Next Article in Journal
Galectin-1 Attenuates PDGF-Mediated AKT Signaling in Retinal Pigment Epithelial Cells
Previous Article in Journal
Interferon-Induced Transmembrane Protein 1 (IFITM1) Is Downregulated in Neurofibromatosis Type 1-Associated Malignant Peripheral Nerve Sheath Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neonatal Fungemia by Non-Candida Rare Opportunistic Yeasts: A Systematic Review of Literature

by
Alexandra Mpakosi
1,*,
Vasileios Cholevas
2,
Joseph Meletiadis
3,
Martha Theodoraki
4 and
Rozeta Sokou
4,5,*
1
Department of Microbiology, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece
2
School of Medicine, University of Bologna, 40125 Bologna, Italy
3
Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
4
Neonatal Intensive Care Unit, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece
5
Neonatal Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(17), 9266; https://doi.org/10.3390/ijms25179266
Submission received: 6 July 2024 / Revised: 23 August 2024 / Accepted: 24 August 2024 / Published: 27 August 2024
(This article belongs to the Special Issue Molecular Mechanisms and Pathophysiology of Sepsis)

Abstract

:
Fungal colonization poses a significant risk for neonates, leading to invasive infections such as fungemia. While Candida species are the most commonly identified pathogens, other rare yeasts are increasingly reported, complicating diagnosis and treatment due to limited data on antifungal pharmacokinetics. These emerging yeasts, often opportunistic, underscore the critical need for early diagnosis and targeted therapy in neonates. This systematic review aims to comprehensively analyze all published cases of neonatal fungemia caused by rare opportunistic yeasts, examining geographical distribution, species involved, risk factors, treatment approaches, and outcomes. Searching two databases (PubMed and SCOPUS), 89 relevant studies with a total of 342 cases were identified in the 42-year period; 62% of the cases occurred in Asia. Pichia anomala (31%), Kodamaea ohmeri (16%) and Malassezia furfur (15%) dominated. Low birth weight, the use of central catheters, prematurity, and the use of antibiotics were the main risk factors (98%, 76%, 66%, and 65%, respectively). 22% of the cases had a fatal outcome (80% in Asia). The highest mortality rates were reported in Trichosporon beigelii and Trichosporon asahii cases, followed by Dirkmeia churashimamensis cases (80%, 71%, and 42% respectively). Low birth weight, the use of central catheters, the use of antibiotics, and prematurity were the main risk factors in fatal cases (84%, 74%, 70%, and 67%, respectively). 38% of the neonates received fluconazole for treatment but 46% of them, died. Moreover, the rare yeasts of this review showed high MICs to fluconazole and this should be taken into account when planning prophylactic or therapeutic strategies with this drug. In conclusion, neonatal fungemia by rare yeasts is a life-threatening and difficult-to-treat infection, often underestimated and misdiagnosed.

1. Introduction

In recent years, there have been increasing cases of invasive fungal infections caused by rare yeasts, which comprise non-Candida and non-Cryptococcus species. These yeasts, although ubiquitous in the environment, have been defined as rare because they are almost never associated with human infections. However, these emerging species are increasingly harming human health, alarming the global scientific community. In 2022, recognizing the seriousness of this global health issue, the World Health Organization (WHO) introduced its first list of fungal “priority pathogens”: https://iris.who.int/bitstream/handle/10665/363682/9789240060241-eng.pdf, access on 31 July 2024. This important initiative highlights the urgent need for policy reforms and enhanced research efforts, especially in understanding the spread of fungal diseases, emerging patterns of antifungal resistance, and identifying the populations most at risk for these infections. A significant concern in the management of these infections is the increasing resistance to existing antifungal therapies. This trend complicates treatment and highlights the need for new and more effective antifungal agents. Additionally, there is growing recognition of the role of fungal infections in worsening outcomes, particularly in critically ill and immunocompromised patients, including neonates.
Neonates, especially premature infants, are vulnerable organisms, easily colonized by fungi due to their immature immune system, poor skin, insufficient mucosal barriers, and multiple risk factors favoring fungal prevalence against normal flora.
Invasive fungal infections (IFIs) are a significant contributor to morbidity and mortality among neonates in neonatal intensive care units (NICUs), particularly affecting preterm infants and those with very low birth weight (VLBW, <1500 g birth weight [BW]) [1]. These neonates are especially vulnerable due to their compromised immune systems, exposure to broad-spectrum antibiotics, immature epithelial barriers, and frequent need for invasive procedures, which collectively heighten their risk for opportunistic fungal infections. The bulk of IFIs are attributed to Candida species [2]. Notably, invasive candidiasis (IC) ranks as the third leading cause of late-onset sepsis in VLBW neonates and is a major factor in morbidity and mortality within the NICU [3,4,5,6]. The prevalence of IC in NICUs varies widely, ranging from 0.5% to 20%, and is significantly influenced by the specific center and patient demographics. It is inversely related to gestational age and birth weight, with the highest rates (5–20%) observed among neonates with extremely low birth weight (ELBW) [2,3]. Recently, a global change in the epidemiology of candidemia has been observed, marked by the emergence of resistant non-albicans Candida species. Among these, C. auris has been spreading globally, causing outbreaks in hospitals among both pediatric and adult patients, particularly in ICUs, and has been highlighted in the CDC’s report on urgent threats https://www.cdc.gov/antimicrobial-resistance/?CDC_AAref_Val=https://www.cdc.gov/DrugResistance/Biggest-Threats, access on 30 July 2024.
Fungal colonization is an important risk factor facilitating invasion, dissemination and eventually causing fungemia. Although Candida is the most commonly identified species of fungemia in neonates, other yeasts are also sporadically reported worldwide raising concerns due to difficulties in identification, limited pharmacokinetic data of the available antifungals, and lack of sufficient clinical and microbiological information. These rare yeasts are emerging, opportunistic pathogens that may lead to underdiagnosis of fungemia, delayed initiation of antifungal therapy, and often have fatal outcomes [4]. Therefore, an early and specific diagnosis and targeted treatment are critical, especially for neonates. To the best of our knowledge, there are no previous reviews of neonatal fungemia cases by rare opportunistic yeasts. The present study aims to provide a systematic review of all neonatal fungemia cases by rare opportunistic yeasts published in the literature.

2. Methods

A research methodological protocol was developed based on the PRISMA guidelines (presented as a Supplementary Material) [7] to collect and evaluate studies related to the subject under investigation. This protocol is registered in the PROSPERO database (CRD 42024561133).

2.1. Formulation of the Research Question

In order to clearly formulate the research question and facilitate a literature review, the PICO (Population, Intervention, Comparison, Outcome) framework was used:
  • Population: Neonates
  • Intervention: Rare opportunistic yeasts
  • Comparison: Not required for this review
  • Outcome: Cases of neonatal fungemia

2.2. Development of the Review Protocol

Research question: To conduct a review of all cases of neonatal fungemia from non-Candida rare opportunistic yeasts that have been published in the international literature.
Inclusion Criteria:
  • Case reports and case series studies reporting on cases of neonatal fungemia from non-Candida rare opportunistic yeasts.
  • Study designs focused on non-Candida rare opportunistic yeast infections, mainly defined as positive cultures from blood in neonates.
  • Exclusion Criteria:
  • Studies referring to other forms of fungal infections.
  • Cases not involving neonates.
  • Duplicate publications of the same cases.
  • Review articles, systematic reviews, and meta-analyses. Conference proceedings will be excluded.
  • Neonatal fungemia by Candida species
Main Outcomes:
  • Clinical presentation of non-Candida rare yeasts infections in neonates
  • Risk factors and characteristics of the affected population
  • Outcomes of the infections, including mortality

2.3. Search Strategy-Data Sources

The systematic literature review was conducted from June 2024 to July 2024. A systematic review of the existing literature was performed by searching the electronic databases PubMed and Scopus, with a final search date of 1 July 2024. The literature review was conducted using the following keywords: “neonatal fungemia”, “rare yeasts”, “non-Candida species”, “fungal infections”, “fungemia”, “fungaemia”, “neonates”, “preterm infants”, “preterm neonate*”, “infant*”, “newborn*”, “case report”, “case series”, “case”, in combination with Boolean operators (AND, OR). Additionally, to minimize the risk of missing studies and fully cover the extent of available literature, a manual electronic search and review of the references of each selected study, as well as references from previous systematic reviews in the same research field, was conducted.

2.4. Conflict Resolution

Screening, data extraction, and quality assessment were conducted independently by two researchers (AM and RS), with conflicts resolved through discussion and consensus between the researchers or, if necessary, by a third researcher (JM).

2.5. Data Synthesis and Presentation

Data were recorded in tabular form according to the following criteria: type of opportunistic yeasts, day of life when the infection manifested, comorbidities, risk factors, prematurity, administration of total parenteral nutrition, placement of central catheters, mechanical ventilation, and prior administration of broad-spectrum antibiotics, number of participants, number of events, and other relevant criteria for grouping the study population (subpopulations of neonates: preterm neonates, very low birth weight neonates, cardiac surgery or other surgical cases, neonates with congenital anomalies, etc.), year of publication.

3. Results

From the electronic database search, a total of 1990 studies were retrieved. Among these, 142 duplicate entries were identified and subsequently removed using the duplicate removal tool in the reference management software (EndNote X8). After carefully reading the titles and abstracts of the remaining studies, 1704 studies were excluded either because their subject matter did not serve the purpose of the review or because they met some of the exclusion criteria already visible at the title and abstract level. The careful reading of the full text of the 144 remaining studies revealed that only 89 studies met all the inclusion criteria and were included in this review. In total, 342 cases of neonates with rare opportunistic yeast infections were recorded. The flow diagram is presented in Figure 1.

3.1. Species Taxonomy and Geographical Distribution

The first report of neonatal fungemia from rare non-Candida yeasts was in 1981. Since then, there have been several cases and studies examining the incidence, diagnosis, and treatment of these infections. Over the past 42 years, 342 cases of neonatal fungemia caused by rare non-Candida yeasts have been reported. (Table 1 and Table 2, Figure 2)
Regarding the geographical distribution of all reported cases, 221 (65%) were from Asia, 55 (16%) from Europe, 48 (14%) from the USA, 15 (4%) from South America, and 3 (1%) from Africa (Figure 3 and Figure 4). More specifically, Pichia anomala (synonyms: Candida pelliculosa, Hansenula anomala, Wickerhamomyces anomalus)was isolated in 108 (31%) cases (82% in Asia), Kodamaea ohmeri (previously known as Pichia ohmeri) in 54 (16%) cases (94% in Asia), Malassezia furfur in 51 (15%) cases (48% in USA, 48% in Europe), Malassezia pachydermatis in 29 (8%) cases (55% in USA), Pichia fabianii (synonym: Cyberlindnera fabianii) in 25 (7%) cases (84% in Asia), Trichosporon asahii in 14 (4%) cases (92% of the cases were in Asia), Dirkmeia churashimaensis in 12 (3%) cases (100% in Asia), Pichia kudriavzevii in 9 (3%) cases (100% in Asia), Trichosporon beigelii in 6 (2%) cases (83% in USA), Saccharomyces cerevisiae in 6 (2%) cases (67% in Asia), Rhodotorula mucilaginosa in 5 cases (1%) (80% in Europe), Cryptococcus laurentii (synonym: Papiliotrema laurentii) in 5 (1%) cases (75% in Asia), Rhodotorula glutinis in 4 (1%) cases (100% in Asia), Trichosporon mucoides in 3 cases (100% in Asia), Moesziomyces (Pseudozyma) aphidis in 2 cases (in Asia and in Europe), Lodderomyces elongisporus in 2 cases (in Latin America and in Asia), Trichosporon spp. in one case in Europe, Malassezia sympodialis in one case in South America, Moesziomyces (Pseudozyma) bullatus in one case in Africa, Trichosporon asteroides in one case in Europe, Cryptococcus neoformans in one case in Asia, Aureobasidium melanogenum in one case in Asia, and Cryptococcus terreus in one case in Europe (Figure 5).

3.2. Risk Factors

Low birth weight, the use of central catheters, and prematurity are the three dominant risk factors for fungemia development (Figure 6).

3.3. Outcome

In 12% of the cases, the outcome was not reported. Among the remaining cases, 22% of the neonates, died. Moreover, 80% of the cases with a fatal outcome were reported from Asia (Figure 7). The highest mortality rates were noted in cases of fungemia caused by the Trichosporon species (Figure 8).
Neonates with Moesziomyces bullatus, Aureobasidium melanogenum, and Lodderomyces elongisporus fungemia also died.
The following risk factors were most commonly reported in total cases, compared to those associated with survival or exitus (Table 3).

3.4. Antifungal Susceptibility

Antifungal susceptibility testing was not reported in all cases. Nevertheless, the available data are presented in Table 4.

3.5. Treatment

The antifungal treatments varied notably among the cases, with Fluconazole and Amphotericin B being the most frequently administered medications. Timely removal (or replacement) of central venous catheters within 24 h of a positive blood culture is a crucial aspect of treatment. Conversely, delayed removal or replacement of central venous catheters in neonates with fungemia has been linked to higher mortality and morbidity. Our review and the available data indicated that only 18% of catheters were removed as part of the treatment intervention (Figure 9 and Table 5).
Most yeasts of this review have been recognized for decades as fungal contaminants but not as human pathogens. Systemic infections caused by them have been considered to be rare and most published studies to date are sporadic cases from all over the world. According to the data above, the majority of neonatal fungemia cases were reported in Asia, where developing countries often have low socio-economic conditions and living standards, with shortages in resources, infrastructure, and modern methods of diagnosing infections, as well as insufficient surveillance, legislation, infection control measures, and inadequately trained medical personnel who lack sufficient information about rare fungi [96]. To the best of our knowledge, from 1981 to 1990 no cases of neonatal fungemia from Asia were published and this is probably explained by the above.
Fluconazole prophylaxis is recommended worldwide in preterm infants < 1000gr from birth and until they no longer require central or peripheral catheters, as it is assumed to decrease mortality [96]. According to the respective guidelines, a third-generation cephalosporin therapy, duration of treatment with a systemic broad-spectrum antibiotic for more than 10 days, fungal colonization, and the use of a central venous catheter, are additional factors for prophylactic administration of fluconazole in such vulnerable organisms [97].
A previous systematic review and meta–analysis showed that prophylactic administration of fluconazole in different regimens may reduce mortality. Furthermore, prophylactic administration of the same regimens of fluconazole in ELBW neonates may reduce the incidence of mortality due to invasive fungal infections [98]. In addition, amphotericin B is not commercially available in 42 developing countries and is not licensed in 22 countries, making fluconazole an effective alternative therapy [96]. Nevertheless, the overuse of fluconazole in these countries, as a cheap prophylaxis in preterm low birth weight neonates, may contribute to infection development by fluconazole-resistant species [96,99]. The current study highlighted, in agreement with previous findings, the high fluconazole-resistant rates of rare yeast species such as Pichia spp., Kodamaea ohmeri, Trichosporon spp., Rhodotorula spp., Saccharomyces cerevisiae, etc. [100,101]. Therefore, the isolates of the present review showed elevated fluconazole MICs. This may explain why fluconazole was not the appropriate antifungal agent against those uncommon yeast species, despite the fact that it had been preferred as a treatment in 38% of all the fungemia cases. Unfortunately, 46% of the neonates who had received fluconazole therapy died. Therefore, the prophylactic use of fluconazole in neonatal intensive care units (NICUs) should be done with care to avoid resistance development and the subsequent selection of resistant species or species with reduced azole susceptibility [101]. Alternatively, nystatin is inexpensive, well-tolerated, safe, and so far with good results when prophylactically administered to preterm infants with risk factors [102,103].

4. Discussion

In our review the highest mortality was revealed in Trichosporon fungemia cases: 80% by T. beigelii and 71% by T. asahii. Previous studies have demonstrated that Trichosporon species were more competent than other basidiomycetes such as Rhodotorula spp. and Cryptococcus spp., to produce dense biofilms, resistant to triazoles or amphotericin B [104,105,106]. This seems to agree with our results in which all the neonatal fungemia cases by Rhodotorula spp. and Cryptococcus spp., showed zero mortality rates, despite their ability to produce biofilms on the medical devices. Previous research revealed the isolation of Trichosporon spp. from the hospital environment and the skin of premature neonates. Moreover, T. beigelii (T. cutaneum) was more often reported in these older studies as the causative organism of invasive infections. Nevertheless, T. asahii is now recognized as the dominant cause of invasive trichosporonosis threatening patients in immunosuppression and risk factors, such as neutropenia, prematurity, AIDS, extensive burns, use of catheters, broad-spectrum antibiotics or corticosteroids, mechanical ventilation, heart valve surgery, etc. Trichosporonosis is a difficult-to-treat infection due to the frequent resistance of yeast, as well as to amphotericin B, fluconazole, and combinations of the two. Nevertheless, voriconazole, posaconazole, and ravuconazole seem to act more effectively against Trichosporon species [105].
According to our data, P. fabianii fungemia cases demonstrated 29% mortality. P. fabianii (Candida fabianii, Cyberlindnera fabianii) is an opportunistic, emerging yeast species and may cause invasive bloodstream infections often with fatal outcomes, mainly due to its characteristic ability to produce biofilms causing resistance to antifungals [107]. Previous studies reported that P. fabianii may develop resistance to fluconazole, voriconazole, caspofungin, and amphotericin B [108,109].
P. anomala (Wickerhamomyces anomalus) is a plant pathogen and opportunistic cause of fungemia in both immunocompetent and immunocompromised patients. In our study, 17% of the cases had a fatal outcome. Pichia anomala has previously been shown to exhibit variable antifungal susceptibility with fluconazole resistance predominance [110,111]. This seems to agree with our findings and must be taken into account in NICUs for effective antifungal prophylactic and therapeutic strategies.
Fungemia by P. kudriavzevii (Candida krusei) is another life-threatening and difficult-to-treat infection due to its intrinsic resistance to fluconazole and the ability for rapid resistance development to other antifungal drugs. Vulnerable patients suffering from gastrointestinal diseases, who had previously received antibiotics, especially carbapenems, and prophylactic fluconazole, are at greater risk of developing invasive infection. In addition, whenever outbreaks occurred, sink traps and surfaces, intravenous dextrose multi-electrolyte infusion bottles, suction devices, pediatric emergency wards, and hands of health care personnel were reported as the most common sources of transmission [110]. Moreover, this species is able to produce a robust biofilm, formed by multiple layers of pseudohyphae in the polymer matrix [111]
All 12 of the neonatal fungemia cases by Dirkmeia occurred in the same NICU of a multispecialty hospital in Delhi with a 42% mortality rate (5/12). Surveillance cultures were obtained, but the source of infection was not identified. Infection control measures appeared to be effective as no further cases of Dirkmeia fungemia were reported [82]. All the twelve isolates of Dirkmeia had high minimum inhibitory concentrations for echinocandins and low minimum inhibitory concentrations for azoles, amphotericin B, and flucytosine.
39% of the neonates who developed fungemia by K. ohmeri, died. Chakrabarti et al. showed that prolonged hospitalization, piperacillin-tazobactam administration, endotracheal intubation, and mechanical ventilation, were major risk factors for the fungemia development by this rare yeast. They also demonstrated that the patients with K. ohmeri fungemia had significantly higher (50%) mortality than other groups of patients (some of them with C. tropicalis fungemia and others without fungemia), mainly due to the virulence mechanisms of the yeast species [61]. The identification of K. ohmeri in the laboratory is a challenge. On Candida chromogenic agar the color of K. ohmeri colonies changes from pink to blue within 48 h. Usually, species identification is performed by automated identification systems such as API 20C, Vitek 2 ID YST, and Microscan with molecular methods. For molecular identification PCR amplification is used followed by sequencing of 18S rRNA, the D1/D2 domains of 26SrRNA, the internal transcriber spacer 1/2 (ITS) of the ribosomal DNA,28SrRNA, and/or 5.8SrRNA. The pulsed field gel electrophoresis for karyotyping has been also used. Restriction endonuclease analysis of NotI-digested DNA (REAG-N) is used for genotyping of the clinical isolates of K. ohmeri. Fluorescent amplified fragment length polymorphism was used by Chakrabarti et al. for molecular typing. Moreover, in their study, K. ohmeri strains presented high fluconazole MICs [61].
Among all the Malassezia spp. fungemia cases, M. furfur dominated, from which 25% had a fatal outcome, followed by M. pachydermatis, with 4% mortality, and one case of M. sympodialis (the newborn survived). It was previously demonstrated that M. furfur presents high MICs to azoles. Therefore, the administration of prophylactic fluconazole in patients with risk factors such as prematurity, may lead to resistant strains and M. furfur colonization [112]. In catheter-related neonatal colonization by M. furfur, the removal of the catheter and discontinuation of intravenous lipid administration are usually sufficient and effective strategies. Furthermore, it is demonstrated that in life-threatening M. furfur fungemia, the preferred antifungal drug is amphotericin B [79]. In this review, M. furfur isolates showed elevated MICs for echinocandins.
This study also revealed the first case of neonatal fungemia due to Aureobasidium melanogenum. Aureobasidium species can survive in extreme ecological conditions, usually found in wet, oligotrophic environments, while their natural habitats are often subjected to osmotic stress and in many cases to solar radiation. Aureobasidium species can also survive on wet surfaces such as in bathrooms and saunas, in hospital environments, and can colonize organs and human skin [113]. Wang M et al. compared the two species A. pullulans and A. melanogenum and found that Aureobasidium melanogenum showed significantly better survival at 37 °C than A. pullulans, possibly due to its response to elevated temperatures with elevated melanin production which A. pullulans did not. This mechanism probably enhanced the virulence and pathogenicity of A. melanogenum [113]. This is why its accurate and timely identification is key to proper treatment as A. melanogenum also exhibits significant antifungal resistance. Unfortunately, A. melanogenum can be easily confused with Candida species on Gram-stained smears, while it cannot be easily identified by conventional diagnostic methods, such as VITEK 2 and MALDI-TOF MS, leaving molecular techniques as the only diagnostic selection [94].
It has recently been suggested that climate change and particularly global warming have affected the ecosystem microbiome with dramatic effects on human health. High temperatures and heat waves can lead to microbes adapting, making them thermotolerant and allowing them to survive at the human body temperature. This is particularly important for environmental fungi, most of which grow best below 37 °C. According to this view, more fungal species such as Aureobasidium melanogenum with resistance to environmental stress and increased pathogenicity will appear more and more frequently due to the global climate crisis threatening vulnerable organisms [114]. The same hypothesis has been made for Candida auris, which probably acquired the property of heat resistance due to global warming, which transformed it from an environmental species to an opportunistic human pathogen [115].
In this study, two fatal neonatal fungemia cases from another opportunistic yeast pathogen, Lodderomyces elongisporus, were also included. Its close relationship with C. parapsilosis confuses correct diagnosis, delaying treatment [92,93]. Clinical isolates of L. elongisporus have previously been misidentified as C. parapsilosis by conventional methods such as API 20C, ID 32C, and Vitek 2, while MALDI-TOF MS and molecular methods were required for correct identification [116]. However, it has been suggested that CHROMagar can be used as a preliminary identification method in laboratories where molecular methods are not available as on this agar isolates of L. elongisporus form turquoise blue colonies instead of the white to light pink colonies of C. parapsilosis [117]. This is particularly important for the timely selection of the correct antifungal therapy, as echinocandins appear to be quite effective for treating L. elongisporus infections but not for C. parapsilosis infections mainly due to the different amino acid sequence of beta-1,3 glucan synthase, which is the target of this class of antifungals [116].
This study, also showed that low birth weight, the use of central catheters, prematurity, and the use of broad-spectrum antibiotics constituted the strongest risk factors for neonatal fungemia cases as well as for the cases with fatal outcomes. It is, after all, recognized that birth weight is the major predictor of nosocomial infection development and our results are in accordance with this [118].
The birth process plays a major role in neonatal colonization by fungi. During the natural delivery, the neonatal skin is directly colonized from the maternal vaginal mycobiome. It has been previously shown that this mode of delivery is related to high colonization of Malassezia [119]. On the other hand, premature neonates, are mainly born via cesarian section and are thus colonized by maternal skin and the hospital environment. It has also been shown that the babies are immediately colonized by Malassezia species after birth with a predominance of Malassezia furfur, Malassezia sympodialis, and Malassezia restricta [119]. Moreover, it has been found that the neonatal gut mycobiome is already shaped at the age of 10 days with a prevalence of Candida albicans and Malassezia spp. Furthermore, Saccharomyces cerevisiae has been found to be the dominant fungal species in the gut mycobiome of both mothers and their 1–2-year-old babies [120]. In addition, the gut microbiome of a premature newborn contains a higher proportion of fungi than an adult’s. Moreover, factors associated with prematurity, such as an immature gastrointestinal system and intestinal barrier, an underdeveloped immune response, administration of antibiotics, and antifungal prophylaxis, can cause hematogenous dissemination of fungi and fungemia [121,122]. The type of prophylactic antifungal drug plays an important role in the composition of the gut mycobiome: a prophylactic administration of azoles may lead to prevalence of Candida glabrata and other azole-resistant species, while a prophylactic echinocandin strategy may cause prevalence of Candida parapsilosis or other echinocandin resistant species [123,124].
Neonatal infections within 48 h of birth are therefore associated with pathogens transmitted from the mother’s vagina. Other modes of transmission of microorganisms to neonates are by contact with a person or a contaminated source, droplets, or airborne transmission. Fungi are widely distributed in the environment and thus can be transmitted very easily by human agents or inanimate objects and threaten the lives of such vulnerable patients. In addition to the immature immune system, skin, or mucosal barriers, other risk factors for the development of life-threatening neonatal infections such as fungemia, include the use of catheters, invasive procedures, steroid administration, prolonged hospitalization, mechanical respiratory support, prolonged parenteral nutrition, necrotizing enterocolitis, etc. [118,121]. The colonization of the catheter or the skin at the insertion site, contamination of intravenous fluids, low frequency of catheter tubing changes, or catheter replacement may also predispose individuals to bloodstream infections. Removal of central venous catheters is therefore critical to the outcome of fungemia [123]. Moreover, it has been previously reported that the retention of a catheter is associated with increased mortality rates in Candida bloodstream infections [124]. The strong association between not removing the catheter and the fatal outcome was also revealed in the present review.
According to our findings, intubation and mechanical ventilation have been found in 34% of all the fungemia cases and in 65% of the fatal cases. Mechanical ventilation is recognized as a risk factor for bloodstream infections due to colonization of humidified air, or due to traumatism from the endotracheal tube and its suctioning [125]. Prolonged hospital stay also dominated in the cases with lethal outcomes (43%) compared with total cases (18%). This is understandable because a prolonged hospital stay means prolonged presence of indwelling catheters, performance of invasive procedures, and prolonged use of broad-spectrum antibiotics that favor the colonization of fungal pathogens [126]. Total parenteral nutrition and intralipids may also predispose to fungemia due to the risk of contamination or translocation of pathogens across the immature neonatal gastrointestinal mucosa and bloodstream dissemination [127]. Moreover, intralipids are predispose to infections by Malassezia spp. [118].
Regarding treatment management, our systematic review revealed that antifungal therapies varied significantly among cases, with Fluconazole and Amphotericin B being the most commonly administered medications. Timely removal (or replacement) of central venous catheters within 24 h of a positive blood culture is a critical component of effective treatment. Conversely, delayed removal or replacement of central venous catheters in neonates with fungemia has been associated with increased mortality and morbidity [128]. Our review and the available data showed that only 18% of catheters were removed as part of the treatment intervention. A multicenter prospective study involving 13 neonatal intensive care units was conducted in Italy to evaluate the incidence of bacterial sepsis and invasive fungal infections, fungal colonization, risk factors for sepsis and mortality rates in neonates and infants under 3 months old undergoing major surgery. They concluded that preventive measures, including early removal of vascular catheters and the use of fluconazole prophylaxis, should be implemented to mitigate the risk of bacterial and fungal infection in infants undergoing abdominal surgery, especially those with fungal colonization [129].
As mentioned above, fungal infections from fungi that were previously not considered pathogenic have been reported more and more frequently in recent years, which was also evident in our study. Given the rapid development of multidrug resistance of these emerging fungi, these infections are difficult to manage and are often associated with high mortality rates. Several studies have hypothesized an environmental pathway for this development of fungal drug resistance. For example, the widespread use of antifungals in agriculture may have caused rapid species evolution and selection of resistant strains [130]. Such opportunistic fungi can then especially threaten immunocompromised individuals including neonates.
Interestingly, our study revealed a steady increase in reported cases from 1991 to 2000, followed by a near tripling of cases from 2001 to 2023. The increase in neonatal fungemia is partially attributed to the enhanced survival rates of very low birth weight (VLBW) and extremely low birth weight (ELBW) infants. This progress has been achieved due to the regionalization of perinatal care, a better understanding of the pathophysiology of extremely premature neonates, the administration of antenatal steroids, and the critical use of postnatal surfactant therapy. Surfactant therapy has significantly improved outcomes in preterm infants by reducing the incidence and severity of neonatal respiratory distress syndrome (RDS), a common complication in very preterm infants. The introduction of surfactants has enhanced lung function, decreased the need for mechanical ventilation, and improved overall survival rates. Advances in surfactant formulations and administration techniques have further optimized its effectiveness [130]. Additionally, the use of intravenous nutrition and ongoing technological advancements have contributed to the overall improvement in preterm infant care.
Despite significant advances in perinatal medicine, caring for extremely preterm infants remains challenging due to their high mortality and morbidity risks. While survival rates have improved, these infants continue to face serious health issues. Effective management strategies, supported by meta-analyses and randomized controlled trials, are essential for reducing mortality and impairments. However, the efficacy and safety of some emerging strategies are still uncertain. Neonatal sepsis is a major concern, exacerbated by immature immune systems, prolonged hospitalizations, and frequent invasive procedures. Empiric broad-spectrum antibiotics are commonly used to treat sepsis, with therapy adjusted once pathogens are identified. Prophylactic maternal antibiotics and infection control measures, such as hand hygiene and central line care, are recommended. Nonetheless, overuse of antibiotics has led to increased rates of multidrug-resistant organisms and related complications, including bronchopulmonary dysplasia (BPD), necrotizing enterocolitis (NEC), and fungal infections. Furthermore, climate change, agricultural practices, occupational hazards, deforestation, human migration patterns, soil dispersion, patient immunosuppression, and advancements in infection detection and diagnostic testing all contribute to the rise of fungal diseases. The increasing rates of fungal morbidity and mortality are closely linked to antifungal resistance, drug tolerance, and biofilm formation [131]. Antifungal tolerance refers to the partial growth of fungi after 24 h of exposure to inhibitory drug concentrations, as evidenced in susceptibility tests. In contrast, antifungal resistance is characterized by the complete lack of a toxic effect on fungal pathogens despite treatment [132]. Current antifungal therapies are limited to a few drug classes, including polyenes, azoles, echinocandins, allylamines, and flucytosine. While allylamines are primarily used for superficial infections, the other four classes are highly effective against invasive mycosis. Moreover, evidence from randomized trials indicates that fluconazole prophylaxis may increase the risk of colonization with fluconazole-susceptible dose-dependent or resistant fungi. However, it does not significantly alter the risk of invasive infections caused by these fungi. The risk of breakthrough infections remains a concern and should be investigated further in large prospective studies [133]. Hence when managing a patient with or at risk for invasive fungal infections (IFI), clinicians must consider various factors to tailor therapy and optimize outcomes. Recent advancements have expanded antifungal options, each varying in spectrum, pharmacokinetics, indications, safety, cost, and usability. Matching these drug characteristics with patient-specific factors is crucial for effective treatment with minimal toxicity. Decision-making becomes especially complex for highly immunocompromised patients, such as ELBW neonates with rare IFIs, where a high level of suspicion is needed for early initiation of therapy. To achieve the best outcomes, extended antifungal treatment should be combined with addressing underlying risk factors and performing radical debridement of affected tissues.

5. Study Limitations

It is important to acknowledge the limitations of our study and interpret its findings with caution. The primary limitation is that our analysis relied exclusively on data from case reports and case series, which are known for their limited generalizability and validity, and their inability to establish causal relationships. Additionally, there may be significant biases related to publication, the retrospective nature of the study, and the focus on rare or atypical cases. However, some case reports and series can still provide valuable insights, especially in the absence of data from randomized controlled trials (RCTs) and observational studies. This is particularly relevant when a few studies suggest a significant and probable causal link in the context of an emerging epidemic or a newly introduced medication [134,135].
It should also be noted that due to the ongoing flux in the nomenclature of fungal agents, several Candida spp. have been renamed non-Candida genus, which are no longer considered causative agents for candidiasis. For example, Candida pelliculosa (now renamed to Wickerhamomyces anomalus/Pichia anomala) or Candida norvegensis (now renamed to Pichia norvegensis) [136].
In addition, significant neonatal risk factors were not available for all cases. There was also a lack of evidence on antifungal prophylaxis, antifungal susceptibility testing, and treatment administered. Moreover, fungemia-attributable mortality data was not available for all cases as well. Such gaps together with significant heterogeneity between review studies in recording baseline data were the main inhibiting factors for conducting a meta-analysis.

6. Conclusions

The present study has provided us with extended insight into the epidemiology of neonatal fungemia by non-Candida rare opportunistic yeasts among geographically different areas all over the world for an extended time period.
A rapid identification of these rare yeasts is a key element for adequate therapy and better outcomes for these vulnerable patients. Although recent advancements have paved the way with modern techniques and methodologies, fungemia by these opportunistic pathogens remains underestimated. Furthermore, it is obvious that the compliance of healthcare workers to general principles of hygiene is crucial in neonatal intensive care units. The problem is bigger in developing countries mainly due to a deficiency of resources.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25179266/s1, Reference [137] is cited in the supplementary materials.

Author Contributions

Conceptualization, A.M. and R.S.; methodology, A.M. and R.S.; investigation, A.M., R.S., V.C., J.M. and M.T.; data curation, A.M., R.S., J.M., V.C. and M.T.; writing—original draft preparation, A.M., R.S., J.M., V.C. and M.T.; writing—review and editing, A.M., R.S., J.M., V.C. and M.T.; supervision, A.M. and R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Stoll, B.J.; Hansen, N.; Fanaroff, A.A.; Wright, L.L.; Carlo, W.A.; Ehrenkranz, R.A.; Lemons, J.A.; Donovan, E.F.; Stark, A.R.; Tyson, J.E.; et al. Late-Onset Sepsis in Very Low Birth Weight Neonates: The Experience of the NICHD Neonatal Research Network. Pediatrics 2002, 110, 285–291. [Google Scholar] [CrossRef]
  2. Weimer, K.E.D.; Smith, P.B.; Puia-Dumitrescu, M.; Aleem, S. Invasive fungal infections in neonates: A review. Pediatr. Res. 2021, 91, 404–412. [Google Scholar] [CrossRef] [PubMed]
  3. Taliaka, P.K.; Tsantes, A.G.; Konstantinidi, A.; Houhoula, D.; Tsante, K.A.; Vaiopoulos, A.G.; Piovani, D.; Nikolopoulos, G.K.; Bonovas, S.; Iacovidou, N.; et al. Risk Factors, Diagnosis, and Treatment of Neonatal Fungal Liver Abscess: A Systematic Review of the Literature. Life 2023, 13, 167. [Google Scholar] [CrossRef]
  4. Nelson, R. Emergence of resistant Candida auris. Lancet Microbe 2023, 4, e396. [Google Scholar] [CrossRef]
  5. Sokou, R.; Palioura, A.E.; Kopanou Taliaka, P.; Konstantinidi, A.; Tsantes, A.G.; Piovani, D.; Tsante, K.A.; Gounari, E.A.; Iliodromiti, Z.; Boutsikou, T.; et al. Candida auris Infection, a Rapidly Emerging Threat in the Neonatal Intensive Care Units: A Systematic Review. J. Clin. Med. 2024, 13, 1586. [Google Scholar] [CrossRef] [PubMed]
  6. Miceli, M.H.; Díaz, J.A.; Lee, S.A. Emerging opportunistic yeast infections. Lancet Infect. Dis. 2011, 11, 142–151. [Google Scholar] [CrossRef]
  7. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. J. Clin. Epidemiol. 2009, 62, 1006–1012. [Google Scholar] [CrossRef]
  8. Redline, R.W.; Dahms, B.B. Malassezia Pulmonary Vasculitis in an Infant on Long-Term Intralipid Therapy. N. Engl. J. Med. 1981, 305, 1395–1398. [Google Scholar] [CrossRef]
  9. Powell, D.A.; Aungst, J.; Snedden, S.; Hansen, N.; Brady, M. Broviac catheter-related Malassezia furfur sepsis in five infants receiving intravenous fat emulsions. J. Pediatr. 1984, 105, 987–990. [Google Scholar] [CrossRef]
  10. Redline, R.W.; Redline, S.S.; Boxerbaum, B.; Dahms, B.B. Systemic Malassezia furfur infections in patients receiving intralipid therapy. Hum. Pathol. 1985, 16, 815–822. [Google Scholar] [CrossRef]
  11. Murphy, N.; Damjanovic, V.; Hart, C.A.; Buchanan, C.R.; Whitaker, R.; Cooke, R.W.I. Infection and colonisation of neonates by hansenula anomala. Lancet 1986, 327, 291–293. [Google Scholar] [CrossRef] [PubMed]
  12. Dankner, W.M.; Spector, S.A.; Fierer, J.; Davis, C.E. Malassezia fungemia in neonates and adults: Complication of hyperalimentation. Rev. Infect. Dis. 1987, 9, 743–753. [Google Scholar] [CrossRef] [PubMed]
  13. Alpert, G.; Bell, L.M.; Campos, J.M. Malassezia furfur fungemia in infancy. Clin. Pediatr. 1987, 26, 528–531. [Google Scholar] [CrossRef]
  14. Shek, Y.H.; Tucker, M.C.; Viciana, A.L.; Manz, H.J.; Connor, D.H. Malassezia furfur--disseminated infection in premature infants. Am. J. Clin. Pathol. 1989, 92, 595–603. [Google Scholar] [CrossRef]
  15. Surmont, I.; Gavilanes, A.; Vandepitte, J.; Devlieger, H.; Eggermont, E. Malassezia furfur fungaemia in infants receiving intravenous lipid emulsions. A rarity or just underestimated? Eur. J. Pediatr. 1989, 148, 435–438. [Google Scholar] [CrossRef]
  16. Weiss Malassezia Furfur Fungemia Associated with Central Venous Catheter Lipid Emulsion Infusion—Aναζήτηση Google. Available online: https://www.google.com/search?q=weiss+Malassezia+furfur+fungemia+associated+with+central+venous+catheter+lipid+emulsion+infusion&sca_esv=578293274&ei=soJBZf2BAoyGxc8Pn--UgA0&udm=&ved=0ahUKEwj9ooTkraGCAxUMQ_EDHZ83BdAQ4dUDCBA&uact=5&oq=weiss+Malassezia+furf (accessed on 1 November 2023).
  17. Hruszkewycz, V.; Holtrop, P.C.; Batton, D.G.; Morden, R.S.; Gibson, P.; Band, J.D. Complications associated with central venous catheters inserted in critically ill neonates. Infect. Control Hosp. Epidemiol. 1991, 12, 544–548. [Google Scholar] [CrossRef] [PubMed]
  18. Masure, O.; Leostic, C.; Abalain, M.; Chastel, C.; Yakoub-Agha, I.; Berthou, C.; Briere, J. Malassezia furfur septicaemia in a child with leukaemia. J. Infect. 1991, 23, 335–336. [Google Scholar] [CrossRef]
  19. Giacoia, G.P. Trichosporon beigelii: A Potential Cause of Sepsis in Premature Infants. South. Med. J. 1992, 85, 1247–1248. [Google Scholar] [CrossRef]
  20. Herbrecht, R.; Waller, J.; Dufour, P.; Koenig, H.; Lioure, B.; Marcellin, L.; Oberling, F. Rare opportunistic fungal diseases in patients with organ or bone marrow transplantation. Agressologie 1992, 33, 77–80. [Google Scholar]
  21. Fisher, D.J.; Christy, C.; Spafford, P.; Maniscalco, W.M.; Hardy, D.J.; Graman, P.S. Neonatal Trichosporon beigelii infection: Report of a cluster of cases in a neonatal intensive care unit. Pediatr. Infect. Dis. J. 1993, 12, 149–155. [Google Scholar] [CrossRef]
  22. Welbel, S.F.; McNeil, M.M.; Pramanik, A.; Silberman, R.; Oberle, A.D.; Midgley, G.; Crow, S.; Jarvis, W.R. Nosocomial Malassezia pachydermatis bloodstream infections in a neonatal intensive care unit. Pediatr. Infect. Dis. J. 1994, 13, 104–108. [Google Scholar] [CrossRef] [PubMed]
  23. Yoss, B.S.; Sautter, R.L.; Brenker, H.J. Trichosporon beigelii, a new neonatal pathogen. Am. J. Perinatol. 1997, 14, 113–117. [Google Scholar] [CrossRef] [PubMed]
  24. Johnson, L.B.; Bradley, S.F.; Kauffman, C.A. Fungaemia due to Cryptococcus laurentii and a review of non-neoformans cryptococcaemia. Mycoses 1998, 41, 277–280. [Google Scholar] [CrossRef] [PubMed]
  25. Chang, H.J.; Miller, H.L.; Watkins, N.; Arduino, M.J.; Ashford, D.A.; Midgley, G.; Aguero, S.M.; Pinto-Powell, R.; von Reyn, C.F.; Edwards, W.; et al. An epidemic of Malassezia pachydermatis in an intensive care nursery associated with colonization of health care workers’ pet dogs. N. Engl. J. Med. 1998, 338, 706–711. [Google Scholar] [CrossRef]
  26. Sweet, D.; Reid, M. Disseminated neonatal Trichosporon beigelii infection: Successful treatment with liposomal amphotericin B. J. Infect. 1998, 36, 120–121. [Google Scholar] [CrossRef]
  27. Singh, K.; Chakrabarti, A.; Narang, A.; Gopalan, S. Yeast colonisation & fungaemia in preterm neonates in a tertiary care centre. Indian J. Med. Res. 1999, 110, 169. [Google Scholar]
  28. Perapoch, J.; Planes, A.M.; Querol, A.; López, V.; Martínez-Bendayán, I.; Tormo, R.; Fernández, F.; Peguero, G.; Salcedo, S. Fungemia with Saccharomyces cerevisiae in two newborns, only one of whom had been treated with ultra-levura. Eur. J. Clin. Microbiol. Infect. Dis. 2000, 19, 468–470. [Google Scholar] [CrossRef]
  29. Ma, J.S.; Chen, P.Y.; Chen, C.H.; Chi, C.S. Neonatal fungemia caused by Hansenula anomala: A case report. J. Microbiol. Immunol. Infect. 2000, 33, 267–270. [Google Scholar]
  30. Wong, A.R.; Ibrahim, H.; Van Rostenberghe, H.; Ishak, Z.; Radzi, M.J. Hansenula anomala infection in a neonate. J. Paediatr. Child. Health 2000, 36, 609–610. [Google Scholar] [CrossRef]
  31. Chakrabarti, A.; Singh, K.; Narang, A.; Singhi, S.; Batra, R.; Rao, K.L.; Ray, P.; Gopalan, S.; Das, S.; Gupta, V.; et al. Outbreak of Pichia anomala infection in the pediatric service of a tertiary-care center in Northern India. J. Clin. Microbiol. 2001, 39, 1702–1706. [Google Scholar] [CrossRef]
  32. Aragão, P.A.; Oshiro, I.C.V.; Manrique, E.I.; Gomes, C.C.; Matsuo, L.L.; Leone, C.; Moretti-Branchini, M.L.; Levin, A.S. Pichia anomala outbreak in a nursery: Exogenous source? Pediatr. Infect Dis. J. 2001, 20, 843–848. [Google Scholar] [CrossRef] [PubMed]
  33. Cheng, M.-F.; Chiou, C.C.; Liu, Y.-C.; Wang, H.-Z.; Hsieh, K.-S. Cryptococcus laurentii Fungemia in a Premature Neonate. J. Clin. Microbiol. 2001, 39, 1608–1611. [Google Scholar] [CrossRef]
  34. Chryssanthou, E.; Broberger, U.; Petrini, B. Malassezia pachydermatis fungaemia in a neonatal intensive care unit. Acta Paediatr. 2007, 90, 323–327. [Google Scholar] [CrossRef]
  35. Panagopoulou, P.; Evdoridou, J.; Bibashi, E.; Filioti, J.; Sofianou, D.; Kremenopoulos, G.; Roilides, E. Trichosporon asahii: An unusual cause of invasive infection in neonates. Pediatr. Infect. Dis. J. 2002, 21, 169–170. [Google Scholar] [CrossRef] [PubMed]
  36. Yıldıran, A.; Kücüködük, Ş.; Saniç, A.; Belet, N.; Güvenli, A. Disseminated Trichosporon asahii Infection in a Preterm. Am. J. Perinatol. 2003, 20, 269–271. [Google Scholar] [CrossRef] [PubMed]
  37. Gökahmetoğlu, S.; Nedret Koç, A.; Güneş, T.; Cetin, N. Case reports. Trichosporon mucoides infection in three premature newborns. Mycoses 2002, 45, 123–125. [Google Scholar] [CrossRef]
  38. Baron, E.; Anaissie, E.; Dumphy, F.; McCredie, K.; Fainstein, V. Hansenula anomala fungemia. Clin. Infect. Dis. 1988, 10, 1182–1186. [Google Scholar] [CrossRef]
  39. Bakır, M.; Çerikcioğlu, N.; Tırtır, A.; Berrak, S.; Özek, E.; Canpolat, C. Pichia anomala fungaemia in immunocompromised children. Mycoses 2004, 47, 231–235. [Google Scholar] [CrossRef] [PubMed]
  40. Belet, N.; Dalgiç, N.; Oncel, S.; Ciftçi, E.; Ince, E.; Güriz, H.; Barlas, M.; Doğru, U. Catheter-related fungemia caused by Saccharomyces cerevisiae in a newborn. Pediatr. Infect. Dis. J. 2005, 24, 1125. [Google Scholar] [CrossRef]
  41. Taj-Aldeen, S.J.; Doiphode, S.H.; Han, X.Y. Kodamaea (Pichia) ohmeri fungaemia in a premature neonate. J. Med. Microbiol. 2006, 55, 237–239. [Google Scholar] [CrossRef]
  42. Bhally, H.S.; Jain, S.; Shields, C.; Halsey, N.; Cristofalo, E.; Merz, W.G. Infection in a neonate caused by Pichia fabianii: Importance of molecular identification. Med. Mycol. 2006, 44, 185–187. [Google Scholar] [CrossRef] [PubMed]
  43. Paula, C.R.; Krebs, V.L.; Auler, M.E.; Ruiz, L.S.; Matsumoto, F.E.; Silva, E.H.; Diniz, E.M.; Vaz, F.A. Nosocomial infection in newborns by Pichia anomala in a Brazilian intensive care unit. Med. Mycol. 2006, 44, 479–484. [Google Scholar] [CrossRef] [PubMed]
  44. Perniola, R.; Faneschi, M.L.; Manso, E.; Pizzolante, M.; Rizzo, A.; Damiani, A.S.; Longo, R. Rhodotorula mucilaginosa outbreak in neonatal intensive care unit: Microbiological features, clinical presentation, and analysis of related variables. Eur. J. Clin. Microbiol. Infect. Dis. 2006, 25, 193–196. [Google Scholar] [CrossRef] [PubMed]
  45. Vivas, R.; Beltran, C.; Munera, M.I.; Trujillo, M.; Restrepo, A.; Garcés, C. Fungemia due to Kodamaea ohmeri in a young infant and review of the literature. Med. Mycol. Case Rep. 2016, 13, 5–8. [Google Scholar] [CrossRef]
  46. Ngerncham, S.; Lapphra, K. Cryptococcus neoformans septicemia in an immunocompetent neonate: First case report in Thailand. Southeast Asian J. Trop. Med. Public Health 2008, 39, 697–700. [Google Scholar]
  47. Téllez-Castillo, C.J.; Gil-Fortuño, M.; Centelles-Sales, I.; Sabater-Vidal, S.; Serrano, F.P. Trichosporon asahii fatal infection in a preterm newborn. Rev. Chil. Infectología 2008, 25, 213–215. [Google Scholar]
  48. Poojary, A.; Sapre, G. Kodamaea ohmeri infection in a neonate. Indian Pediatr. 2009, 46, 629–631. [Google Scholar]
  49. Pereira, D.N.; Nader, S.S.; Nader, P.; Martins, P.G.; Furlan, S.P.; Hentges, C.R. Disseminated Trichosporon spp infection in preterm newborns: A case report. J. Pediatr. 2009, 85, 459–461. [Google Scholar]
  50. Noni, M.; Stathi, A.; Velegraki, A.; Malamati, M.; Kalampaliki, A.; Zachariadou, L.; Michos, A. Rare Invasive Yeast Infections in Greek Neonates and Children, a Retrospective 12-Year Study. J. Fungi 2020, 6, 194. [Google Scholar] [CrossRef]
  51. Grenouillet, F.; Millon, L.; Chamouine, A.; Thiriez, G.; Schulze, O.; Leroy, J. Pichia fabianii Fungemia in a Neonate. Pediatr. Infect. Dis. J. 2010, 29, 191. [Google Scholar] [CrossRef]
  52. Duggal, S.; Jain, H.; Tyagi, A.; Sharma, A.; Chugh, T.D. Rhodotorula fungemia: Two cases and a brief review. Med. Mycol. 2011, 49, 879–882. [Google Scholar] [PubMed]
  53. Oliveri, S.; Trovato, L.; Betta, P.; Romeo, M.G.; Nicoletti, G. Malassezia furfur fungaemia in a neonatal patient detected by lysis-centrifugation blood culture method: First case reported in Italy. Mycoses 2011, 54, 638. [Google Scholar] [CrossRef]
  54. Al-Sweih, N.; Khan, Z.U.; Ahmad, S.; Devarajan, L.; Khan, S.; Joseph, L.; Chandy, R. Kodamaea ohmeri as an emerging pathogen: A case report and review of the literature. Med. Mycol. 2011, 49, 766–770. [Google Scholar] [PubMed]
  55. Vashishtha, V.M.; Mittal, A.; Garg, A. A fatal outbreak of Trichosporon asahii Sepsis in a neonatal intensive care unit. Indian Pediatr. 2012, 49, 745–747. [Google Scholar] [CrossRef]
  56. Lin, H.-C.; Lin, H.-Y.; Su, B.-H.; Ho, M.-W.; Ho, C.-M.; Lee, C.-Y.; Lin, M.-H.; Hsieh, H.-Y.; Lin, H.-C.; Li, T.-C.; et al. Reporting an outbreak of Candida pelliculosa fungemia in a neonatal intensive care unit. J. Microbiol. Immunol. Infect. 2013, 46, 456–462. [Google Scholar] [CrossRef]
  57. da Silva, C.M.; de Carvalho Parahym, A.M.; Leão, M.P.; de Oliveira, N.T.; de Jesus Machado Amorim, R.; Neves, R.P. Fungemia by Candida pelliculosa (Pichia anomala) in a neonatal intensive care unit: A possible clonal origin. Mycopathologia 2013, 175, 175–179. [Google Scholar] [CrossRef]
  58. Iatta, R.; Cafarchia, C.; Cuna, T.; Montagna, O.; Laforgia, N.; Gentile, O.; Rizzo, A.; Boekhout, T.; Otranto, D.; Montagna, M.T. Bloodstream infections by Malassezia and Candida species in critical care patients. Med. Mycol. 2014, 52, 264–269. [Google Scholar] [CrossRef]
  59. Chioukh, F.Z.; Ben Hmida, H.; Ben Ameur, K.; Toumi, A.; Monastiri, K. Saccharomyces cerevisiae fungemia in a premature neonate treated receiving probiotics. Med. Mal. Infect. 2013, 43, 359–360. [Google Scholar] [CrossRef]
  60. Liu, C.X.; Yang, J.H.; Dong, L.; Mai, J.Y.; Zhang, L.; Zhu, J.H. Clinical features and homological analysis of Pichia ohmeri-caused hospital-acquired fungemia in premature infants. Zhonghua Yi Xue Za Zhi 2013, 93, 285–288. [Google Scholar]
  61. Wu, Y.; Wang, J.; Li, W.; Jia, H.; Che, J.; Lu, J.; Liu, L.; Cheng, Y. Pichia fabianii blood infection in a premature infant in China: Case report. BMC Res. Notes 2013, 6, 77. [Google Scholar] [CrossRef] [PubMed]
  62. Kodamaea Ohmeri as an Emerging Pathogen in Mainland China—Aναζήτηση Google. Available online: https://www.google.com/search?q=%0D%0AKodamaea+ohmeri+as+an+Emerging+Pathogen+in+Mainland+China&sca_esv=578502807&ei=aWtCZfutD86Rxc8PoKSAsAc&udm=&ved=0ahUKEwi7htvbi6OCAxXOSPEDHSASAHYQ4dUDCBA&uact=5&oq=%0D%0AKodamaea+ohmeri+as+an+Emerging+Pathogen+in+Mainland+China&gs_lp=Egxnd3Mtd2l6LXNlcnAiOgpLb2RhbWFlYSBvaG1lcmkgYXMgYW4gRW1lcmdpbmcgUGF0aG9nZW4gaW4gTWFpbmxhbmQgQ2hpbmFIwwtQnwhYnwhwAXgBkAEAmAEAoAEAqgEAuAEDyAEA-AEB-AECqAIA4gMEGAAgQYgGAQ&sclient=gws-wiz-serp (accessed on 1 November 2023).
  63. Chakrabarti, A.; Rudramurthy, S.M.; Kale, P.; Hariprasath, P.; Dhaliwal, M.; Singhi, S.; Rao, K.L.N. Epidemiological study of a large cluster of fungaemia cases due to Kodamaea ohmeri in an Indian tertiary care centre. Clin. Microbiol. Infect. 2014, 20, O83–O89. [Google Scholar] [CrossRef] [PubMed]
  64. Borade, A.; Kapdi, M.; Suryavanshi, K. Kodamaea Ohmeri—An Emerging Fungal Pathogen in Neonatal Intensive Care Unit. Pediatr. Oncall 2014, 11, 114–116. [Google Scholar] [CrossRef]
  65. Prakash, A.; Wankhede, S.; Singh, P.K.; Agarwal, K.; Kathuria, S.; Sengupta, S.; Barman, P.; Meis, J.F.; Chowdhary, A. First neonatal case of fungaemia due to Pseudozyma aphidis and a global literature review. Mycoses 2014, 57, 64–68. [Google Scholar] [CrossRef]
  66. Al-Sweih, N.; Ahmad, S.; Joseph, L.; Khan, S.; Khan, Z. Malassezia pachydermatis fungemia in a preterm neonate resistant to fluconazole and flucytosine. Med. Mycol. Case Rep. 2014, 5, 9–11. [Google Scholar] [CrossRef]
  67. Biswal, D.; Sahu, M.; Mahajan, A.; Advani, S.H.; Shah, S. Kodameae ohmeri—An Emerging Yeast: Two Cases and Literature Review. J. Clin. Diagn. Res. 2015, 9, DD01. [Google Scholar] [CrossRef]
  68. Mlinarić-Missoni, E.; Hatvani, L.; Kocsube, S.; Vágvölgyi, C.; Škarić, I.; Lukić-Grlić, A. Cyberlindnera fabianii in the neonatal and paediatric intensive care unit: Case reports. JMM Case Rep. 2015, 2, e000032. [Google Scholar] [CrossRef]
  69. Basu, S.; Tilak, R.; Kumar, A. Multidrug-resistant Trichosporon: An unusual fungal sepsis in preterm neonates. Ann. Trop. Med. Parasitol. 2015, 109, 202–206. [Google Scholar] [CrossRef]
  70. Okolo, O.M.; Van Diepeningen, A.D.; Toma, B.; Nnadi, N.E.; Ayanbimpe, M.G.; Onyedibe, I.K.; Sabitu, M.Z.; Banwat, B.E.; Groenewald, M.; Scordino, F.; et al. First report of neonatal sepsis due to Moesziomyces bullatus in a preterm low-birth-weight infant. JMM Case Rep. 2015, 2, e000011. [Google Scholar] [CrossRef]
  71. Socarras, J.A.; Rojas Torres, J.P.; Vargas Soler, J.A.; Guerrero, C. Kodamaea ohmeri infection in a newborn with a mediastinal mass. Arch. Argent. Pediatr. 2016, 114, e319–e322. [Google Scholar]
  72. Yılmaz-Semerci, S.; Demirel, G.; Taştekin, A. Wickerhamomyces anomalus blood stream infection in a term newborn with pneumonia. Turk. J. Pediatr. 2017, 59, 349–351. [Google Scholar] [CrossRef]
  73. Nagarathnamma, T.; Chunchanur, S.K.; Rudramurthy, S.M.; Vineetha, K.R.; Ramamurthy, K.; Joseph, J.; Ambica, R. Outbreak of Pichia kudriavzevii fungaemia in a neonatal intensive care unit. J. Med. Microbiol. 2017, 66, 1759–1764. [Google Scholar] [CrossRef]
  74. Kulkarni, V.; Manasa, B.M. Rhodotorula: An emerging pathogen in NICU. Perinatology 2017, 18, 77–80. [Google Scholar]
  75. Kumar, A.; Roy, P.; Rai, G.; Das, S.; Ansari, M.A. Cyberlindnera fabianii and Wickerhamomyces anomalous fungemia in newborns: An experience from a North Indian tertiary-care centre. Indian J. Med. Spéc. 2017, 8, 131–133. [Google Scholar] [CrossRef]
  76. Roy, U.; Jessani, L.G.; Rudramurthy, S.M.; Gopalakrishnan, R.; Dutta, S.; Chakravarty, C.; Jillwin, J.; Chakrabarti, A. Seven cases of Saccharomyces fungaemia related to use of probiotics. Mycoses 2017, 60, 375–380. [Google Scholar] [CrossRef] [PubMed]
  77. Gupta, M.; Mishra, A.K.; Singh, S.K. Cryptococcus laurentii fungemia in a low birth weight preterm neonate: India. J. Infect. Public. Health 2018, 11, 896–897. [Google Scholar] [CrossRef] [PubMed]
  78. Pedrosa, A.F.; Lisboa, C.; Rodrigues, A.G. Malassezia infections with systemic involvement: Figures and facts. J. Dermatol. 2018, 45, 1278–1282. [Google Scholar] [CrossRef] [PubMed]
  79. Iatta, R.; Battista, M.; Miragliotta, G.; Boekhout, T.; Otranto, D.; Cafarchia, C. Blood culture procedures and diagnosis of Malassezia furfur bloodstream infections: Strength and weakness. Med. Mycol. 2017, 56, 828–833. [Google Scholar] [CrossRef]
  80. Al-Sweih, N.; Ahmad, S.; Khan, S.; Joseph, L.; Asadzadeh, M.; Khan, Z. Cyberlindnera fabianii fungaemia outbreak in preterm neonates in Kuwait and literature review. Mycoses 2018, 62, 51–61. [Google Scholar] [CrossRef]
  81. Chen, I.T.; Chen, C.C.; Huang, H.C.; Kuo, K.C. Malassezia furfur Emergence and Candidemia Trends in a Neonatal Intensive Care Unit During 10 Years: The Experience of Fluconazole Prophylaxis in a Single Hospital. Adv. Neonatal Care 2020, 20, E3–E8. [Google Scholar] [CrossRef]
  82. Huang, C.-Y.; Peng, C.-C.; Hsu, C.-H.; Chang, J.-H.; Chiu, N.-C.; Chi, H. Systemic Infection Caused by Malassezia pachydermatis in Infants: Case Series and Review of the Literature. Pediatr. Infect. Dis. J. 2020, 39, 444–448. [Google Scholar] [CrossRef]
  83. Chow, N.A.; Chinn, R.; Pong, A.; Schultz, K.; Kim, J.; Gade, L.; Jackson, B.R.; Beer, K.D.; Litvintseva, A.P. Use of whole-genome sequencing to detect an outbreak of Malassezia pachydermatis infection and colonization in a neonatal intensive care unit-California, 2015–2016. Infect. Control. Hosp. Epidemiol. 2020, 41, 851–853. [Google Scholar] [CrossRef] [PubMed]
  84. Chowdhary, A.; Sharada, K.; Singh, P.K.; Bhagwani, D.K.; Kumar, N.; de Groot, T.; Meis, J.F. Outbreak of Dirkmeia churashimaensis Fungemia in a Neonatal Intensive Care Unit, India. Emerg. Infect. Dis. 2020, 26, 764. [Google Scholar] [CrossRef]
  85. Pandey, N.; Paul, P.; Kumar, D.; Tilak, R. Case report of a rare yeast Cyberlindnera fabianii fungemia in preterm twin neonates from North India: Diagnostic and therapeutic challenge. Indian J. Case Rep. 2020, 6, 563–565. [Google Scholar] [CrossRef]
  86. Al-Otaibi, H.; Asadzadeh, M.; Ahmad, S.; Al-Sweih, N.; Joseph, L. Papiliotrema laurentii fungemia in a premature, very low-birth-weight neonate in Kuwait successfully treated with liposomal amphotericin B. J. Med. Mycol. 2021, 31, 101123. [Google Scholar] [CrossRef]
  87. Galvis-Marín, J.C.; Giraldo-Ospina, B.; Martínez-Ríos, J.B.; Echeverri-Peláez, S. Fungemia by malassezia sympodialis in a neonatal intensive care unit in Colombia. Infectio 2021, 25, 130–134. [Google Scholar] [CrossRef]
  88. Cai, Z.; Wei, W.; Cheng, Z. Candida pelliculosa sepsis in a neonate: A casereport. J. Int. Med. Res. 2021, 49, 0300060520982804. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, Z.; Cao, Y.; Li, Y.; Chen, X.; Ding, C.; Liu, Y. Risk factors and biofilm formation analyses of hospital-acquired infection of Candida pelliculosa in a neonatal intensive care unit. BMC Infect. Dis. 2021, 21, 620. [Google Scholar] [CrossRef]
  90. Yang, Y.; Wu, W.; Ding, L.; Yang, L.; Su, J.; Wu, B. Two different clones of Candida pelliculosa bloodstream infection in a tertiary neonatal intensive care unit. J. Infect. Dev. Ctries. 2021, 15, 870–876. [Google Scholar] [CrossRef]
  91. Mpakosi, A.; Siopi, M.; Demetriou, M.; Falaina, V.; Theodoraki, M.; Meletiadis, J. Fungemia due to Moesziomyces aphidis (Pseudozyma aphidis) in a premature neonate. Challenges in species identification and antifungal susceptibility testing of rare yeasts. J. Med. Mycol. 2022, 32, 101258. [Google Scholar] [CrossRef]
  92. Asadzadeh, M.; Al-Sweih, N.; Ahmad, S.; Khan, S.; Alfouzan, W.; Joseph, L. Fatal Lodderomyces elongisporus Fungemia in a Premature, Extremely Low-Birth-Weight Neonate. J. Fungi. 2022, 8, 906. [Google Scholar] [CrossRef]
  93. da Silva, C.M.; Jucá, M.B.; Melo AS de, A.; Lima, S.L.; Galvão, P.V.M.; Macêdo, D.P.C.; Neves, R.P. Neonatal fungemia caused by Lodderomyces elongisporus: First case report in Latin America. Diagn. Microbiol. Infect. Dis. 2023, 107, 116077. [Google Scholar] [CrossRef] [PubMed]
  94. Davies, A.A.; Osuagwu, C.S.; Aikhomu, V.; Oladele, R.O.O. Cryptococcus laurentii fungaemia in a Tertiary Hospital in Nigeria: Case reports. Microbes Infect. Dis. 2023, 4, 1428–1434. [Google Scholar] [CrossRef]
  95. Samaddar, A.; Sharma, A. First case of neonatal fungemia caused by Aureobasidium melanogenum. J. Med. Mycol. 2023, 33, 101334. [Google Scholar] [CrossRef] [PubMed]
  96. Ashraf, A.A.; Karnaker, V.K.; Sreelatha, S.V.; Ramdas, S.; Nair, S.; Varma, S.R. A Rare Case of Fungaemia Due to Kodamaea ohmeri in a Neonate. Online J. Health Allied Sci. 2023, 22, 11. [Google Scholar]
  97. Kaur, H.; Chakrabarti, A. Strategies to Reduce Mortality in Adult and Neonatal Candidemia in Developing Countries. J. Fungi 2017, 3, 41. [Google Scholar] [CrossRef]
  98. McCrossan, B.; McHenry, E.; O‘Neill, F.; Ong, G.; Sweet, D.G. Selective fluconazole prophylaxis in high-risk babies to reduce invasive fungal infection. Arch. Dis. Child.-Fetal Neonatal Ed. 2007, 92, F454–F458. [Google Scholar] [CrossRef]
  99. Anaraki, M.R.; Nouri-Vaskeh, M.; Oskoei, S.A. 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, 172–179. [Google Scholar] [CrossRef]
  100. Bretagne, S.; Renaudat, C.; Desnos-Ollivier, M.; Sitbon, K.; Lortholary, O.; Dromer, F.; the Cochrane Adverse Effects Methods Group. Predisposing factors and outcome of uncommon yeast species-related fungaemia based on an exhaustive surveillance programme (2002–2014). J. Antimicrob. Chemother. 2017, 72, 1784–1793. [Google Scholar] [CrossRef]
  101. Borman, A.M.; Muller, J.; Walsh-Quantick, J.; Szekely, A.; Patterson, Z.; Palmer, M.D.; Fraser, M.; Johnson, E.M. Fluconazole Resistance in Isolates of Uncommon Pathogenic Yeast Species from the United Kingdom. Antimicrob. Agents Chemother. 2019, 63, 10–1128. [Google Scholar] [CrossRef]
  102. Lee, J.; Kim, H.-S.; Shin, S.H.; Choi, C.W.; Kim, E.-K.; Choi, E.H.; Kim, B.I.; Choi, J.-H. Efficacy and safety of fluconazole prophylaxis in extremely low birth weight infants: Multicenter pre-post cohort study. BMC Pediatr. 2016, 16, 67. [Google Scholar] [CrossRef]
  103. Iosifidis, E.; Papachristou, S.; Roilides, E. Advances in the Treatment of Mycoses in Pediatric Patients. J. Fungi. 2018, 4, 115. [Google Scholar] [CrossRef]
  104. Nunes, J.M.; Bizerra, F.C.; Carmona e Ferreira, R.; Colombo, A.L. Molecular Identification, Antifungal Susceptibility Profile, and Biofilm Formation of Clinical and Environmental Rhodotorula Species Isolates. Antimicrob. Agents Chemother. 2013, 57, 382–389. [Google Scholar] [CrossRef]
  105. Martinez, L.R.; Casadevall, A. Cryptococcus neoformans Biofilm Formation Depends on Surface Support and Carbon Source and Reduces Fungal Cell Susceptibility to Heat, Cold, and UV Light. Appl. Environ. Microbiol. 2007, 73, 4592–4601. [Google Scholar] [CrossRef]
  106. Iturrieta-González, I.A.; Padovan, A.C.B.; Bizerra, F.C.; Hahn, R.C.; Colombo, A.L. Multiple Species of Trichosporon Produce Biofilms Highly Resistant to Triazoles and Amphotericin B. PLoS ONE 2014, 9, e109553. [Google Scholar] [CrossRef]
  107. Pfaller, M.A.; Diekema, D.J.; Merz, W.G. Infections caused by non-Candida, non-Cryptococcus yeasts. Clin. Mycol. 2009, 10, 251–270. [Google Scholar]
  108. Jindal, N.; Arora, S.; Dhuria, N.; Arora, D. Cyberlindnera (Pichia) fabianii infection in a neutropenic child: Importance of molecular identification. JMM Case Rep. 2015, 2, e000033. [Google Scholar] [CrossRef]
  109. Hamal, P.; Ostransky, J.; Dendis, M.; Horváth, R.; Ruzicka, F.; Buchta, V.; Vejsova, M.; Sauer, P.; Hejnar, P.; Raclavsky, V. A case of endocarditis caused by the yeast Pichia fabianii with biofilm production and developed in vitro resistance to azoles in the course of antifungal treatment. Sabouraudia 2008, 46, 601–605. [Google Scholar] [CrossRef] [PubMed]
  110. Barchiesi, F.; Tortorano, A.M.; Di Francesco, L.F.; Cogliati, M.; Scalise, G.; Viviani, M.A. In-vitro activity of five antifungal agents against uncommon clinical isolates of Candida spp. J. Antimicrob. Chemother. 1999, 43, 295–299. [Google Scholar] [CrossRef]
  111. Chitasombat, M.N.; Kofteridis, D.P.; Jiang, Y.; Tarrand, J.; Lewis, R.E.; Kontoyiannis, D.P. Rare opportunistic (non-Candida, non-Cryptococcus) yeast bloodstream infections in patients with cancer. J. Infect. 2012, 64, 68–75. [Google Scholar] [CrossRef]
  112. Kaur, H.; Shankarnarayana, S.A.; Hallur, V.; Muralidharan, J.; Biswal, M.; Ghosh, A.K.; Ray, P.; Chakrabarti, A.; Rudramurthy, S.M. Prolonged Outbreak of Candida krusei Candidemia in Paediatric Ward of Tertiary Care Hospital. Mycopathologia 2020, 185, 257–268. [Google Scholar] [CrossRef] [PubMed]
  113. Pannanusorn, S.; Fernandez, V.; Römling, U. Prevalence of biofilm formation in clinical isolates of Candida species causing bloodstream infection. Mycoses 2012, 56, 264–272. [Google Scholar] [CrossRef] [PubMed]
  114. Rojas, F.D.; Sosa, M.d.L.A.; Fernández, M.S.; Cattana, M.E.; Córdoba, S.B.; Giusiano, G.E. Antifungal susceptibility of Malassezia furfur, Malassezia sympodialis, and Malassezia globosa to azole drugs and amphotericin B evaluated using a broth microdilution method. Med. Mycol. 2014, 52, 641–646. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, M.; Danesi, P.; James, T.Y.; Al-Hatmi, A.M.S.; Najafzadeh, M.J.; Dolatabadi, S.; Ming, C.; Liou, G.; Kang, Y.; de Hoog, S. Comparative pathogenicity of opportunistic black yeasts in Aureobasidium. Mycoses 2019, 62, 803–811. [Google Scholar] [CrossRef]
  116. Smith, D.F.Q.; Casadevall, A. Disaster Microbiology—A New Field of Study. mBio 2022, 13, e0168022. [Google Scholar] [CrossRef] [PubMed]
  117. Casadevall, A.; Kontoyiannis, D.P.; Robert, V. On the Emergence of Candida auris: Climate Change, Azoles, Swamps, and Birds. mBio 2019, 10, 10–1128. [Google Scholar] [CrossRef]
  118. Wang, Y.; Xu, J. Lodderomyces elongisporus: An emerging human fungal pathogen. PLoS Pathog. 2023, 19, e1011613. [Google Scholar] [CrossRef]
  119. Al-Obaid, K.; Ahmad, S.; Joseph, L.; Khan, Z. Lodderomyces elongisporus: A bloodstream pathogen of greater clinical significance. New Microbes New Infect. 2018, 26, 20–24. [Google Scholar] [CrossRef]
  120. Heath, J.A.; Zerr, D.M. Infections Acquired in the Nursery: Epidemiology and Control. Infect. Dis. Fetus Newborn Infant. 2006, 2006, 1179–1205. [Google Scholar]
  121. Hobi, S.; Cafarchia, C.; Romano, V.; Barrs, V.R. Malassezia: Zoonotic Implications, Parallels and Differences in Colonization and Disease in Humans and Animals. J. Fungi. 2022, 8, 708. [Google Scholar] [CrossRef]
  122. Schei, K.; Avershina, E.; Øien, T.; Rudi, K.; Follestad, T.; Salamati, S.; Ødegård, R.A. Early gut mycobiota and mother-offspring transfer. Microbiome. BioMed 2017, 5, 107. [Google Scholar] [CrossRef]
  123. Golubkova, A.; Hunter, C.J. Development of the Neonatal Intestinal Barrier, Microbiome, and Susceptibility to NEC. Microorganisms 2023, 11, 1247. [Google Scholar] [CrossRef] [PubMed]
  124. Fan, Y.; Wu, L.; Zhai, B. The mycobiome: Interactions with host and implications in diseases. Curr. Opin. Microbiol. 2023, 75, 102361. [Google Scholar] [CrossRef]
  125. O‘Grady, N.P.; Alexander, M.; Burns, L.A.; Dellinger, E.P.; Garland, J.; Heard, S.O.; Lipsett, P.A.; Masur, H.; Mermel, L.A.; Pearson, M.L.; et al. Summary of Recommendations: Guidelines for the Prevention of Intravascular Catheter-related Infections. Clin. Infect. Dis. 2011, 52, 1087–1099. [Google Scholar] [CrossRef] [PubMed]
  126. Fu, J.; Ding, Y.; Jiang, Y.; Mo, S.; Xu, S.; Qin, P. Persistent candidemia in very low birth weight neonates: Risk factors and clinical significance. BMC Infect. Dis. 2018, 18, 558. [Google Scholar] [CrossRef] [PubMed]
  127. Saiman, L.; Ludington, E.; Pfaller, M.; Rangel-Frausto, S.; Wiblin, R.T.; Dawson, J.; Blumberg, H.M.; Patterson, J.E.; Rinaldi, M.; Edwards, J.E. Risk factors for candidemia in Neonatal Intensive Care Unit patients. The National Epidemiology of Mycosis Survey study group. Pediatr. Infect. Dis. J. 2000, 19, 319–324. [Google Scholar] [CrossRef] [PubMed]
  128. De Rose, D.U.; Santisi, A.; Ronchetti, M.P.; Martini, L.; Serafini, L.; Betta, P.; Maino, M.; Cavigioli, F.; Cocchi, I.; Pugni, L.; et al. Invasive Candida Infections in Neonates after Major Surgery: Current Evidence and New Directions. Pathogens 2021, 10, 319. [Google Scholar] [CrossRef]
  129. Auriti, C.; De Rose, D.; Santisi, A.; Martini, L.; Ronchetti, M.; Ravà, L.; Antenucci, V.; Bernaschi, P.; Serafini, L.; Catarzi, S.; et al. Incidence and risk factors of bacterial sepsis and invasive fungal infection in neonates and infants requiring major surgery: An Italian multicentre prospective study. J. Hosp. Infect. 2022, 130, 122–130. [Google Scholar] [CrossRef]
  130. McPherson, C.; Wambach, J.A. Prevention and Treatment of Respiratory Distress Syndrome in Preterm Neonates. Neonatal Netw. 2018, 37, 169–177. [Google Scholar] [CrossRef]
  131. Cosio, T.; Pica, F.; Fontana, C.; Pistoia, E.S.; Favaro, M.; Valsecchi, I.; Zarabian, N.; Campione, E.; Botterel, F.; Gaziano, R. Stephanoascus ciferrii Complex: The Current State of Infections and Drug Resistance in Humans. J. Fungi 2024, 10, 294. [Google Scholar] [CrossRef]
  132. Garvey, M.; Rowan, N.J. Pathogenic Drug Resistant Fungi: A Review of Mitigation Strategies. Int. J. Mol. Sci. 2023, 24, 1584. [Google Scholar] [CrossRef]
  133. Brion, L.P.; Uko, S.E.; Goldman, D.L. Risk of resistance associated with fluconazole prophylaxis: Systematic review. J. Infect. 2007, 54, 521–529. [Google Scholar] [CrossRef] [PubMed]
  134. Loke, Y.K.; Price, D.; Herxheimer, A.; the Cochrane Adverse Effects Methods Group. Systematic reviews of adverse effects: Framework for a structured approach. BMC Med. Res. Methodol. 2007, 7, 32. [Google Scholar] [CrossRef]
  135. Nambiema, A.; Sembajwe, G.; Lam, J.; Woodruff, T.; Mandrioli, D.; Chartres, N.; Fadel, M.; Le Guillou, A.; Valter, R.; Deguigne, M.; et al. A Protocol for the Use of Case Reports/Studies and Case Series in Systematic Reviews for Clinical Toxicology. Front. Med. 2021, 8, 708380. [Google Scholar] [CrossRef] [PubMed]
  136. Sharma, M.; Chakrabarti, A. Candidiasis and Other Emerging Yeasts. Curr. Fungal Infect. Rep. 2023, 17, 15–24. [Google Scholar] [CrossRef]
  137. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of the systematic review.
Figure 1. Flow diagram of the systematic review.
Ijms 25 09266 g001
Figure 2. Published cases over the years.
Figure 2. Published cases over the years.
Ijms 25 09266 g002
Figure 3. Geographical distribution of the cases.
Figure 3. Geographical distribution of the cases.
Ijms 25 09266 g003
Figure 4. Geographical distribution of the cases over the decades.
Figure 4. Geographical distribution of the cases over the decades.
Ijms 25 09266 g004
Figure 5. Neonatal fungemia by rare yeast species: Published data since 1981.
Figure 5. Neonatal fungemia by rare yeast species: Published data since 1981.
Ijms 25 09266 g005
Figure 6. Reported risk factors. (Intubation + mech. Ventilation: Intubation + mechanical ventilation, RDS: Respiratory distress syndrome).
Figure 6. Reported risk factors. (Intubation + mech. Ventilation: Intubation + mechanical ventilation, RDS: Respiratory distress syndrome).
Ijms 25 09266 g006
Figure 7. Mortality rates by continent.
Figure 7. Mortality rates by continent.
Ijms 25 09266 g007
Figure 8. Mortality rates by species.
Figure 8. Mortality rates by species.
Ijms 25 09266 g008
Figure 9. Treatment of fungemia cases (from available data). Amphotericin B (AmB), Fluconazole (FLC), Voriconazole (VOR), Miconazole (MIC), Caspofungin (CAS), 5-FC (5-flucytosine), KTC (ketoconazole).
Figure 9. Treatment of fungemia cases (from available data). Amphotericin B (AmB), Fluconazole (FLC), Voriconazole (VOR), Miconazole (MIC), Caspofungin (CAS), 5-FC (5-flucytosine), KTC (ketoconazole).
Ijms 25 09266 g009
Table 1. Neonatal fungemia cases by rare opportunistic yeasts published in literature, since 1981.
Table 1. Neonatal fungemia cases by rare opportunistic yeasts published in literature, since 1981.
LocationYearCases Yeast SpeciesIdentification MethodReference
USA19811 caseMalassezia furfurNA[8]
USA19844 casesMalassezia furfurNA[9]
USA19853 casesMalassezia furfurNA[10]
UK19867 casesHansenula anomalaAPI 20C [11]
USA19875 casesMalassezia furfurNA[12]
USA19876 casesMalassezia furfurNA[13]
USA19893 casesMalassezia furfurNA[14]
BELGIUM19896 casesMalassezia furfurNA[15]
NY19911 caseMalassezia furfurNA[16]
USA19912 casesMalassezia furfurNA[17]
FRANCE19911 caseMalassezia furfurNA[18]
USA19921 caseTrichosporon beigeliiNA[19]
EUROPE19921 caseTrichosporon asteroidesNA[20]
NY19932 casesTrichosporon beigeliiBacT/Alert[21]
ATLANTA19945 casesMalassezia pachydermatisNA[22]
USA19972 casesTrichosporon beigeliiNA[23]
USA19971 caseCryptococcus laurentiiNA[24]
USA19988 casesMalassezia pachydermatisMolecular[25]
UK19981 caseTrichosporon beigeliiAPI 32 ATB [26]
CHANDIGARH199910 casesPichia anomalaNA[27]
SPAIN20002 caseSaccharomyces cerevisiaeVitek 2
API 20C
Molecular
[28]
TAIWAN20001 case Hansenula anomalaNA[29]
MALAYSIA20001 caseHansenula anomalaNA[30]
INDIA200133 casesPichia anomalaMorphology
Biochemical tests
[31]
BRAZIL20014 casesPichia anomalaNA[32]
TAIWAN20011 caseCryptococcus laurentiiMorphology
India ink
Vitek 2
[33]
SWEDEN20018 casesMalassezia pachydermatisNA[34]
EUROPE20021 caseTrichosporon asahiiAPI ID 32C[35]
TURKEY20031 caseTrichosporon asahiiNA[36]
TURKEY20033 casesTrichosporon mucoidesAPI AUX C[37]
ARGENTINA20031 case Hansenula anomalaAPI ID 32C[38]
TURKEY20041 casePichia anomalaAPI ID 32C[39]
TURKEY20051 caseSaccharomyces cerevisiaeNA[40]
QATAR20061 caseKodamaea ohmeriVitek 2
API ID 32C
Molecular
[41]
USA20061 casePichia fabianiiMolecular [42]
BRAZIL20062 casesPichia anomalaVitek 2[43]
ITALY20064 casesRhodotorula mucilaginosaVitek 2
API ID 32C
[44]
SOUTH KOREA20071 caseKodamaea ohmeriVitek 2
API20C
[45]
THAILAND20081 caseCryptococcus neoformansNA[46]
EUROPE20081 caseTrichosporon asahiiVitek 2[47]
INDIA20091 caseKodamaea ohmeriBacT/Alert 3D
API ID 32C
[48]
BRAZIL20091 caseTrichosporon spp.Morphology[49]
GREECE20091 caseCryptococcus terreusMorphology
MALDI-TOF MS Molecular
[50]
FRANCE20101 casePichia fabianiiMolecular [51]
INDIA20111 caseRhodotorula mucilaginosaMorphology API 20C [52]
ITALY20111 caseMalassezia furfurMorphology
Molecular
[53]
KUWAIT20111 caseKodamaea ohmeriVitek 2
API 20C AUX Molecular
[54]
INDIA20128 casesTrichosporon asahiiVitek 2
[55]
TAIWAN20136 casesP. anomala (C. pelliculosa)API-32C
Mini API
Molecular
[56]
SOUTH AMERICA20134 casesP. anomala (C. pelliculosa)Morphology
Molecular
[57]
GREECE20131 caseMalassezia furfurMorphology
MALDI-TOF MS Molecular
[50]
ITALY20136 casesMalassezia furfurVitek2
MALDI-TOF MS Molecular
[58]
TUNISIA20131 caseSaccharomyces cerevisiaeNA[59]
CHINA20136 casesPichia ohmeriMolecular [60]
CHINA20131 casePichia fabianiiAPI 20C AUX Molecular [61]
CHINA20131 caseKodamaea ohmeriAPI 20C AUX
Vitek 2
Molecular
[62]
INDIA201338 cases Kodamaea ohmeriMolecular [63]
INDIA20141 caseKodamaea ohmeriBacT/ALERT
Vitek2
[64]
INDIA20141 casePseudozyma aphidisAPI ID 32C
Vitek 2
Molecular
[65]
KUWAIT20141 caseMalassezia pachydermatisVitek 2
MALDI–TOF MS Molecular
[66]
INDIA20152 casesKodamaea ohmeriVitek 2
Molecular
[67]
CROATIA20152 casesCyberlindnera fabianiiMolecular [68]
ASIA20153 casesTrichosporon asahiiNA[69]
NIGERIA20151 caseMoesziomyces bullatusMolecular[70]
COLOMBIA20161 caseKodamaea ohmeriVitek 2[71]
TURKEY20171 caseWickerhamomyces anomalusVitek 2[72]
INDIA20179 casesPichia kudriavzeviiMolecular [73]
INDIA20174 casesRhodotorula glutinisNA[74]
INDIA20178 casesCyberlindnera fabianiiMolecular [75]
INDIA20171 caseWickerhamomyces anomalusMolecular [75]
INDIA20172 casesSaccharomyces cerevisiaeMolecular [76]
INDIA20181 caseCryptococcus laurentiiVitek 2[77]
PORTUGAL20181 caseMalassezia furfurMALDI–TOF MS[78]
ITALY20189 casesMalassezia furfurBacT/Alert[79]
KUWAIT201910 casesCyberlindnera fabianiiMolecular[80]
TAIWAN20191 caseMalassezia furfurBacT/Alert[81]
TAIWAN20204 casesMalassezia pachydermatisBD BACTEC FX[82]
CALIFORNIA20203 casesMalassezia pachydermatisMolecular [83]
INDIA202012 casesDirkmeia churashimaensisMolecular [84]
INDIA20202 casesCyberlindnera fabianiiMALDI-TOF-MS[85]
KUWAIT20211 casePapiliotrema laurentiiVitek 2
Molecular
[86]
COLOMBIA20211 caseMalassezia sympodialisMolecular [87]
CHINA20211 caseP. anomala (C. pelliculosa)Molecular [88]
CHINA202121 casesP. anomala (C. pelliculosa)Vitek MS[89]
CHINA202114 casesP. anomala (C. pelliculosa)Molecular [90]
GREECE20221 caseMoesziomyces aphidisVitek 2
Molecular
[91]
KUWAIT20221 caseLodderomyces elongisporusVitek 2
Molecular
[92]
BRAZIL20231 caseLodderomyces elongisporusMolecular [93]
NIGERIA20231 caseCryptococcus laurentiiVitek 2[94]
INDIA20231 caseAureobasidium melanogenumMolecular [95]
INDIA20231 caseKodamaea ohmeriVitek 2
MALDI–TOF
[96]
NA Not available, API Analytical Profile Index, MALDI-TOF Matrix-assisted laser desorption/ionization time-of-flight, MALDI-TOF MS matrix-assisted laser desorption ionization-time of flight mass spectrometry.
Table 2. Cases per decade.
Table 2. Cases per decade.
YearsCases
1981–199035
1991–200039
2001–201069
2011–2023199
Table 3. Risk factors (total cases/survival/exitus).
Table 3. Risk factors (total cases/survival/exitus).
Risk Factors
Total Cases
n = 342
NA Outcome n = 41
Survival
n = 235/301 (78 %)
Exitus
n = 66/301 (22%)
Low birth weight335/342 (98%)181/235 (77%)55/66 (84%)
Central catheters260/342 (76%) 178/235 (76%)49/66 (74%)
Prematurity226/342 (66%) 141/235 (60%)44/66 (67%)
Broad spectrum antibiotics222/342 (65%) 153/235 (65%)46/66 (70%)
Parenteral nutrition120/342 (35%) 89/235 (38%)14/66 (21%)
Intubation/mechanical ventilation116/342 (34%) 56/235 (24%)43/66 (65%)
Sepsis96/342 (28%) 63/235 (27%)28/66 (43%)
Thrombocytopenia72/342 (21%) 61/235 (26%)14/66 (21%)
Prolonged hospitalization61/342 (18%)61/235 (26%)28/66 (43%)
Respiratory distress syndrome41/342 (12%) 28/235 (12%)12/66 (19%)
Use of fluconazole27/342 (8%) 9/235 (4%)3/66 (5%)
NA (Not available) Outcome n = 41 (12%). Available Outcome n = 301 (88%).
Table 4. Antifungal susceptibility testing of species. Minimum inhibitory concentrations in (mg/L).
Table 4. Antifungal susceptibility testing of species. Minimum inhibitory concentrations in (mg/L).
YeastNumber of IsolatesAMBFLCVRCPOSITCISA5-FCAFGMFGCASMethod UsedReferences
Kodamaea ohmeri10.06432NANA0.25NA<0.002NANANAEtest [41]
10.02340.0470.0120.125 0.0320.0640.1250.25Etest[54]
38 0.25–10.5–640.03–80.06–40.06–4NANANANA0.12–1CLSI[63]
20.2540.25NANANA1NANA0.25VITEK2[67]
1<0.5NANANANANA<1NANA<0.25VITEK2[64]
Cyberlindnera/Pichia fabianii10.038NANANANA0.125NANANANCCLS[42]
10.52NANANANA0.25NANANAEtest[51]
80.75–120.5–16NANANANANANANANAEtest[75]
11≤10.125NA2NA≤4NANANACLSI[61]
2NANANANANANANA0.016–0.0641–40.125–0.19EUCAST[68]
Pichia anomala (Candida pelliculosa)20.023–0.0323–40.064–0.125NANANANANANANAEtest[43]
33NAOnly one isolate >64NANANANANANANANACLSI[31]
61–20.125NA2NANANANA0.25–0.52CLSI[56]
21<0.52–40.125–0.25NA0.125–0.25NA<4NANANACLSI[89]
Pichia kudriavzevii912–160.250.25–10.25–0.5NANA0.25–0.50.25–0.50.25–0.5CLSI[73]
Moesziomyces (Pseudozyma)
aphidis
12<0.12580.030.03≤0.016>64>8>8>8EUCAST[91]
Moesziomyces (Pseudozyma) bullatus111280.030.030.12NA64888Sensititre YeastOne Y010 microdilution method[70]
Dirkmeia (Pseudozyma) churashimaensis120.1981 to 40.03–0.1250.03–0.250.03–0.250.03–0.1250.157>8>8>8CLSI[84]
Rhodotoroula mucilaginosa40.25>2562NA2NA0.125NANANANA[44]
11.5>2560.38NANANANANANA>16Etest[52]
Cryptococcus laurentii10.254NANANANANANANANANCCLS[33]
10.252NANANANANANANANANA[77]
Cryptococcus terreus10.125–1.5128–2560.125–0.50.25–0.5NA18–328–328–328–32broth microdilution method Micronaut-AM and E-test[50]
Malassezia furfur10.190.250.0940.0940.1250.516323232MIC method on modified RPMI 1640 agar[50]
Malassezia pachydermatis10.19>2560.0120.016NANA>32NANA>32Etest[66]
AMB: Amphotericin B, FLC: Fluconazole, VRC: Voriconazole, POS: Posaconazole, ITC: Itraconazole, ISA: Isavuconazole, 5-FC: Flucytosine, AFG: Anidulafungin, MFG: Micafungin, CAS: Caspofungin, NA: Not available, MIC: minimum inhibitory concentration, CLSI: Clinical and Laboratory Standards Institute, NCCLS: National Committee for Clinical Laboratory Standards, EUCAST: European Committee on Antimicrobial Susceptibility Testing.
Table 5. Treatment administered to neonates who survived and to whom died (survival/exitus). Amphotericin B (AmB), Fluconazole (FLC), Voriconazole (VOR), Miconazole (MIC), Caspofungin (CAS), 5-FC (5-flucytosine), KTC (ketoconazole).
Table 5. Treatment administered to neonates who survived and to whom died (survival/exitus). Amphotericin B (AmB), Fluconazole (FLC), Voriconazole (VOR), Miconazole (MIC), Caspofungin (CAS), 5-FC (5-flucytosine), KTC (ketoconazole).
TreatmentSurvivalExitus
CVC removal21%3%
Fluconazole (FLC)30%46%
Voriconazole (VOR)5%3%
Miconazole (MIC)3%2%
Caspofungin (CAS)2%0% or NA
Amphotericin B (AmB)21%33%
AmB + FLC8%10%
AmB + 5-FC (5-flucytosine) 4%3%
AmB + FLC + 5-FC4%2%
AmB + KTC (ketoconazole)1%0% or NA
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mpakosi, A.; Cholevas, V.; Meletiadis, J.; Theodoraki, M.; Sokou, R. Neonatal Fungemia by Non-Candida Rare Opportunistic Yeasts: A Systematic Review of Literature. Int. J. Mol. Sci. 2024, 25, 9266. https://doi.org/10.3390/ijms25179266

AMA Style

Mpakosi A, Cholevas V, Meletiadis J, Theodoraki M, Sokou R. Neonatal Fungemia by Non-Candida Rare Opportunistic Yeasts: A Systematic Review of Literature. International Journal of Molecular Sciences. 2024; 25(17):9266. https://doi.org/10.3390/ijms25179266

Chicago/Turabian Style

Mpakosi, Alexandra, Vasileios Cholevas, Joseph Meletiadis, Martha Theodoraki, and Rozeta Sokou. 2024. "Neonatal Fungemia by Non-Candida Rare Opportunistic Yeasts: A Systematic Review of Literature" International Journal of Molecular Sciences 25, no. 17: 9266. https://doi.org/10.3390/ijms25179266

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