3,221
Views
44
CrossRef citations to date
0
Altmetric
Review

Challenging issues in neonatal candidiasis

Pages 1769-1778 | Accepted 20 Apr 2010, Published online: 01 Jun 2010

Abstract

In an era of quality improvement and ‘getting to zero (infections and/or related mortality),’ neonatal candidiasis is ripe for evidence-based initiatives. Knowledge of each institution’s invasive Candida infection (ICI) incidence and infection-related mortality is critical to evaluate disease burden and effective interventions. Evidenced-based interventions include: antifungal prophylaxis, starting with appropriate dosing, and prompt removal of central venous catheters (CVC). There is A-I evidence supporting antifungal prophylaxis with fluconazole, and it should be considered in every neonatal intensive care unit (NICU). The literature supports targeting infants <1000 g and/or ≤27 weeks, because this group has high infection-related mortality and neurodevelopmental impairment in 57% of survivors. Antifungal prophylaxis has been shown to nearly eliminate infection-related mortality. Interventions start with prenatal initiatives, with women being treated for vaginal candidiasis, especially with preterm labor or complications. Targeting modifiable risk factors, including restriction policies for use of third- and fourth-generation cephalosporins, carbapenems, H2-antagonists, proton pump inhibitors, and postnatal steroids; guidelines for CVC care and removal; and feeding practices, with promotion of early feedings and breast milk, may also reduce risk. A few studies have emerged on empiric antifungal therapy with sepsis evaluations for preterm infants <1500 g and other high-risk patients that have shown favorable effects of eliminating mortality, but these have not been compared to appropriate antifungal therapy and central line removal. Further study of empiric therapy, prospective treatment studies with higher targeted dosing of amphotericin B preparations, fluconazole, and new antifungals with prompt CVC removal may contribute to a 100% survival rate for those infants >1000 g and ≥28 weeks not receiving antifungal prophylaxis. Evaluation of ICI incidence and mortality by gestational age and birth week should be followed in each NICU, to evaluate infection control and prevention.

Introduction

A comprehensive review of invasive Candida infections (ICI) and published in the neonatal intensive care unit (NICU) has been previously presented and publishedCitation1. This follow-up review focuses on future challenges that remain in eliminating these infections and their associated mortality and neurodevelopmental impairment in the NICU.

Case 1

An intubated 24-week gestation infant at day of life 23 develops new-onset thrombocytopenia and hypotension. Cultures are obtained and nafcillin and gentamicin started. He was on a slow-feeding advance with breast milk, and was made NPO (nothing by mouth) due to an ileus with this sepsis evaluation. After 2 days of antibiotics, there is no growth from his cultures. Platelet count has decreased from 155,000 to 65,000/mL.

For almost every sepsis evaluation, empiric therapy is initiated for the most common and likely organisms. In the NICU, this varies depending on the timing of infection. For early-onset infections, the most common organisms have been group B streptococcus and Escherichia coli; therefore, ampicillin or penicillin and an aminoglycoside are most commonly administered pending culture results. For late-onset infections in very-low birth weight (VLBW, <1500 g) infants, gram-positive organisms occur in 70% of infections, with coagulase-negative staphylococcus (CoNS) species occurring 50% of the time, followed by gram-negative infections (20%) and fungal infections (10%)Citation2. Nafcillin or oxacillin with an aminoglycoside are most often initiated pending culture results. Despite CoNS being the most common organism, most centers do not start with vancomycin due to concern for resistance. It is also juxtaposed with the fact that only one in four sepsis evaluations actually yield an organismCitation2. Therefore, although CoNS is responsible for 50% of the infections, with each work up, only 12.5% of the time is it due to CoNS. Translating this into fungal infections, 2.5% (one in 40) of sepsis evaluations would be due to a fungal bloodstream infection (BSI) in VLBW infants.

Should an antifungal be used and, if so, which one should be used as empiric therapy with sepsis evaluations is an issue in neonatology, with few studies centered on this topic. Nearly all the studies have focused on the VLBW infant population. It has been proposed to use risk factors to determine how and when empiric antifungal therapy should be considered. New-onset thrombocytopenia (<100,000/mL) in VLBW infants occurs in 85% of fungemia cases and persists for a mean of >2 daysCitation3. Comparatively, 75% of gram-negative and 45% of gram-positive infections are accompanied by thrombocytopenia <100,000/mL. The fall in platelet count is 50% in fungemia cases, nearly 40% in gram-negative, and 25% in gram-positive bacteremia cases. Other studies concur that it may not be organism specific although it is common with ICICitation4,Citation5. The same authors have proposed using the following risk factors alone or in combination to determine therapy: gestational age, exposure to third-generation cephalosporins in the previous 7 days, and thrombocytopeniaCitation6. These have not been prospectively studied to date.

A few small, single-center studies where empiric antifungal therapy was used showed that Candida-related mortality was eliminated. Makhoul et al. administered empiric antifungal therapy to all VLBW infants when undergoing evaluation for late-onset sepsisCitation7. All 35 infants who developed fungal sepsis received empiric antifungal therapy and survived. In one retrospective study, empiric antifungal therapy was used in neonates who were <1500 g or considered ‘very sick’ in the NICUCitation8. Empiric antifungal therapy was started in those high-risk patients if they had (a) received vancomycin plus a third-generation cephalosporin for 7 days, and (b) had one or more of the following infection risk factors: total parenteral nutrition, mechanical ventilation, steroids, H2 blocker use, or signs of a Candida rash or thrush. Candida-related mortality occurred in 11 of 18 historic control patients who did not receive empiric therapy compared with none of six of those receiving empiric therapy.

Many factors differ among NICUs, and mortality rates also may differ among them depending on: their infected patients (how many are extremely low birth weight or have necrotizing enterocolitis [NEC]); the starting dose of antifungal therapy; immediate central line removal with treatment of proven fungal BSIs; and how aggressive the intensive care is within the unit. This last factor is hard to quantify.

The goal of empiric therapy is to attack the infection when it is believed that the number of organisms infecting a host would be lower, at around presentation compared with 24 to 48 hours later when the cultures most commonly become positiveCitation9. Empiric antifungal therapy may also work via intermittently decreasing the fungal burden in select patients. For example in VLBWs, it would be used for approximately 40 sepsis evaluations to every one culture-proven fungal infection. Its use may also decrease the fungal burden in the NICU in general and prevent possible horizontal transmission. Another issue with empiric antifungal therapy is that it is only part of the treatment for fungal BSIs, with delayed line removal contributing to mortality as well.

Finally, empiric therapy cannot be discussed alone without consideration of whether prophylaxis would be more beneficial for a patient group. The studies of fluconazole prophylaxis have both significantly reduced fungal infections, as well as nearly eliminated Candida-related mortality. At one center, Candida-related mortality was eliminated not only in the patients who received fluconazole prophylaxis, but also in the entire NICU, including those patients >1000 g who did not receive prophylaxisCitation10. The NICU used appropriate antifungal dosing of amphotericin B (AmB) and had prompt catheter removal. These investigations highlight what is lacking in studies to date: standardized treatment in terms of antifungal used, dose administered and prompt catheter removal.

What is the best evidence-based approach: waiting for positive cultures, or using empiric antifungal therapy with all sepsis evaluations in infants <1500 g, or a risk-based approach in those infants <1500 g? (). There are no randomized controlled trials (RCTs) in this area to control for the many variables affecting survival and compare prompt treatment with appropriate dosing and line removal to empiric therapy. Eliminating Candida-related mortality is critical, and since prophylaxis has proven to prevent infection and eliminate mortality and is the best approach in infants <1000 g, study of empiric therapy should be focused on those patients >1000 g, and considered in all patients who are not responding to empiric antibacterials after 48 hours and where thrombocytopenia persists and new cultures are drawn. More prospective studies are needed.

Table 1.  Should empiric antifungal therapy be administered with sepsis evaluations?.

Case 2

An intubated 36-week gestation infant with gastroschisis has been NPO, had a silo in place for 14 days, and had been receiving antibiotics for 21 days when at day of life 23 develops new-onset thrombocytopenia and hypotension. Blood culture grows Candida parapsilosis. Amphotericin B deoxycholate is started at 1 mg/kg per day, a peripheral intravenous (IV) line is placed, and the central venous catheter (CVC) is removed.

Treatment of invasive fungal infections

Treatment studies have not uniformly defined exact dosing combined with prompt CVC removal (for BSI). AmB deoxycholate (1 mg/kg) and lipid preparations (5 mg/kg) provide coverage for the most common neonatal Candida speciesCitation11–13. They are the best choices pending species identification. Most species have favorable susceptibility to AmB, and 95% to 97% of neonatal isolates are susceptible to fluconazole. Additionally, if fluconazole is being used for prophylaxis, a different antifungal should be used for treatment in that NICU.

Pharmacokinetic data have recently been published for fluconazole, micafungin, and caspofunginCitation14–23. With studies examining pharmacokinetic data targeting comparable levels in adults, neonatal data are needed regarding safety. As new dosing information has emerged, there are also case reports of possible adverse effects of these medications. Conversely, new studies have added safety data for AmB deoxycholate and have demonstrated a lack of significant nephrotoxicity; they reiterate the need for careful monitoring and adjustment of intake of sodium and potassium and monitoring renal functionCitation24–26. Studies of liposomal AmB products indicate that 5 mg/kg should be the starting dose and administered over 2 hoursCitation27,Citation28. Recent publications of fluconazole demonstrate that a dose of 12 mg/kg is comparable to adult dosing; however, the safety of this dosing has not yet been studied in neonatesCitation19,Citation20. The echinocandins may have effects that are unknown; monitoring of liver function tests, electrolytes, blood urea nitrogen, creatinine, calcium, phosphorus, and magnesium should be done daily for several days initially and then periodically if normal throughout treatment. Regarding caspofungin dosing, neonatal clinical studies had used 1 to 2 mg/kg, but pharmacokinetic data indicate that 2.5 mg/kg should be used. For micafungin dosing, pharmacokinetic data suggest a dose of 10 mg/kgCitation21–23,Citation29,Citation30. A micafungin RCT is currently suspended for further pharmacodynamic studies (www.Clinicaltrials.gov Identifier: NCT00815516). Echinocandins may have higher minimum inhibitory concentrations (MICs) with C parapsilosis; speciation and susceptibility testing should be performed with their use in neonatesCitation31. The severity of ICI with its high mortality and morbidity in many cases has lead to the need to better study the current antifungals, new dosing information, and new antifungals for neonates. As mentioned in other sections, the need to standardize central line removal with candidemia in studies of antifungals for treatment is critical to interpret outcomes.

What is the best antifungal for treatment? AmB deoxycholate, AmB lipid preparations, and fluconazole are the most commonly used antifungals in neonates, but head-to-head data have not demonstrated a definitive superior agent. Data are limited in standard dosing, starting with the highest safe dose on day 1 of treatment, and use with prompt CVC removal with fungemia. A-II evidence supports AmB deoxycholate starting at 1 mg/kg per day, with a recent study demonstrating safety when increasing up to 1.5 mg/kg if neededCitation32. Lipid preparations need a similar approach, starting at 5 mg/kg so underdosing does not affect efficacy, and one study has examined increasing to 7 mg/kgCitation33,Citation34. Fluconazole dosing of 6 mg/kg has been studied for efficacy and safety; use of higher dosing should be studied for its safety and effect on resistance patterns. The major concern would be the effect of higher dosing on liver function. The dosing of 6 mg/kg achieves blood levels higher than the average area under the curve (AUC)/MICs of most Candida species affecting neonates.

The newer echinocandins are still undergoing extensive study on dosing and safety. Observational studies and pharmacokinetic studies are emerging, but to date there are no RCTs comparing these to other antifungals. There are data with caspofungin in neonates using 1 mg/kg and 2 mg/kg, but new data support higher dosing of 2.5 mg/kgCitation14,Citation15,Citation23. Micafungin has been studied at doses up to 15 mg/kg, but only in a few patients and with limited safety data. There are limited data to guide dosing of anidulafungin. Neonatal data are needed for all the echinocandins in four major areas: optimal dose, resistance patterns with regards to C parapsilosis, safety, and central nervous system penetrationCitation31,Citation35,Citation36. Their role in treatment or prophylaxis will depend on these findings and the general principle to use one agent for prophylaxis and a different agent for treatment. Their use currently should be in consultation with pediatric infectious disease specialists or as part of research studies.

Central venous catheter removal with fungemia

Multiple studies have consistently demonstrated improved outcomes with prompt CVC removal (A-II evidence)Citation12,Citation13,Citation37,Citation38. Removal and use of peripheral IV access until there is documented clearance from the blood with three or more negative blood cultures is supported in the literatureCitation38. In cases where maintaining central venous access is critical to life-saving measures, removal and replacement at a different site should be done promptly as a second optionCitation38.

Case 2: continued

Blood cultures remain positive for 5 consecutive days. Consultation occurs with pediatric infectious diseases on whether increasing AmB deoxycholate to 1.5 mg/kg, changing to one of the lipid amphotericin preparations, adding fluconazole, or adding one of the new echinocandins would help clear the candidemia, or if there might be a fungal abscess that was undetected in the initial screening for end-organ dissemination.

Screening and re-screening for end-organ dissemination

Most studies report an average time to clearance of 3 to 5 days. Re-screening for end-organ dissemination if cultures are still positive after 5 days of antifungal therapy is recommended. While adding a second antifungal agent could be considered, no prospective study has examined this approach to determine if clearance occurs earlierCitation9,Citation37,Citation38.

Combination therapy

There are limited data in neonates examining combination therapy. Linder et al. reported the use of a second antifungal agent if one of the following was present: (1) fungal sepsis with any abscess; (2) positive urine culture; and/or (3) 10 days of persistently positive culturesCitation34. Patients were treated for 14 days or more after negative culture until radiographic resolution of abscess, if present. Overall mortality was 15%. Fungal clearance occurred in 36 (67%) with monotherapy and 52 (96%) with polytherapy. Amphotericin (deoxycholate or a lipid preparation) was used as primary treatment, with fluconazole added as the second agent in most casesCitation34. Similar to treatment data, combination therapy studies need to include data on prompt central line removal. There is a paucity of neonatal data in this area in general, with none of it controlling for a second agent, dosing, or prompt CVC removal.

Case 3

A 25-week infant is born via spontaneous vaginal delivery after premature rupture of membranes 7 days prior to delivery, followed by preterm labor not stopped by tocolytics. Infant is intubated in the delivery room and umbilical catheters placed in the NICU. Fluconazole prophylaxis is started on day of life 1. On day of life 4, a peripherally inserted central catheter (PICC) is placed and umbilical lines removed. Full enteral feeds are reached on day 24; PICC is removed and fluconazole prophylaxis is discontinued.

Prevention

With single and multicenter studies, meta-analyses, and Cochrane review, and the American Academy of Pediatrics (AAP) and the Infectious Diseases Society of America (IDSA) recommendation that there is A-I evidence supporting the use of fluconazole prophylaxis, every NICU should examine its high-risk population and institute prevention measures in an effort to ‘get to zero’ -- ‘zero’ infections and ‘zero’ mortalityCitation12,Citation13,Citation39. Incorporating known risk factors into an infection control medication stewardship, early feeding, CVC bundles, and antifungal prophylaxis in infants <1000 g can nearly eliminate invasive fungal infections and achieve zero fungal-related mortality.

Preventative strategies can be divided into these main areas: prenatal detection and eradication of maternal vaginal infections, medication, feeding and CVC stewardship in the NICU, and antifungal prophylaxis.

Prenatal detection and eradication of maternal vaginal candidiasis

While most prenatal screening is for viral and bacterial infections, pregnancies complicated by preterm labor or possible delivery should consider screening and treatment of maternal vaginal candidiasisCitation40,Citation41. This can be a simple approach to eradication of a pathogen that commonly colonizes preterm infants and in one third of cases leads to invasive infectionsCitation42.

Neonatal medication and feeding stewardship

Three medication practices have been associated with an increased risk of ICI: use of broad-spectrum antibiotics (third- or fourth-generation cephalosporins, carbapenems), acid inhibitors (H2 blockers, proton pump inhibitors), and postnatal steroidsCitation38,Citation43–46. Use of these medications during the high-risk period of infection for preterm infants when they have central venous lines, are not on full enteral feedings, and/or are intubated should be restricted to documented gram-negative infections, gastritis (limit to 3 days), and severe lung disease, respectively. Each NICU establishing guidelines with respect to medication stewardship will lead to the improved compliance. Initial antibiotic steps include using an aminoglycoside instead of a cephalosporin for empiric coverage during sepsis evaluations, and use with documented gram-negative infections, meningitis or resistant infections. Regarding postnatal steroids, dexamethasone has been associated with ICI, while hydrocortisone prophylaxis in one multicenter RCT did not increase ICI but that high-risk group of intubated infants <1000 g had a rate of fungal BSI of 10%Citation46,Citation47.

Having feeding protocols and promoting breast milk feedings may aid in the prevention of NEC, which is associated with a high rate of fungal infections of 16.5%Citation37,Citation48. Early feedings started in the first 3 days of life if possible in hemodynamically stable infants is associated with lower rates of fungal infections in infants <1000 gCitation38,Citation45. Early feeding may or may not be feasible in all patients, but in those who are stable, feeding, preferentially with breast milk, will help establish a more favorable and diverse microbiome, thereby helping to prevent fungal proliferation and dissemination.

While these risk factors are associated with increased rates of infection, they have not been definitely proven or subjected to prospective study or RCTs in neonates. Prospective studies of the effect of risk factor reduction on infection would be beneficial.

Central venous catheter management (‘bundles’)

Currently the placement and management of CVCs, with attention to team work, sterile practices, and hub and dressing care, have become a focus of many collaboratives (such as the New York State California Perinatal Quality Care Collaborative http://www.cpqcc.org/quality_improvement/qi_toolkits/hospital_acquired_infection_prevention_rev_march_2008) and for many NICUs, with the emergence of evidence-based data beginning to appear in the literatureCitation49–53. There are many differences between these bundles, for example, the use or not of chlorhexidine and the use of disks and their safety in preterm infants and other neonatesCitation54–57. Publication of the results of these and different central line bundles is critical to our understanding of how to prevent central line-associated bloodstream infection (CLABSI). It is also critical for these studies and the quality care collaborative to report their infection rates and effect on specific infections (CoNS, gram-positive, gram-negative, and Candida BSIs). The study by Aly et al. demonstrates significant reduction in all CLABSIs and elimination of Candida infectionsCitation49. Most importantly, their central line bundle includes antifungal prophylaxis in addition to placement and maintenance interventions. Other studies have lacked the necessary breakdown by type of infections.

While the contribution of CLABSI prevention bundles hopefully will contribute to reducing fungal CLABSIs, it is critical to understand that not all Candida infections are catheter related; the majority disseminate from other sites, such as the respiratory and gastrointestinal tract, the urinary tract, and the skin ().

Table 2.  Site of action of antifungal prevention strategies.

Antifungal prophylaxis

In this area, there is A-I evidence from single and multicenter RCTs of fluconazole prophylaxis demonstrating efficacy without adverse effects or the development of significant antifungal resistanceCitation10,Citation58–69. Its strongest effect is when it is targeted to high-risk patients <1000 g and begun in the first 2 days of lifeCitation39. Dosing of 3 mg/kg twice-a-week until IV (central or peripheral) access is no longer needed should be used based on its efficacy, safety, and resistance data. This targeted group of preterm infants <1000 g has the greatest number of risk factors, highest Candida-related mortality, and 57% neurodevelopmental impairment in survivorsCitation38,Citation70,Citation71. Both the AAP and the IDSA have statements supporting its use in preterm infants <1000 gCitation12,Citation13.

Currently there are 14 studies of fluconazole prophylaxis in over 3100 patients consistently demonstrating efficacy and safety, with an overall reduction in ICI by >80% and near elimination of Candida-related mortalityCitation10,Citation58–69. Subanalysis demonstrates that there is also significant efficacy in the smallest infants (both when <750 g and <1000 g are examined) and youngest infants (<27 weeks) of approximately 90%Citation39 (). Safety and resistance have been studied as well, and detailed in recent reviewsCitation1,Citation39. No significant adverse effects have been documented, and no significant emergence of resistance due to prophylaxis has been noted, both short term for individual patients or long term for NICUsCitation10,Citation39,Citation41,Citation58–69,Citation72.

Table 3.  Comparison of antifungal prophylaxis agents.

Nystatin prophylaxis has been studied in one RCT and three retrospective or epidemiologic studies. Nystatin was the first antifungal studied in preterm infants. Sims et al. examined its use in an RCT of 67 intubated infants <1250 g, and found a decrease in BSI and urinary tract infectionsCitation73. Ozturk et al. in a quasi-randomized prospective study demonstrated that antifungal prophylaxis was significantly more effective in decreasing fungal BSI when started in the first 48 hours after birth (by 90% to 3.6%) compared with screening for colonization and targeting that subgroup (by 62% to 14%)Citation74. In a retrospective study of infants <1500 g (n = 1055) in the United Kingdom, nystatin prophylaxis was associated with a 54% decrease in fungal BSI (5.5% to 2.5%, p < 0.0001)Citation75. Nystatin prophylaxis was not given to infants with peritonitis or NEC. In an epidemiologic study (1993–2006; Australia and New Zealand), oral nystatin prophylaxis decreased BSI or meningitis by 56% in <1500 g infants (1.23% to 0.54%, p < 0.0001) and 54% in <1000 g infants (2.67% to 1.23%, p < 0.0001)Citation76.

Many comparisons between fluconazole and nystatin prophylaxis remain unanswered (). One recent RCT compared oral fluconazole to nystatin prophylaxis started in the first 7 days in infants <1500 g until they reached full enteral feedingsCitation69. Both were given orally. ICI occurred in two of 38 (5.3%) fluconazole-treated infants compared with 6 of 42 (14.3%) nystatin-treated infants. All-cause mortality was zero in the fluconazole-treated group compared with six of 42 (7.5%) in the nystatin-treated group (p = 0.03). Of the six deaths, three were due to NEC, one was due to focal bowel perforation, and two were sepsis-related (including one Candida-related death), and raised the question of whether nystatin is safe to give enterally to extremely preterm infants when they are advancing on feeds. These safety data are lacking among most of the nystatin epidemiologic data, and need to be included. Bloody stool has been reported as an adverse gastrointestinal reaction to nystatin. The oral suspension of nystatin contains a high concentration of sucrose (50% by weight) and a very high osmolality of 1893 mOsm/kg and osmolarity of 3002 mOsm/LCitation77,Citation78. Comparatively, oral fluconazole has an osmolarity of 300 mOsm/L. In the neonatal literature, there are several reports of the association of hyperosmolar oral medications and NECCitation77,Citation79. The authors noted that the amounts of nystatin given to the infants were small, and the high concentration of sucrose might have altered the intestinal flora or facilitated bacterial invasion.

In all other fluconazole prophylaxis studies, fluconazole was given IV in the first weeks, and in a few studies transitioned to enteral administration when patients were on full enteral feeds without a CVC to finish up 4 or 6 weeks of prophylaxis. Fluconazole is 90% absorbed compared with nystatin, which is nonabsorbable. Because fungal infections can colonize and disseminate from multiple sites, including the skin, gastrointestinal tract, respiratory tract, and/or IV lines (central or peripheral), enteral fluconazole would not cover central line colonization and dissemination and would only achieve 90% of IV levels. This may explain the higher rate of ICI with enteral fluconazole in this study compared with the IV fluconazole prophylaxis studies.

The widespread institution and use of antifungal prophylaxis in high-risk preterm infants needs to be instituted due to the vulnerability of this population. The AAP and IDSA are helping to move this forward, targeting antifungal prophylaxis in high-risk preterm infants <1000 g. The prevention of group B streptococcus infections met similar debates over variability of rates in different countries and NICUs, number needed to treat, effect on mortality, and long-term neurodevelopmental outcomes. In extremely preterm (<28 weeks gestation) and extremely low birth weight infants, multiple hurdles exist for survival without neurodevelopmental impairment, which include not only invasive fungal infections, but intraventricular hemorrhage, periventricular leukomalacia, bacterial infections, NEC, focal bowel perforation, bronchopulmonary dysplasia, patent ductus arteriosus, and parental education level. To significantly improve our outcomes for this vulnerable population, multiple interventions are needed as a ‘bundle’ to increase survival without disability and antifungal prophylaxis is a vital part of this. In preventing ICI, for which there is A-I evidence for fluconazole prophylaxis, ICI can be eliminated as a cause of mortality or neurodevelopmental impairment for our patients born today. Delaying prevention adds risk to these preterm infants of developing an ICI, dying, or surviving with disability in the face of A-I evidence. It is important for NICUs and infection control and prevention societies or groups to examine the magnitude of its effect on NICUs regionally and nationally. For the United States, there are nearly 30,000 preterm infants born each year <1000 g or ≤27 weeks (National Vital Statistics, Centers for Disease Control and Prevention, 2007) which translates into antifungal prophylaxis preventing 2000 to 3000 ICI, 200 to 300 Candida-related deaths, and 400 to 500 survivors with neurodevelopmental impairment each year.

Each year we wait denies this A-I level of evidence from benefiting our patients. All agree that there is significant efficacy in preventing ICI with the use of antifungal prophylaxis; some of the debate focuses on whether fluconazole or nystatin would be betterCitation39,Citation80,Citation81. The data overwhelmingly point to fluconazole for prophylaxis (). To have the greatest effect and target the highest risk period, antifungal prophylaxis should be started in the first 2 days of life and continue until IV access (central or peripheral) is no longer needed. Future research may test the hypothesis of whether the combination of IV fluconazole with enteral nystatin can get us to zero infections.

Table 4.  Preventative strategies and levels of evidence.

Conclusions

Treatment and prevention can help get us close to zero ICI and zero Candida-related mortality (). Further studies need to address treatment of infection in conjunction with central line removal, optimal dosing of older (AmB preparations, fluconazole) and new antifungals (echinocandins), empiric therapy, and the effect of central line bundles on all ICI. We need to move forward now, as the benefit of prophylaxis outweighs the risks, and a significant number of infants each year continue to develop ICI, suffer neurodevelopmental impairment, or die when we have A-I evidence for antifungal prophylaxis. Evaluation of these strategies should continue to be assessed by individual, larger group, and national NICU epidemiologic data.

Table 5.  Top 10 ways to ‘get to zero’ invasive Candida infections and infection-related mortality.

Transparency

Declaration of funding

This activity is supported by an educational grant from Merck & Co., Inc.

Declaration of financial/other relationships

D.A.K. has disclosed that he has no real or apparent conflicts of interest to report.

Some peer reviewers receive honoraria from CMRO for their review work. Peer reviewer 1 has disclosed that he/she has received research grants from Children’s Cancer Foundation of Baltimore and is on speakers bureaus of Enzon Pharmaceuticals and Sigma Tau Pharmaceuticals. Peer reviewer 2 has no relevant financial relationships to disclose.

Acknowledgment

The authors of this supplement thank Global Education Exchange, LLC, for editorial support.

References

  • Kaufman DA. Neonatal candidiasis: clinical manifestations, management, and prevention strategies. J Pediatr 2010;156:S47-52
  • Stoll BJ, Hansen N, Fanaroff AA, et al. Late-onset sepsis in very low birth weight neonates: the experience of the NICHD Neonatal Research Network. Pediatrics 2002;110:285-91
  • Guida JD, Kunig AM, Leef KH, et al. Platelet count and sepsis in very low birth weight neonates: is there an organism-specific response?. Pediatrics 2003;111:1411-15
  • Manzoni P, Mostert M, Galletto P, et al. Is thrombocytopenia suggestive of organism-specific response in neonatal sepsis?. Pediatr Int 2009;51:206-10
  • Bhat MA, Bhat JI, Kawoosa MS, et al. Organism-specific platelet response and factors affecting survival in thrombocytopenic very low birth weight babies with sepsis. J Perinatol 2009;29:702-8
  • Benjamin DK Jr, DeLong ER, Steinbach WJ, et al. Empirical therapy for neonatal candidemia in very low birth weight infants. Pediatrics 2003;112:543-7
  • Makhoul IR, Kassis I, Smolkin T, et al. Review of 49 neonates with acquired fungal sepsis: further characterization. Pediatrics 2001;107:61-6
  • Procianoy RS, Eneas MV, Silveira RC. Empiric guidelines for treatment of Candida infection in high-risk neonates. Eur J Pediatr 2006;165:422-3
  • Schelonka RL, Moser SA. Time to positive culture results in neonatal Candida septicemia. J Pediatr 2003;142:564-5
  • Healy CM, Campbell JR, Zaccaria E, et al. Fluconazole prophylaxis in extremely low birth weight neonates reduces invasive candidiasis mortality rates without emergence of fluconazole-resistant Candida species. Pediatrics 2008;121:703-10
  • Baley JE. Neonatal candidiasis: the current challenge. Clin Perinatol 1991;18:263-80
  • Candidiasis. In: Pickering L, Kimberlin DW, Baker CJ, Long SS, eds. Red Book: 2009 Report of the Committee on Infectious Diseases. Elk Grove Village, IL: American Academy of Pediatrics, 2009:245-9
  • Pappas PG, Kauffman CA, Andes D, et al. Clinical practice guidelines for the management of candidiasis: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis 2009;48:503-35
  • Odio CM, Araya R, Pinto LE, et al. Caspofungin therapy of neonates with invasive candidiasis. Pediatr Infect Dis J 2004;23:1093-7
  • Natarajan G, Lulic-Botica M, Rongkavilit C, et al. Experience with caspofungin in the treatment of persistent fungemia in neonates. J Perinatol 2005;25:770-7
  • Yalaz M, Akisu M, Hilmioglu S, et al. Successful caspofungin treatment of multidrug resistant Candida parapsilosis septicaemia in an extremely low birth weight neonate. Mycoses 2006;49:242-5
  • Smith PB, Steinbach WJ, Cotten CM, et al. Caspofungin for the treatment of azole resistant candidemia in a premature infant. J Perinatol 2007;27:127-9
  • Natale F, Castronovo A, Regoli D, et al. Successful treatment with caspofungin of refractory Candida krusei candidemia in a very low birth weight preterm infant. Pediatr Infect Dis J 2009;28:452
  • Wade KC, Wu D, Kaufman DA, et al. Population pharmacokinetics of fluconazole in young infants. Antimicrob Agents Chemother 2008;52:4043-9
  • Wade KC, Benjamin DK Jr, Kaufman DA, et al. Fluconazole dosing for the prevention or treatment of invasive candidiasis in young infants. Pediatr Infect Dis J 2009;28:717-23
  • Smith PB, Walsh TJ, Hope W, et al. Pharmacokinetics of an elevated dosage of micafungin in premature neonates. Pediatr Infect Dis J 2009;28:412-15
  • King KY, Edwards MS, Word BM. Hepatitis associated with micafungin use in a preterm infant. J Perinatol 2009;29:320-2
  • Saez-Llorens X, Macias M, Maiya P, et al. Pharmacokinetics and safety of caspofungin in neonates and infants less than 3 months of age. Antimicrob Agents Chemother 2009;53:869-75
  • Le J, Adler-Shohet FC, Nguyen C, et al. Nephrotoxicity associated with amphotericin B deoxycholate in neonates. Pediatr Infect Dis J 2009;28:1061-3
  • Holler B, Omar SA, Farid MD, et al. Effects of fluid and electrolyte management on amphotericin B-induced nephrotoxicity among extremely low birth weight infants. Pediatrics 2004;113:e608-6
  • Turcu R, Patterson MJ, Omar S. Influence of sodium intake on amphotericin B-induced nephrotoxicity among extremely premature infants. Pediatr Nephrol 2009;24:497-505
  • Wurthwein G, Groll AH, Hempel G, et al. Population pharmacokinetics of amphotericin B lipid complex in neonates. Antimicrob Agents Chemother 2005;49:5092-8
  • Kotwani RN, Gokhale PC, Bodhe PV, et al. A comparative study of plasma concentrations of liposomal amphotericin B (L-AMP-LRC-1) in adults, children and neonates. Int J Pharm 2002;238:11-15
  • Natarajan G, Lulic-Botica M, Aranda JV. Refractory neonatal candidemia and high-dose micafungin pharmacotherapy. J Perinatol 2009;29:738-43
  • Benjamin DK Jr, Smith PB, Arrieta A, et al. Safety and pharmacokinetics of repeat-dose micafungin in young infants. Clin Pharmacol Ther 2010;87:93-9
  • Silva AP, Miranda IM, Lisboa C, et al. Prevalence, distribution, and antifungal susceptibility profiles of Candida parapsilosis, C.orthopsilosis, and C. metapsilosis in a tertiary care hospital. J Clin Microbiol 2009;47:2392-7
  • Jeon GW, Koo SH, Lee JH, et al. A comparison of AmBisome to amphotericin B for treatment of systemic candidiasis in very low birth weight infants. Yonsei Med J 2007;48:619-26
  • Juster-Reicher A, Flidel-Rimon O, Amitay M, et al. High-dose liposomal amphotericin B in the therapy of systemic candidiasis in neonates. Eur J Clin Microbiol Infect Dis 2003;22:603-7
  • Linder N, Klinger G, Shalit I, et al. Treatment of candidaemia in premature infants: comparison of three amphotericin B preparations. J Antimicrob Chemother 2003;52:663-7
  • Petraitiene R, Petraitis V, Hope WW, et al. Cerebrospinal fluid and plasma (1–>3)-beta-D-glucan as surrogate markers for detection and monitoring of therapeutic response in experimental hematogenous Candida meningoencephalitis. Antimicrob Agents Chemother 2008;52:4121-9
  • 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;197:163-71
  • Chapman RL, Faix RG. Persistently positive cultures and outcome in invasive neonatal candidiasis. Pediatr Infect Dis J 2000;19:822-7
  • Benjamin DK Jr, 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;117:84-92
  • Kaufman DA. Fluconazole prophylaxis: can we eliminate invasive Candida infections in the neonatal ICU?. Curr Opin Pediatr 2008;20:332-40
  • Waggoner-Fountain LA, Walker MW, Hollis RJ, et al. Vertical and horizontal transmission of unique Candida species to premature newborns. Clin Infect Dis 1996;22:803-8
  • Freydiere AM, Piens MA, Andre JM, et al. Successful treatment of Candida glabrata peritonitis with fluconazole plus flucytosine in a premature infant following in vitro fertilization. Eur J Clin Microbiol Infect Dis 2005;24:704-5
  • Kaufman D, Fairchild KD. Clinical microbiology of bacterial and fungal sepsis in very-low-birth-weight infants. Clin Microbiol Rev 2004;17:638-80
  • Cotten CM, McDonald S, Stoll B, et al. The association of third-generation cephalosporin use and invasive candidiasis in extremely low birth-weight infants. Pediatrics 2006;118:717-22
  • Makhoul IR, Bental Y, Weisbrod M, et al. Candidal versus bacterial late-onset sepsis in very low birthweight infants in Israel: a national survey. J Hosp Infect 2007;65:237-43
  • Saiman L, Ludington E, Pfaller M, et al. Risk factors for candidemia in neonatal intensive care unit patients. The National Epidemiology of Mycosis Survey study group. Pediatr Infect Dis J 2000;19:319-24
  • Stoll BJ, Temprosa M, Tyson JE, et al. Dexamethasone therapy increases infection in very low birth weight infants. Pediatrics 1999;104:e63
  • Watterberg KL, Gerdes JS, Cole CH, et al. Prophylaxis of early adrenal insufficiency to prevent bronchopulmonary dysplasia: a multicenter trial. Pediatrics 2004;114:1649-57
  • Noyola DE, Fernandez M, Moylett EH, et al. Ophthalmologic, visceral, and cardiac involvement in neonates with candidemia. Clin Infect Dis 2001;32:1018-23
  • Aly H, Herson V, Duncan A, et al. Is bloodstream infection preventable among premature infants? A tale of two cities. Pediatrics 2005;115:1513-18
  • Garland JS, Henrickson K, Maki DG. The 2002 Hospital Infection Control Practices Advisory Committee Centers for Disease Control and Prevention guideline for prevention of intravascular device-related infection. Pediatrics 2002;110:1009-13
  • Curry S, Honeycutt M, Goins G, et al. Catheter-associated bloodstream infections in the NICU: getting to zero. Neonatal Netw 2009;28:151-5
  • Schulman J, Wirtschafter DD, Kurtin P. Neonatal intensive care unit collaboration to decrease hospital-acquired bloodstream infections: from comparative performance reports to improvement networks. Pediatr Clin North Am 2009;56:865-92
  • Schulman J, Stricof RL, Stevens TP, et al. Development of a statewide collaborative to decrease NICU central line-associated bloodstream infections. J Perinatol 2009;29:591-9
  • Garland JS, Uhing MR. Strategies to prevent bacterial and fungal infection in the neonatal intensive care unit. Clin Perinatol 2009;36:1-13
  • Garland JS, Alex CP, Mueller CD, et al. A randomized trial comparing povidone-iodine to a chlorhexidine gluconate-impregnated dressing for prevention of central venous catheter infections in neonates. Pediatrics 2001;107:1431-6
  • Visscher M, deCastro MV, Combs L, et al. Effect of chlorhexidine gluconate on the skin integrity at PICC line sites. J Perinatol 2009;29:802-7
  • Garland JS, Alex CP, Uhing MR, et al. Pilot trial to compare tolerance of chlorhexidine gluconate to povidone-iodine antisepsis for central venous catheter placement in neonates. J Perinatol 2009;29:808-13
  • Kaufman D, Boyle R, Hazen KC, et al. Fluconazole prophylaxis against fungal colonization and infection in preterm infants. N Engl J Med 2001;345:1660-6
  • Kicklighter SD, Springer SC, Cox T, et al. Fluconazole for prophylaxis against candidal rectal colonization in the very low birth weight infant. Pediatrics 2001;107:293-8
  • Kaufman D, Boyle R, Hazen KC, et al. Twice weekly fluconazole prophylaxis for the prevention of candidemia in preterm infants <1000 grams. J Pediatr 2005;147:172-9
  • Bertini G, Perugi S, Dani C, et al. Fluconazole prophylaxis prevents invasive fungal infection in high-risk, very low birth weight infants. J Pediatr 2005;147:162-5
  • Healy CM, Baker CJ, Zaccaria E, et al. Impact of fluconazole prophylaxis on incidence and outcome of invasive candidiasis in a neonatal intensive care unit. J Pediatr 2005;147:166-71
  • Manzoni P, Arisio R, Mostert M, et al. Prophylactic fluconazole is effective in preventing fungal colonization and fungal systemic infections in preterm neonates: a single-center, 6-year, retrospective cohort study. Pediatrics 2006;117:e22-32
  • Uko S, Soghier LM, Vega M, et al. Targeted short-term fluconazole prophylaxis among very low birth weight and extremely low birth weight infants. Pediatrics 2006;117:1243-52
  • Aghai ZH, Mudduluru M, Nakhla TA, et al. Fluconazole prophylaxis in extremely low birth weight infants: association with cholestasis. J Perinatol 2006;26:550-5
  • Manzoni P, Stolfi I, Pugni L, et al. A multicenter, randomized trial of prophylactic fluconazole in preterm neonates. N Engl J Med 2007;356:2483-95
  • Weitkamp JH, Ozdas A, Lafleur B, et al. Fluconazole prophylaxis for prevention of invasive fungal infections in targeted highest risk preterm infants limits drug exposure. J Perinatol 2008;28:405-11
  • Aziz M, Patel AL, Losavio J, et al. Efficacy of fluconazole prophylaxis for prevention of invasive fungal infection in extremely low birth weight infants. Pediatr Infect Dis J 2010;29:352-6.
  • Violaris K, Carbone T, Bateman D, et al. Comparison of fluconazole and nystatin oral suspensions for prophylaxis of systemic fungal infection in very low birthweight infants. Am J Perinatol 2010;27:73-8
  • Johnsson H, Ewald U. The rate of candidaemia in preterm infants born at a gestational age of 23–28 weeks is inversely correlated to gestational age. Acta Paediatr 2004;93:954-8
  • Stoll BJ, Hansen NI, Adams-Chapman I, et al. Neurodevelopmental and growth impairment among extremely low-birth-weight infants with neonatal infection. JAMA 2004;292:2357-65
  • Manzoni P, Leonessa M, Galletto P, et al. Routine use of fluconazole prophylaxis in a neonatal intensive care unit does not select natively fluconazole-resistant Candida subspecies. Pediatr Infect Dis J 2008;27:731-7
  • Sims ME, Yoo Y, You H, et al. Prophylactic oral nystatin and fungal infections in very-low-birthweight infants. Am J Perinatol 1988;5:33-6
  • Ozturk MA, Gunes T, Koklu E, et al. Oral nystatin prophylaxis to prevent invasive candidiasis in neonatal intensive care unit. Mycoses 2006;49:484-92
  • Ganesan K, Harigopal S, Neal T, et al. Prophylactic oral nystatin for preterm babies under 33 weeks gestation decreases fungal colonisation and invasive fungaemia. Arch Dis Child Fetal Neonatal Ed 2009;94:F275-8
  • Howell AJ, Isaacs D, Halliday R. Oral nystatin prophylaxis and neonatal fungal infections. Arch Dis Child Fetal Neonatal Ed 2009;94:F429-33
  • Jew RK, Owen D, Kaufman D, et al. Osmolality of commonly used medications and formulas in the neonatal intensive care unit. Nutr Clin Pract 1997;12:158-63
  • Ernst JA, Williams JM, Glick MR, et al. Osmolality of substances used in the intensive care nursery. Pediatrics 1983;72:347-52
  • Mutz AE, Obladen MW. Hyperosmolar oral medication and necrotizing enterocolitis. Pediatrics 1985;75:371-2
  • Kaufman D. Fluconazole prophylaxis decreases the combined outcome of invasive Candida infections or mortality in preterm infants. Pediatrics 2008;122:1158-9
  • Isaacs D. Fungal prophylaxis in very low birth weight neonates: nystatin, fluconazole or nothing?. Curr Opin Infect Dis 2008;21:246-50