utilization, use of groin catheters, mechanical ventilation, broad-spectrum antibiotics, H2 receptor blockers, systemic steroids, parenteral nutrition, and IV lipid emulsion are possible modifiable targets.Citation1,Citation4

Antibiotic stewardship is a particularly important intervention to consider. Broad spectrum antibiotic therapy kills the commensal bacteria that combat Candida proliferation.Citation8 An analysis of extremely low birth weight infants demonstrated a nearly 2-fold increased risk of invasive candidiasis when exposed to cephalosporins,Citation12 and carbapenems may also be associated with increased risk of invasive candidiasis in very low birth weight infants.Citation60 Judicious use of broad-spectrum antibiotics in neonates is a critical intervention to preclude the selection for Candida overgrowth and predisposition to infection that precedes invasive candidiasis.

Prophylaxis

Antifungal prophylaxis for high-risk neonates may prevent invasive candidiasis infection (). The 2016 IDSA guidelines recommend prophylaxis for neonates weighing less than 1 kg at birth in nurseries with high rates of invasive candidiasis, defined as greater than 10%.Citation7 The first-line regimen is IV or oral fluconazole 3–6 mg/kg twice weekly for six weeks.Citation7 If an alternative agent is necessary, the IDSA recommends oral nystatin 100,000 units three times daily for six weeks or oral bovine lactoferrin 100 mg daily.Citation7 The ESMID strongly recommends IV or oral fluconazole 3–6 mg/kg twice weekly for all neonates weighing less than 1 kg in NICUs with high frequency of Candida infections; moderately recommends the use of oral nystatin 100,000 units three times daily to decrease the Candida burden in the gastrointestinal tract; and suggests bovine lactoferrin 100 mg/day alone or in combination with a lactobacillus probiotic for neonates weighing less than 1.5 kg.Citation9,Citation91 The ESCMID also recommends against the use of miconazole gel due to concerns that it may increase fluconazole resistance.Citation9 Although not recommended by either organization, others advise the use of micafungin for antifungal prophylaxis in high-risk neonates when Candida auris colonization is suspected or NICU incidence is high.Citation30

Table 5 Prevention of Invasive Candidiasis in Neonates as Recommended by the IDSA and ESCMID

Fluconazole is the drug most recommended for prophylaxis against invasive candidiasis in neonates. There is data to support fluconazole prophylaxis in extremely low birth weight infants; a 2021 meta-analysis and systemic review indicated a significant decrease in invasive candidiasis mortality in extremely low birth weight infants on fluconazole prophylaxis compared to those not on prophylaxis.Citation141 A 2014 randomized, blinded, placebo-controlled trial of 361 infants from 32 different NICUs demonstrated a significantly lower incidence of invasive candidiasis in infants receiving 42 days of fluconazole prophylaxis compared to those receiving a placebo; however, there was not a significant difference in the composite primary end point of death or invasive candidiasis.Citation142 Other studies showed no difference in incidence of invasive fungal infectionsCitation143 or mortality in extremely low birth weight infants.Citation142 Fluconazole prophylaxis may not be particularly effective given the rise of triazole-resistant species; as epidemiology changes, attention will need to be paid to the rates of resistance, and consideration of alternative agents for prophylaxis may be necessary. Additionally, fluconazole prophylaxis has been associated with an increased minimal inhibitory concentration for Candida isolates that colonize infants who receive prophylaxis, potentially necessitating higher treatment doses and therefore increasing risk of drug toxicity.Citation144

Nystatin is also recommended in invasive candidiasis prophylaxis guidelines and has been studied in neonates for decades.Citation145 This medication’s oral formulation is not systemically absorbed; instead, it works against fungi in the GI tract, which is a major source of Candida colonization.Citation146 Oral nystatin is also less expensive than the IV formulation and is non-inferior for neonatal antifungal prophylaxis.Citation147 One prospective, open-labelled, randomized controlled trial in an Indonesian NICU demonstrated significantly lower incidence of fungal colonization, but not overall survival, in neonates who received oral nystatin prophylaxis as compared to those who did not.Citation146 The side effects and safety profile of nystatin have not been well studied in infants, and there are no blinded randomized control trials showing its benefit.

Another compound mentioned in prophylaxis guidelines is lactoferrin, a mammalian milk glycoprotein involved in innate immune host defenses.Citation148 This compound appears to be safe and well tolerated, but there is mixed data on its efficacy. Although some studies have determined that bovine lactoferrin, with or without a lactobacillus probiotic, reduces the incidence of sepsis in very low birth weight infants,Citation148 other studies, including a large, double-blind placebo trial, did not demonstrate beneficial impact on incidence of late-onset sepsis or necrotizing enterocolitis.Citation149

Probiotics are considered to minimize risk of necrotizing enterocolitis in neonates, but their role in fungal infections is not well characterized. Infants in the NICU are at risk for microbiome alterations through factors such as delivery via cesarean section, antimicrobial treatment, delayed enteral feeding, and prolonged hospitalizations.Citation150 There are mixed data on the efficacy of probiotics in the prevention of neonatal sepsis. A 2022 randomized intervention trial in extremely preterm infants determined that daily administration of a probiotic mixture accelerated the maturation of the neonatal microbiome, significantly decreased inflammatory cytokine stool burden, and resulted in a significant decrease in the relative abundance of Candida species.Citation151 A 2014 Cochrane review of over 5000 preterm infants given enteral probiotic supplementation determined that probiotics did not change incidence of nosocomial sepsis, while a 2017 systemic review found that the use of probiotics was beneficial for the prevention of late-onset sepsis in preterm infants.Citation152,Citation153 In 2021, the American Academy of Pediatrics stated that the routine use of probiotics in preterm infants was not recommended due to the lack of FDA regulated products, contradictory safety and efficacy data, and potential for harm in this vulnerable population.Citation154 In September 2023, the FDA cautioned that microorganisms contained in probiotics have been implicated as causes of serious neonatal bacterial and fungal infection, especially in preterm and low birthweight infants.Citation155–158 These recent changes may influence the clinical use of probiotics in the neonatal population in general.

Conclusion

Invasive candidiasis in neonates leads to substantial short- and long-term morbidity and mortality and poses a significant threat to both short- and long-term outcomes. Although infants with invasive candidiasis often present with non-specific symptoms, awareness of risk factors and utilization of high-yield diagnostic methodologies can identify affected neonates in a timely manner. Understanding the extent of disease involvement and prompt treatment with supportive measures as well as targeted systemic antifungal therapy can optimize short-term and long-term outcomes of affected infants. Prevention strategies, including risk factor mitigation and antifungal prophylaxis, should be considered both at an individual and unit level to reduce the incidence of invasive candidiasis and its effects on preterm infants, who are among the most vulnerable populations at risk for Candida infections.

Disclosure

RGG has received support from industry for research services from Provepharm Inc and Tellus Therapeutics, outside the submitted work. AB reports training grants from Eunice Kennedy Shriver National Institute of Child Health and Human Development (1T32HD094671) and research support from Thrasher Research Foundation, outside the submitted work. The authors report no other conflicts of interest in this work.

Acknowledgments

We would like to thank Carson Stahlsmith, PharmD, BCPPS (Department of Pharmacy, Duke University Hospital, Durham, North Carolina, USA) for reviewing the manuscript for accuracy of information in the therapeutics section, editing, revising, and correcting errors. We would like to thank Jordan Mah, MD (Department of Medicine, Division of Infectious Diseases, Durham, North Carolina, USA) for reviewing the laboratory testing section of the manuscript, editing, revising, and verifying information.

References

  • Arsenault AB, Bliss JM. Neonatal candidiasis: new insights into an old problem at a unique host-pathogen interface. Curr Fungal Infect Rep. 2015;9(4):246–252. doi:10.1007/s12281-015-0238-x
  • Rowen JL. Mucocutaneous candidiasis. Semin Perinatol. 2003;27(5):406–413. doi:10.1016/s0146-0005(03)00066-1
  • Benjamin DK, Stoll BJ, Gantz MG, et al. Neonatal candidiasis: epidemiology, risk factors, and clinical judgment. Pediatrics. 2010;126(4):e865–73. doi:10.1542/peds.2009-3412
  • Kilpatrick R, Scarrow E, Hornik C, Greenberg RG. Neonatal invasive candidiasis: updates on clinical management and prevention. Lancet Child Adolesc Health. 2022;6(1):60–70. doi:10.1016/s2352-4642(21)00272-8
  • Filippidi A, Galanakis E, Maraki S, et al. The effect of maternal flora on Candida colonisation in the neonate. Mycoses. 2014;57(1):43–48. doi:10.1111/myc.12100
  • Al-Rusan RM, Darwazeh AMG, Lataifeh IM. The relationship of Candida colonization of the oral and vaginal mucosae of mothers and oral mucosae of their newborns at birth. Oral Surg Oral Med Oral Pathol Oral Radiol. 2017;123(4):459–463. doi:10.1016/j.oooo.2017.01.003
  • Pappas PG, Kauffman CA, Andes DR, et al. Clinical practice guideline for the management of candidiasis: 2016 update by the infectious diseases Society of America. Clin Infect Dis. 2015;62(4):e1–e50. doi:10.1093/cid/civ933
  • Kelly MS, Benjamin DK, Smith PB. The epidemiology and diagnosis of invasive candidiasis among premature infants. Clin Perinatol. 2015;42(1):105–17, viii–ix. doi:10.1016/j.clp.2014.10.008
  • Vasileiou E, Apsemidou A, Vyzantiadis TA, Tragiannidis A. Invasive candidiasis and candidemia in pediatric and neonatal patients: a review of current guidelines. Curr Med Mycol. 2018;4(3):28–33. doi:10.18502/cmm.4.3.173
  • Alvarado-Socarras JL, Vargas-Soler JA, Franco-Paredes C, Villegas-Lamus KC, Rojas-Torres JP, Rodriguez-Morales AJ. A cluster of neonatal infections caused by Candida auris at a large referral center in Colombia. J Pediatric Infect Dis Soc. 2021;10(5):549–555. doi:10.1093/jpids/piaa152
  • Kaur H, Chakrabarti A. Strategies to reduce mortality in adult and neonatal candidemia in developing countries. J Fungi. 2017;3(3):41. doi:10.3390/jof3030041
  • Benjamin DK, Stoll BJ, Fanaroff AA, et al. Neonatal candidiasis among extremely low birth weight infants: risk factors, mortality rates, and neurodevelopmental outcomes at 18 to 22 months. Pediatrics. 2006;117(1):84–92. doi:10.1542/peds.2004-2292
  • Cotten CM, McDonald S, Stoll B, Goldberg RN, Poole K, Benjamin DK. The association of third-generation cephalosporin use and invasive candidiasis in extremely low birth-weight infants. Pediatrics. 2006;118(2):717–722. doi:10.1542/peds.2005-2677
  • Cole GT, Halawa AA, Anaissie EJ. The role of the gastrointestinal tract in hematogenous candidiasis: from the laboratory to the bedside. Clin Infect Dis. 1996;22 Suppl 2(Supplement_2):S73–88. doi:10.1093/clinids/22.supplement_2.s73
  • Cook A, Ferreras-Antolin L, Adhisivam B, et al. Neonatal invasive candidiasis in low- and middle-income countries: data from the NeoOBS study. Med Mycol. 2023;61(3). doi:10.1093/mmy/myad010
  • Aliaga S, Clark RH, Laughon M, et al. Changes in the incidence of candidiasis in neonatal intensive care units. Pediatrics. 2014;133(2):236–242. doi:10.1542/peds.2013-0671
  • Centers for Disease Control and Prevention. Invasive Candidiasis statistics; [ updated January 13, 2023]. Available from: https://www.cdc.gov/fungal/diseases/candidiasis/invasive/statistics.html. Accessed September 28, 2023.
  • Scamardo MS, Dolce P, Esposito EP, Raimondi F, Triassi M, Zarrilli R. Trends, risk factors and outcomes of healthcare-associated infections in a neonatal intensive care unit in Italy during 2013–2017. Ital J Pediatr. 2020;46(1):34. doi:10.1186/s13052-020-0799-3
  • Oeser C, Vergnano S, Naidoo R, et al. Neonatal invasive fungal infection in England 2004–2010. Clin Microbiol Infect. 2014;20(9):936–941. doi:10.1111/1469-0691.12578
  • Ting JY, Roberts A, Synnes A, et al. Invasive fungal infections in neonates in Canada: epidemiology and outcomes. Pediatr Infect Dis J. 2018;37(11):1154–1159. doi:10.1097/inf.0000000000001968
  • Howell A, Isaacs D, Halliday R. Oral nystatin prophylaxis and neonatal fungal infections. Arch Dis Child Fetal Neonatal Ed. 2009;94(6):F429–33. doi:10.1136/adc.2008.157123
  • Stoll BJ, Hansen NI, Adams-Chapman I, et al. Neurodevelopmental and growth impairment among extremely low-birth-weight infants with neonatal infection. JAMA. 2004;292(19):2357–2365. doi:10.1001/jama.292.19.2357
  • Greenberg RG, Benjamin DK, Gantz MG, et al. Empiric antifungal therapy and outcomes in extremely low birth weight infants with invasive candidiasis. J Pediatr. 2012;161(2):264–9.e2. doi:10.1016/j.jpeds.2012.01.053
  • Cahan H, Deville JG. Outcomes of neonatal candidiasis: the impact of delayed initiation of antifungal therapy. Int J Pediatr. 2011;2011:813871. doi:10.1155/2011/813871
  • Calley JL, Warris A. Recognition and diagnosis of invasive fungal infections in neonates. J Infect. 2017;74(Suppl 1):S108–s113. doi:10.1016/s0163-4453(17)30200-1
  • Walsh TJ, Katragkou A, Chen T, Salvatore CM, Roilides E. Invasive Candidiasis in infants and children: recent advances in epidemiology, diagnosis, and treatment. J Fungi. 2019;5(1):11. doi:10.3390/jof5010011
  • Benjamin DK, Poole C, Steinbach WJ, Rowen JL, Walsh TJ. Neonatal candidemia and end-organ damage: a critical appraisal of the literature using meta-analytic techniques. Pediatrics. 2003;112(3 Pt 1):634–640. doi:10.1542/peds.112.3.634
  • Lingappan A, Wykoff CC, Albini TA, et al. Endogenous fungal endophthalmitis: causative organisms, management strategies, and visual acuity outcomes. Am J Ophthalmol. 2012;153(1):162–6.e1. doi:10.1016/j.ajo.2011.06.020
  • Alhamoud MA, Alnosair GH, Alhashim HY. Neonatal endogenous endophthalmitis: a case report. Cureus. 2022;14(2):e22256. doi:10.7759/cureus.22256
  • Ferrando G, Castagnola E. Prophylaxis of invasive fungal infection in neonates: a narrative review for practical purposes. J Fungi. 2023;9(2):164. doi:10.3390/jof9020164
  • Goudjil S, Kongolo G, Dusol L, et al. (1-3)-β-D-glucan levels in candidiasis infections in the critically ill neonate. J Matern Fetal Neonatal Med. 2013;26(1):44–48. doi:10.3109/14767058.2012.722716
  • Horvath LL, Hospenthal DR, Murray CK, Dooley DP. Detection of simulated candidemia by the BACTEC 9240 system with plus aerobic/F and anaerobic/F blood culture bottles. J Clin Microbiol. 2003;41(10):4714–4717. doi:10.1128/jcm.41.10.4714-4717.2003
  • Horvath LL, George BJ, Murray CK, Harrison LS, Hospenthal DR. Direct comparison of the BACTEC 9240 and BacT/ALERT 3D automated blood culture systems for candida growth detection. J Clin Microbiol. 2004;42(1):115–118. doi:10.1128/jcm.42.1.115-118.2004
  • Telenti A, Roberts GD. Fungal blood cultures. Eur J Clin Microbiol Infect Dis. 1989;8(9):825–831. doi:10.1007/bf02185855
  • McDonald LC, Weinstein MP, Fune J, Mirrett S, Reimer LG, Reller LB. Controlled comparison of BacT/ALERT FAN aerobic medium and BATEC fungal blood culture medium for detection of fungemia. J Clin Microbiol. 2001;39(2):622–624. doi:10.1128/jcm.39.2.622-624.2001
  • Bersani I, Piersigilli F, Goffredo BM, et al. Antifungal drugs for invasive Candida Infections (ICI) in neonates: future perspectives. Front Pediatr. 2019;7:375. doi:10.3389/fped.2019.00375
  • Keighley C, Cooley L, Morris AJ, et al. Consensus guidelines for the diagnosis and management of invasive candidiasis in haematology, oncology and intensive care settings, 2021. Intern Med J. 2021;51(Suppl 7):89–117. doi:10.1111/imj.15589
  • Simor AE, Porter V, Mubareka S, 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(12). doi:10.1128/JCM.01387-18
  • Schelonka RL, Moser SA. Time to positive culture results in neonatal Candida septicemia. J Pediatr. 2003;142(5):564–565. doi:10.1067/mpd.2003.188
  • Clancy CJ, Nguyen MH. 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(9):1284–1292. doi:10.1093/cid/cit006
  • Oliveri S, Trovato L, Betta P, Romeo MG, Nicoletti G. Experience with the Platelia Candida ELISA for the diagnosis of invasive candidosis in neonatal patients. Clin Microbiol Infect. 2008;14(4):391–393. doi:10.1111/j.1469-0691.2007.01938.x
  • Lucignano B, Cento V, Agosta M, et al. Effective rapid diagnosis of bacterial and fungal bloodstream infections by T2 magnetic resonance technology in the pediatric population. J Clin Microbiol. 2022;60(10):e0029222. doi:10.1128/jcm.00292-22
  • Mylonakis E, Clancy CJ, Ostrosky-Zeichner L, et al. T2 magnetic resonance assay for the rapid diagnosis of candidemia in whole blood: a clinical trial. Clin Infect Dis. 2015;60(6):892–899. doi:10.1093/cid/ciu959
  • Ramos JT, Villar S, Bouza E, et al. Performance of a quantitative PCR-based assay and Beta-d-glucan detection for diagnosis of invasive Candidiasis in Very-Low-Birth-Weight Preterm Neonatal Patients (CANDINEO Study). J Clin Microbiol. 2017;55(9):2752–2764. doi:10.1128/JCM.00496-17
  • George BJ, Horvath LL, Hospenthal DR. Effect of inoculum size on detection of Candida growth by the BACTEC 9240 automated blood culture system using aerobic and anaerobic media. J Clin Microbiol. 2005;43(1):433–435. doi:10.1128/jcm.43.1.433-435.2005
  • Cohen-Wolkowiez M, Smith PB, Mangum B, et al. Neonatal Candida meningitis: significance of cerebrospinal fluid parameters and blood cultures. J Perinatol. 2007;27(2):97–100. doi:10.1038/sj.jp.7211628
  • Berenguer J, Buck M, Witebsky F, Stock F, Pizzo PA, Walsh TJ. Lysis-centrifugation blood cultures in the detection of tissue-proven invasive candidiasis. Disseminated versus single-organ infection. Diagn Microbiol Infect Dis. 1993;17(2):103–109. doi:10.1016/0732-8893(93)90020-8
  • Johnson MD, Lewis RE, Dodds Ashley ES, et al. Core recommendations for antifungal Stewardship: a statement of the mycoses study group education and research consortium. J Infect Dis. 2020;222(Suppl 3):S175–s198. doi:10.1093/infdis/jiaa394
  • Huber S, Hetzer B, Crazzolara R, Orth-Höller D. The correct blood volume for paediatric blood cultures: a conundrum? Clin Microbiol Infect. 2020;26(2):168–173. doi:10.1016/j.cmi.2019.10.006
  • Sundararajan S. Ideal blood inoculant volume for neonatal sepsis evaluation: an alternative approach. Pediatr Res. 2021;90(5):930–933. doi:10.1038/s41390-021-01720-2
  • Miller JM, Binnicker MJ, Campbell S, et al. A guide to utilization of the microbiology laboratory for diagnosis of infectious diseases: 2018 update by the infectious diseases society of America and the American Society for Microbiology. Clin Infect Dis. 2018;67(6):e1–e94. doi:10.1093/cid/ciy381
  • Howie SR. Blood sample volumes in child health research: review of safe limits. Bull World Health Organ. 2011;89(1):46–53. doi:10.2471/blt.10.080010
  • Lancaster DP, Friedman DF, Chiotos K, Sullivan KV, Carroll KC. Blood volume required for detection of low levels and ultralow levels of organisms responsible for neonatal bacteremia by use of Bactec Peds Plus/F, Plus Aerobic/F Medium, and the BD Bactec FX system: an in vitro study. J Clin Microbiol. 2015;53(11):3609–3613. doi:10.1128/jcm.01706-15
  • Harewood FC, Curtis N, Daley AJ, Bryant PA, Gwee A, Connell TG. Adequate or inadequate? The volume of blood submitted for blood culture at a tertiary children’s hospital. Clin Pediatr. 2018;57(11):1310–1317. doi:10.1177/0009922818778042
  • Ruhnke M, Rickerts V, Cornely OA, et al. Diagnosis and therapy of Candida infections: joint recommendations of the German speaking mycological society and the Paul-Ehrlich-Society for chemotherapy. Mycoses. 2011;54(4):279–310. doi:10.1111/j.1439-0507.2011.02040.x
  • Dulanto-Reinoso CM, Martinez-Castellanos MA. Detection of Candida endophthalmitis in a newborn using handheld spectral-domain optical coherence tomography. Case Rep Ophthalmol. 2018;9(3):439–443. doi:10.1159/000493679
  • Zhao D, Qiu G, Luo Z, Zhang Y, Simeoni U. Platelet parameters and (1, 3)-β-D-glucan as a diagnostic and prognostic marker of invasive fungal disease in preterm infants. PLoS One. 2015;10(4):e0123907. doi:10.1371/journal.pone.0123907
  • Manzoni P, Mostert M, Galletto P, et al. Is thrombocytopenia suggestive of organism-specific response in neonatal sepsis? Pediatr Int. 2009;51(2):206–210. doi:10.1111/j.1442-200X.2008.02689.x
  • Guida JD, Kunig AM, Leef KH, McKenzie SE, Paul DA. Platelet count and sepsis in very low birth weight neonates: is there an organism-specific response? Pediatrics. 2003;111(6 Pt 1):1411–1415. doi:10.1542/peds.111.6.1411
  • Benjamin DK, DeLong ER, Steinbach WJ, Cotton CM, Walsh TJ, Clark RH. Empirical therapy for neonatal candidemia in very low birth weight infants. Pediatrics. 2003;112(3 Pt 1):543–547. doi:10.1542/peds.112.3.543
  • Makhoul IR, Kassis I, Smolkin T, Tamir A, Sujov P. Review of 49 neonates with acquired fungal sepsis: further characterization. Pediatrics. 2001;107(1):61–66. doi:10.1542/peds.107.1.61
  • Cohen JF, Ouziel A, Matczak S, et al. Diagnostic accuracy of serum (1,3)-beta-d-glucan for neonatal invasive candidiasis: systematic review and meta-analysis. Clin Microbiol Infect. 2020;26(3):291–298. doi:10.1016/j.cmi.2019.09.010
  • Smith PB, Benjamin DK, Alexander BD, Johnson MD, Finkelman MA, Steinbach WJ. Quantification of 1,3-beta-D-glucan levels in children: preliminary data for diagnostic use of the beta-glucan assay in a pediatric setting. Clin Vaccine Immunol. 2007;14(7):924–925. doi:10.1128/cvi.00025-07
  • Lamoth F, Cruciani M, Mengoli C, et al. β-Glucan antigenemia assay for the diagnosis of invasive fungal infections in patients with hematological malignancies: a systematic review and meta-analysis of cohort studies from the Third European Conference on Infections in Leukemia (ECIL-3). Clin Infect Dis. 2012;54(5):633–643. doi:10.1093/cid/cir897
  • Digby J, Kalbfleisch J, Glenn A, Larsen A, Browder W, Williams D. Serum glucan levels are not specific for presence of fungal infections in intensive care unit patients. Clin Diagn Lab Immunol. 2003;10(5):882–885. doi:10.1128/cdli.10.5.882-885.2003
  • Racil Z, Kocmanova I, Lengerova M, et al. Difficulties in using 1,3-{beta}-D-glucan as the screening test for the early diagnosis of invasive fungal infections in patients with haematological malignancies--high frequency of false-positive results and their analysis. J Med Microbiol. 2010;59(Pt9):1016–1022. doi:10.1099/jmm.0.019299-0
  • Prattes J, Schilcher G, Krause R. Reliability of serum 1,3-beta-D-glucan assay in patients undergoing renal replacement therapy: a review of the literature. Mycoses. 2015;58(1):4–9. doi:10.1111/myc.12267
  • Wheat LJ. Approach to the diagnosis of invasive aspergillosis and candidiasis. Clin Chest Med. 2009;30(2):367–77, viii. doi:10.1016/j.ccm.2009.02.012
  • Cendejas-Bueno E, Falces-Romero I, Laplaza-González M, et al. Candidemia diagnosis with T2 nuclear magnetic resonance in a PICU: a new approach. Pediatr Crit Care Med. 2021;22(2):e109–e114. doi:10.1097/pcc.0000000000002548
  • Mylonakis E, Zacharioudakis IM, Clancy CJ, Nguyen MH, Pappas PG, Warnock DW. Efficacy of T2 magnetic resonance assay in monitoring Candidemia after initiation of antifungal therapy: the Serial Therapeutic and Antifungal Monitoring Protocol (STAMP) trial. J Clin Microbiol. 2018;56(4). doi:10.1128/JCM.01756-17
  • Clancy CJ, Pappas PG, Vazquez J, 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(11):1678–1686. doi:10.1093/cid/cix1095
  • Tan KE, Ellis BC, Lee R, Stamper PD, Zhang SX, Carroll KC. Prospective evaluation of a matrix-assisted laser desorption ionization-time of flight mass spectrometry system in a hospital clinical microbiology laboratory for identification of bacteria and yeasts: a bench-by-bench study for assessing the impact on time to identification and cost-effectiveness. J Clin Microbiol. 2012;50(10):3301–3308. doi:10.1128/jcm.01405-12
  • Sow D, Fall B, Ndiaye M, et al. Usefulness of MALDI-TOF mass spectrometry for routine identification of Candida Species in a resource-poor setting. Mycopathologia. 2015;180(3–4):173–179. doi:10.1007/s11046-015-9905-2
  • Balows A. Manual of clinical microbiology 8th edition: p. R. Murray, E. J. Baron, J. H. Jorgenson, M. A. Pfaller, and R. H. Yolken, eds., ASM Press, 2003, 2113 pages, 2 vol, 2003 + subject & author indices, ISBN: 1-555810255-4, US$ 189.95. Book Reviews. Diagn Microbiol Infect Dis. 2003;47(4):625–626. doi:10.1016/S0732-8893(03)00160-3
  • Fernandez J, Erstad BL, Petty W, Nix DE. Time to positive culture and identification for Candida blood stream infections. Diagn Microbiol Infect Dis. 2009;64(4):402–407. doi:10.1016/j.diagmicrobio.2009.04.002
  • Spanu T, Posteraro B, Fiori B, et al. Direct maldi-tof mass spectrometry assay of blood culture broths for rapid identification of Candida species causing bloodstream infections: an observational study in two large microbiology laboratories. J Clin Microbiol. 2012;50(1):176–179. doi:10.1128/jcm.05742-11
  • Huang AM, Newton D, Kunapuli A, et al. Impact of rapid organism identification via matrix-assisted laser desorption/ionization time-of-flight combined with antimicrobial stewardship team intervention in adult patients with bacteremia and candidemia. Clin Infect Dis. 2013;57(9):1237–1245. doi:10.1093/cid/cit498
  • Harris DM, Hata DJ. Rapid identification of bacteria and Candida using PNA-FISH from blood and peritoneal fluid cultures: a retrospective clinical study. Ann Clin Microbiol Antimicrob. 2013;12(1):2. doi:10.1186/1476-0711-12-2
  • 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. 2018;61(3):179–185. doi:10.1111/myc.12722
  • Lau A, Halliday C, Chen SC, Playford EG, Stanley K, Sorrell TC. Comparison of whole blood, serum, and plasma for early detection of candidemia by multiplex-tandem PCR. J Clin Microbiol. 2010;48(3):811–816. doi:10.1128/jcm.01650-09
  • Aittakorpi A, Kuusela P, Koukila-Kähkölä P, et al. Accurate and rapid identification of Candida spp. frequently associated with fungemia by using PCR and the microarray-based prove-it sepsis assay. J Clin Microbiol. 2012;50(11):3635–3640. doi:10.1128/jcm.01461-12
  • Kidd SE, Chen SC, Meyer W, Halliday CL. A new age in molecular diagnostics for invasive fungal disease: are we ready? Front Microbiol. 2019;10:2903. doi:10.3389/fmicb.2019.02903
  • He B, Yang Q. Updates in laboratory identification of invasive fungal infection in neonates. Microorganisms. 2023;11(4):1001.
  • Yang Q, He B, Chen C, et al. A rapid, visible, and highly sensitive method for recognizing and distinguishing invasive fungal infections via CCP-FRET technology. ACS Infect Dis. 2021;7(10):2816–2825. doi:10.1021/acsinfecdis.1c00393
  • Graff K, Dominguez SR, Messacar K. Metagenomic next-generation sequencing for diagnosis of pediatric meningitis and encephalitis: a review. J Pediatric Infect Dis Soc. 2021;10(Supplement_4):S78–s87. doi:10.1093/jpids/piab067
  • Okumura T, Horiba K, Tetsuka N, et al. Next-generation sequencing-based detection of Ureaplasma in the gastric fluid of neonates with respiratory distress and chorioamnionitis. J Matern Fetal Neonatal Med. 2023;36(1):2207113. doi:10.1080/14767058.2023.2207113
  • Agudelo-Pérez S, Fernández-Sarmiento J, Rivera León D, Peláez RG. Metagenomics by next-generation sequencing (mNGS) in the etiological characterization of neonatal and pediatric sepsis: a systematic review. Front Pediatr. 2023;11:1011723. doi:10.3389/fped.2023.1011723
  • Hill JA, Dalai SC, Hong DK, et al. Liquid Biopsy for invasive mold infections in hematopoietic cell transplant recipients with pneumonia through next-generation sequencing of microbial cell-free DNA in plasma. Clin Infect Dis. 2021;73(11):e3876–e3883. doi:10.1093/cid/ciaa1639
  • Heldman MR, Ahmed AA, Liu W, et al. Serial quantitation of plasma microbial cell-free DNA before and after diagnosis of pulmonary invasive mold infections in hematopoietic cell transplant recipients. J Infect Dis. 2023. doi:10.1093/infdis/jiad255
  • Autmizguine J, Guptill JT, Cohen-Wolkowiez M, Benjamin DK, Capparelli EV. Pharmacokinetics and pharmacodynamics of antifungals in children: clinical implications. Drugs. 2014;74(8):891–909. doi:10.1007/s40265-014-0227-3
  • Hope WW, Castagnola E, Groll AH, et al. ESCMID* guideline for the diagnosis and management of Candida diseases 2012: prevention and management of invasive infections in neonates and children caused by Candida spp. Clin Microbiol Infect. 2012;18(Suppl 7):38–52. doi:10.1111/1469-0691.12040
  • Wade KC, Wu D, Kaufman DA, et al. Population pharmacokinetics of fluconazole in young infants. Antimicrob Agents Chemother. 2008;52(11):4043–4049. doi:10.1128/aac.00569-08
  • Watt K, Benjamin DK, Cohen-Wolkowiez M. Pharmacokinetics of antifungal agents in children. Early Hum Dev. 2011;87(Suppl 1):S61–5. doi:10.1016/j.earlhumdev.2011.01.014
  • Piper L, Smith PB, Hornik CP, et al. Fluconazole loading dose pharmacokinetics and safety in infants. Pediatr Infect Dis J. 2011;30(5):375–378. doi:10.1097/INF.0b013e318202cbb3
  • Pammi M. Treatment of Candida Infection in Neonates. Post T, editor. UpToDate; 2023.
  • Bellmann R, Smuszkiewicz P. Pharmacokinetics of antifungal drugs: practical implications for optimized treatment of patients. Infection. 2017;45(6):737–779. doi:10.1007/s15010-017-1042-z
  • Clancy CJ, Nguyen MH. Emergence of Candida auris: an international call to arms. Clin Infect Dis. 2017;64(2):141–143. doi:10.1093/cid/ciw696
  • Fairuz S, Nair RS, Billa N. Orally administered amphotericin B nanoformulations: physical properties of nanoparticle carriers on bioavailability and clinical relevance. Pharmaceutics. 2022;14(9):1823. doi:10.3390/pharmaceutics14091823
  • Bekersky I, Fielding RM, Dressler DE, Lee JW, Buell DN, Walsh TJ. Pharmacokinetics, excretion, and mass balance of liposomal amphotericin B (AmBisome) and amphotericin B deoxycholate in humans. Antimicrob Agents Chemother. 2002;46(3):828–833. doi:10.1128/aac.46.3.828-833.2002
  • Baley JE, Meyers C, Kliegman RM, Jacobs MR, Blumer JL. Pharmacokinetics, outcome of treatment, and toxic effects of amphotericin B and 5-fluorocytosine in neonates. J Pediatr. 1990;116(5):791–797. doi:10.1016/s0022-3476(05)82674-5
  • Benson JM, Nahata MC. Pharmacokinetics of amphotericin B in children. Antimicrob Agents Chemother. 1989;33(11):1989–1993. doi:10.1128/aac.33.11.1989
  • Koren G, Lau A, Klein J, et al. Pharmacokinetics and adverse effects of amphotericin B in infants and children. J Pediatr. 1988;113(3):559–563. doi:10.1016/s0022-3476(88)80653-x
  • Nath CE, McLachlan AJ, Shaw PJ, Gunning R, Earl JW. Population pharmacokinetics of amphotericin B in children with malignant diseases. Br J Clin Pharmacol. 2001;52(6):671–680. doi:10.1046/j.0306-5251.2001.01496.x
  • Starke JR, Mason EO, Kramer WG, Kaplan SL. Pharmacokinetics of amphotericin B in infants and children. J Infect Dis. 1987;155(4):766–774. doi:10.1093/infdis/155.4.766
  • Lestner JM, Smith PB, Cohen-Wolkowiez M, Benjamin DK, Hope WW. Antifungal agents and therapy for infants and children with invasive fungal infections: a pharmacological perspective. Br J Clin Pharmacol. 2013;75(6):1381–1395. doi:10.1111/bcp.12025
  • Andrew EC, Curtis N, Coghlan B, Cranswick N, Gwee A. Adverse effects of amphotericin B in children; a retrospective comparison of conventional and liposomal formulations. Br J Clin Pharmacol. 2018;84(5):1006–1012. doi:10.1111/bcp.13521
  • Butler KM, Rench MA, Baker CJ. Amphotericin B as a single agent in the treatment of systemic candidiasis in neonates. Pediatr Infect Dis J. 1990;9(1):51–56. doi:10.1097/00006454-199001000-00012
  • Linder N, Klinger G, Shalit I, et al. Treatment of candidaemia in premature infants: comparison of three amphotericin B preparations. J Antimicrob Chemother. 2003;52(4):663–667. doi:10.1093/jac/dkg419
  • Le J, Adler-Shohet FC, Nguyen C, Lieberman JM. Nephrotoxicity associated with amphotericin B deoxycholate in neonates. Pediatr Infect Dis J. 2009;28(12):1061–1063. doi:10.1097/INF.0b013e3181af6201
  • Holler B, Omar SA, Farid MD, Patterson MJ. Effects of fluid and electrolyte management on amphotericin B-induced nephrotoxicity among extremely low birth weight infants. Pediatrics. 2004;113(6):e608–16. doi:10.1542/peds.113.6.e608
  • Jacobs SE, Zagaliotis P, Walsh TJ. Novel antifungal agents in clinical trials. F1000Res. 2021;10:507. doi:10.12688/f1000research.28327.2
  • Hornik CD, Bondi DS, Greene NM, Cober MP, John B. Review of fluconazole treatment and prophylaxis for invasive candidiasis in neonates. J Pediatr Pharmacol Ther. 2021;26(2):115–122. doi:10.5863/1551-6776-26.2.115
  • Chen S, Sun KY, Feng XW, Ran X, Lama J, Ran YP. Efficacy and safety of itraconazole use in infants. World J Pediatr. 2016;12(4):399–407. doi:10.1007/s12519-016-0034-x
  • Mondal RK, Singhi SC, Chakrabarti A, J M. Randomized comparison between fluconazole and itraconazole for the treatment of candidemia in a pediatric intensive care unit: a preliminary study. Pediatr Crit Care Med. 2004;5(6):561–565. doi:10.1097/01.Pcc.0000144712.29127.81
  • Zimmermann T, Yeates RA, Laufen H, Pfaff G, Wildfeuer A. Influence of concomitant food intake on the oral absorption of two triazole antifungal agents, itraconazole and fluconazole. Eur J Clin Pharmacol. 1994;46(2):147–150. doi:10.1007/bf00199879
  • Watt K, Manzoni P, Cohen-Wolkowiez M, et al. Triazole use in the nursery: fluconazole, voriconazole, posaconazole, and ravuconazole. Curr Drug Metab. 2013;14(2):193–202.
  • Shima H, Miharu M, Osumi T, Takahashi T, Shimada H. Differences in voriconazole trough plasma concentrations per oral dosages between children younger and older than 3 years of age. Pediatr Blood Cancer. 2010;54(7):1050–1052. doi:10.1002/pbc.22451
  • Roberts JK, Stockmann C, Constance JE, et al. Pharmacokinetics and pharmacodynamics of antibacterials, antifungals, and antivirals used most frequently in neonates and infants. Clin Pharmacokinet. 2014;53(7):581–610. doi:10.1007/s40262-014-0147-0
  • Frankenbusch K, Eifinger F, Kribs A, Rengelshauseu J, Roth B. Severe primary cutaneous aspergillosis refractory to amphotericin B and the successful treatment with systemic voriconazole in two premature infants with extremely low birth weight. J Perinatol. 2006;26(8):511–514. doi:10.1038/sj.jp.7211532
  • Muldrew KM, Maples HD, Stowe CD, Jacobs RF. Intravenous voriconazole therapy in a preterm infant. Pharmacotherapy. 2005;25(6):893–898. doi:10.1592/phco.2005.25.6.893
  • Krishnan T, Rishi P. Management of a case of Candida endogenous endophthalmitis in a neonate. Ocul Immunol Inflamm. 2014;22(1):77–78. doi:10.3109/09273948.2013.791923
  • Turner RB, Martello JL, Malhotra A. Worsening renal function in patients with baseline renal impairment treated with intravenous voriconazole: a systematic review. Int J Antimicrob Agents. 2015;46(4):362–366. doi:10.1016/j.ijantimicag.2015.05.023
  • Stemler J, Mellinghoff SC, Khodamoradi Y, et al. Primary prophylaxis of invasive fungal diseases in patients with haematological malignancies: 2022 update of the recommendations of the Infectious Diseases Working Party (AGIHO) of the German Society for Haematology and Medical Oncology (DGHO). J Antimicrob Chemother. 2023;13. doi:10.1093/jac/dkad143
  • Ashbee HR, Barnes RA, Johnson EM, Richardson MD, Gorton R, Hope WW. Therapeutic drug monitoring (TDM) of antifungal agents: guidelines from the British Society for Medical Mycology. J Antimicrob Chemother. 2014;69(5):1162–1176. doi:10.1093/jac/dkt508
  • Treviño-Rangel RJ, González GM, Montoya AM, Rojas OC, Elizondo-Zertuche M, Álvarez-Villalobos NA. Recent antifungal pipeline developments against Candida auris: a systematic review. J Fungi. 2022;8(11):1144. doi:10.3390/jof8111144
  • Wiederhold NP. Pharmacodynamics, mechanisms of action and resistance, and spectrum of activity of new antifungal agents. J Fungi. 2022;8(8):857. doi:10.3390/jof8080857
  • Lyman M, Forsberg K, Reuben J, et al. Notes from the field: transmission of pan-resistant and echinocandin-resistant candida auris in health care facilities - Texas and the district of Columbia, January-April 2021. MMWR Morb Mortal Wkly Rep. 2021;70(29):1022–1023. doi:10.15585/mmwr.mm7029a2
  • Szekely J, Rakchang W, Rattanaphan P, Kositpantawong N. Fluconazole and echinocandin resistance of Candida species in invasive candidiasis at a university hospital during pre-COVID-19 and the COVID-19 outbreak. Epidemiol Infect. 2023;151:e146. doi:10.1017/s0950268823001346
  • Hope WW, Seibel NL, Schwartz CL, et al. Population pharmacokinetics of micafungin in pediatric patients and implications for antifungal dosing. Antimicrob Agents Chemother. 2007;51(10):3714–3719. doi:10.1128/aac.00398-07
  • Hope WW, Mickiene D, Petraitis V, et al. The pharmacokinetics and pharmacodynamics of micafungin in experimental hematogenous Candida meningoencephalitis: implications for echinocandin therapy in neonates. J Infect Dis. 2008;197(1):163–171. doi:10.1086/524063
  • Benjamin DK, Kaufman DA, Hope WW, et al. A Phase 3 study of Micafungin versus Amphotericin B deoxycholate in infants with invasive candidiasis. Pediatr Infect Dis J. 2018;37(10):992–998. doi:10.1097/inf.0000000000001996
  • Queiroz-Telles F, Berezin E, Leverger G, et al. Micafungin versus liposomal amphotericin B for pediatric patients with invasive candidiasis: substudy of a randomized double-blind trial. Pediatr Infect Dis J. 2008;27(9):820–826. doi:10.1097/INF.0b013e31817275e6
  • Arrieta AC, Maddison P, Groll AH. Safety of micafungin in pediatric clinical trials. Pediatr Infect Dis J. 2011;30(6):e97–e102. doi:10.1097/INF.0b013e3182127eaf
  • Walsh TJ, Adamson PC, Seibel NL, et al. Pharmacokinetics, safety, and tolerability of caspofungin in children and adolescents. Antimicrob Agents Chemother. 2005;49(11):4536–4545. doi:10.1128/aac.49.11.4536-4545.2005
  • Cohen-Wolkowiez M, Benjamin DK, Piper L, et al. Safety and pharmacokinetics of multiple-dose anidulafungin in infants and neonates. Clin Pharmacol Ther. 2011;89(5):702–707. doi:10.1038/clpt.2011.26
  • Roilides E, Carlesse F, Tawadrous M, et al. Safety, efficacy and pharmacokinetics of anidulafungin in patients 1 month to <2 years of age with invasive candidiasis, including candidemia. Pediatr Infect Dis J. 2020;39(4):305–309. doi:10.1097/inf.0000000000002568
  • Ham YY, Lewis JS, Thompson GR. Rezafungin: a novel antifungal for the treatment of invasive candidiasis. Future Microbiol. 2021;16(1):27–36. doi:10.2217/fmb-2020-0217
  • Thompson GR, Soriano A, Cornely OA, et al. Rezafungin versus caspofungin for treatment of candidaemia and invasive candidiasis (ReSTORE): a multicentre, double-blind, double-dummy, randomised phase 3 trial. Lancet. 2023;401(10370):49–59. doi:10.1016/s0140-6736(22)02324-8
  • Syed YY. Rezafungin: first approval. Drugs. 2023;83(9):833–840. doi:10.1007/s40265-023-01891-8
  • Sigera LSM, Denning DW. Flucytosine and its clinical usage. Ther Adv Infect Dis. 2023;10:20499361231161387. doi:10.1177/20499361231161387
  • Robati Anaraki M, Nouri-Vaskeh M, Abdoli Oskoei S. Fluconazole prophylaxis against invasive candidiasis in very low and extremely low birth weight preterm neonates: a systematic review and meta-analysis. Clin Exp Pediatr. 2021;64(4):172–179. doi:10.3345/cep.2019.01431
  • Benjamin DK, Hudak ML, Duara S, et al. Effect of fluconazole prophylaxis on candidiasis and mortality in premature infants: a randomized clinical trial. JAMA. 2014;311(17):1742–1749. doi:10.1001/jama.2014.2624
  • Lee J, Kim HS, Shin SH, et al. Efficacy and safety of fluconazole prophylaxis in extremely low birth weight infants: multicenter pre-post cohort study. BMC Pediatr. 2016;16(1):67. doi:10.1186/s12887-016-0605-y
  • Autmizguine J, Smith PB, Prather K, et al. Effect of fluconazole prophylaxis on Candida fluconazole susceptibility in premature infants. J Antimicrob Chemother. 2018;73(12):3482–3487. doi:10.1093/jac/dky353
  • Sims ME, Yoo Y, You H, Salminen C, Walther FJ. Prophylactic oral nystatin and fungal infections in very-low-birthweight infants. Am J Perinatol. 1988;5(1):33–36. doi:10.1055/s-2007-999649
  • Rundjan L, Wahyuningsih R, Oeswadi CA, Marsogi M, Purnamasari A. Oral nystatin prophylaxis to prevent systemic fungal infection in very low birth weight preterm infants: a randomized controlled trial. BMC Pediatr. 2020;20(1):170. doi:10.1186/s12887-020-02074-0
  • Mersal A, Alzahrani I, Azzouz M, et al. Oral nystatin versus intravenous fluconazole as neonatal antifungal prophylaxis: non-inferiority trial. J Clin Neonatol. 2013;2(2):88–92. doi:10.4103/2249-4847.116408
  • Manzoni P, Rinaldi M, Cattani S, et al. Bovine lactoferrin supplementation for prevention of late-onset sepsis in very low-birth-weight neonates: a randomized trial. JAMA. 2009;302(13):1421–1428. doi:10.1001/jama.2009.1403
  • Berrington JE, McGuire W, Embleton ND. ELFIN, the United Kingdom preterm lactoferrin trial: interpretation and future questions (1). Biochem Cell Biol. 2021;99(1):1–6. doi:10.1139/bcb-2020-0073
  • Laforest-Lapointe I, Arrieta MC. Patterns of early-life gut microbial colonization during human immune development: an ecological perspective. Front Immunol. 2017;8:788. doi:10.3389/fimmu.2017.00788
  • Samara J, Moossavi S, Alshaikh B, et al. Supplementation with a probiotic mixture accelerates gut microbiome maturation and reduces intestinal inflammation in extremely preterm infants. Cell Host Microbe. 2022;30(5):696–711.e5. doi:10.1016/j.chom.2022.04.005
  • AlFaleh K, Anabrees J. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev. 2014;4:Cd005496. doi:10.1002/14651858.CD005496.pub4
  • Dermyshi E, Wang Y, Yan C, et al. The “Golden Age” of probiotics: a systematic review and meta-analysis of randomized and observational studies in preterm infants. Neonatology. 2017;112(1):9–23. doi:10.1159/000454668
  • Poindexter B, Cummings J, Hand I. Use of probiotics in preterm infants. Pediatrics. 2021;147(6). doi:10.1542/peds.2021-051485
  • Esaiassen E, Cavanagh P, Hjerde E, Simonsen GS, Støen R, Klingenberg C. Bifidobacterium longum subspecies infantis bacteremia in 3 extremely preterm infants receiving probiotics. Emerg Infect Dis. 2016;22(9):1664–1666. doi:10.3201/eid2209.160033
  • Bertelli C, Pillonel T, Torregrossa A, et al. Bifidobacterium longum bacteremia in preterm infants receiving probiotics. Clin Infect Dis. 2015;60(6):924–927. doi:10.1093/cid/ciu946
  • Roy U, Jessani LG, Rudramurthy SM, et al. Seven cases of Saccharomyces fungaemia related to use of probiotics. Mycoses. 2017;60(6):375–380. doi:10.1111/myc.12604
  • Pillai A, Tan J, Paquette V, Panczuk J. Does probiotic bacteremia in premature infants impact clinically relevant outcomes? A case report and updated review of literature. Clin Nutr ESPEN. 2020;39:255–259. doi:10.1016/j.clnesp.2020.05.020