Next Article in Journal
Prevalence and Antimicrobial Resistance of Virulent Listeria monocytogenes and Cronobacter sakazakii in Dairy Cattle, the Environment, and Dried Milk with the In Vitro Application of Natural Alternative Control
Next Article in Special Issue
Neonatal Early Onset Sepsis (EOS) Calculator plus Universal Serial Physical Examination (SPE): A Prospective Two-Step Implementation of a Neonatal EOS Prevention Protocol for Reduction of Sepsis Workup and Antibiotic Treatment
Previous Article in Journal
Acinetobacter baumannii Infection-Related Mortality in Hospitalized Patients: Risk Factors and Potential Targets for Clinical and Antimicrobial Stewardship Interventions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Therapeutic Options and Outcomes for the Treatment of Neonates and Preterms with Gram-Negative Multidrug-Resistant Bacteria: A Systematic Review

Division of Pediatric Infectious Diseases, Department for Women’s and Child’s Health, University of Padua, 35128 Padua, Italy
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(8), 1088; https://doi.org/10.3390/antibiotics11081088
Submission received: 30 June 2022 / Revised: 3 August 2022 / Accepted: 7 August 2022 / Published: 10 August 2022
(This article belongs to the Special Issue Tackling Antimicrobial Resistance in Neonatal Infections)

Abstract

:
(1) Background: Infections caused by multidrug-resistant (MDR) or extensively drug-resistant (XDR) bacteria represent a challenge in the neonatal population due to disease severity and limited therapeutic possibilities compared to adults. The spread of antimicrobial resistance and drug availability differ significantly worldwide. The incidence of MDR bacteria has constantly risen, causing an increase in morbidity, mortality, and healthcare costs in both high-income (HIC) and low- and middle-income countries (LMIC). Therefore, more evidence is needed to define the possible use of newer molecules and to optimize combination regimens for the oldest antimicrobials in neonates. This systematic review aims to identify and critically appraise the current antimicrobial treatment options and the relative outcomes for MDR and XDR Gram-negative bacterial infections in the neonatal population. (2) Methods: A literature search for the treatment of MDR Gram-negative bacterial infections in neonates (term and preterm) was conducted in Embase, MEDLINE, and Cochrane Library. Studies reporting data on single-patient-level outcomes related to a specific antibiotic treatment for MDR Gram-negative bacterial infection in children were included. Studies reporting data from adults and children were included if single-neonate-level information could be identified. We focused our research on four MDROs: Enterobacterales producing extended-spectrum beta-lactamase (ESBL) or carbapenemase (CRE), Pseudomonas aeruginosa, and Acinetobacter baumannii. PROSPERO registration: CRD42022346739 (3) Results: The search identified 11,740 studies (since January 2000), of which 22 fulfilled both the inclusion and exclusion criteria and were included in the analysis. Twenty of these studies were conducted in LMIC. Colistin is the main studied and used molecule to treat Gram-negative MDR bacteria for neonate patients in the last two decades, especially in LMIC, with variable evidence of efficacy. Carbapenems are still the leading antibiotics for ESBL Enterobacterales, while newer molecules (i.e., beta-lactam agents/beta-lactamase inhibitor combination) are promising across all analyzed categories, but data are few and limited to HICs. (4) Conclusions: Data about the treatment of Gram-negative MDR bacteria in the neonatal population are heterogeneous and limited mainly to older antimicrobials. Newer drugs are promising but not affordable yet for many LMICs. Therefore, strategies cannot be generalized but will differ according to the country’s epidemiology and resources. More extensive studies are needed to include new antimicrobials and optimize the combination strategies for the older ones.

1. Background

The emergence of multidrug-resistant organism (MDRO)-associated infections also represents a global challenge for the neonatal population, and the shortage of newer antimicrobial options is a prioritized global health concern [1,2,3]. Infections contribute considerably to neonatal mortality, with sepsis accounting for 15.6% of neonatal deaths (an estimated 430,000–680,000 deaths per annum), which predominantly occur in low- and middle-income countries (LMICs) [4,5,6]. In high-income countries (HICs), neonates admitted to the neonatal intensive care unit (NICU) are at higher risk of infections [7]. Gram-negative bacteria are responsible for approximately 15–30% of late-onset infections (LOIs) in HIC, especially E. coli and Klebsiella species [8,9]. In LMIC, on the other hand, Gram-negative bacteria are the most frequently identified neonatal pathogens [10,11], and more than half are resistant to three or more classes of broad-spectrum antibiotics (Penicillin, Cephalosporins, Fluoroquinolones, and Carbapenems) [12].
Further, MDROs are colonizing neonates across NICUs with an increasing trend in both HICs and LMICs [13,14].
In Europe, third-generation cephalosporin resistance in E. coli increased up to 50% of strains in 2020, while resistance to carbapenems was as high as 25% in Enterobacterales and 50% in Acinetobacter spp.; Pseudomonas aeruginosa has gained multiple resistances against common antimicrobials, with 50% of strains resistant to three or more antibacterial categories [15]. In the same way, in the USA, the 2019 Center for Disease Control and Prevention (CDC) report designated extended-spectrum β-lactamase-producing Enterobacterales (ESBL-E), carbapenem-resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with difficult-to-treat resistance (DTR-P.A.) as urgent or serious threats [16]. In LMICs, resistance patterns are even higher, with a rate of carbapenem-resistant K. pneumoniae that, in 2019, was reported to be up to 34%, while Acinetobacter baumannii was up to 84% [17].
As a result, antibiotics consumption increased worldwide by 65% between 2000 and 2015, mainly older molecules in LMIC; however, polymyxins and tigecycline also increased, with a worrisome trend in high-income countries (HIC) [13,18].
Newer promising agents, such as beta-lactam/beta-lactamase inhibitors (βL-βLI), are studied for resistant Gram-negative infections, but studies in neonatal patients are limited [19]. Further, they are expensive compared to older molecules and are not available worldwide.
This systematic review aims to critically appraise the current antimicrobial treatment options and the relative outcomes for MDR and extensively drug-resistant (XDR) Gram-negative bacterial infections in the neonatal population.

2. Methods

2.1. Literature Search

This systematic review was carried out according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines (Figure 1). Embase, Medical Literature Analysis and Retrieval System Online (MEDLINE), and Cochrane Library were searched for relevant studies, combining Medical Subject Heading (MeSH) and free-text terms for “neonate” and “MDR” and “Outcome assessment” (complete search strategy in Additional File 2). The search strategy involved restrictions on the date (from 1 January 2000 to 7 September 2021) but not on language. All studies on children younger than 18 were searched.
This study is registered with the International Prospective Register of Systematic Reviews (PROSPERO) as record number CRD42022346739.
We included studies with any method of diagnosing MDR infections in the neonate and preterm populations; however, the reasons for defining an organism as MDR were not explicit in most studies, but we can assume the international expert proposal definition was applied [20]. Any site of infection was included. The search results were exported to Rayyan software for further manuscript assessment and handling.

2.2. Study Selection

Assessments of the titles, abstracts, and full texts were conducted independently by three investigators (L.C., M.C., and L.R.). Discussion with a fourth reviewer (D.D.) resolved any disagreement regarding study selection.

2.3. Eligibility Criteria

Eligible study designs included randomized clinical trials, observational studies, prospective or retrospective designs, concomitant or historical control studies, case series, and case reports. Meta-analyses, systematic reviews, and narrative reviews were not included. Studies investigating any antimicrobial treatment for infections caused by MDR Gram-negative bacteria were included.
Studies without pediatric data or about Gram-positive bacterial infections or drug resistance on malaria, human immunodeficiency virus (HIV), viral tuberculosis, and fungal treatment were also excluded.
The population of interest was term and preterm newborns with confirmed MDRO Gram-negative infections with clearly defined resistance that were receiving antimicrobial treatment and presenting clinical and microbiological outcomes.
The primary outcome was infection-related mortality from the initiation of treatment until discharge. Secondary outcomes were clinical success (defined as complete resolution or a substantial improvement in the signs and symptoms of the index infection) and microbiological success (measured by the suppression, eradication, or relapses of bacterial growth).
Studies published between 1 January 2000 and 7 September 2021 were included. Further details are reported in the PICOS (P: problem/patient/population; I: intervention; C: comparison/control; O: outcome; S: Study design) (Additional File 1).

2.4. Data Extraction and Assessment of Study Quality

The following data were extracted using a standardized data collection form:
  • Study characteristics (authors, year of publication, study design, study location, and country);
  • Patient characteristics (age, care setting, and inclusion and exclusion criteria);
  • Type of MDR;
  • Setting;
  • Main results with accuracy measures;
  • Health outcomes (e.g., mortality, clinical response, and microbiological eradication);
  • Main results.
Standardized predetermined study criteria were applied to all full-text documents. The selection process is presented in Figure 1.
The quality and risk of bias in individual studies were jointly assessed at the study and outcome level by all reviewing authors using the Study Quality Assessment Tool from The National Heart, Lung, and Blood Institute [21]. The quality assessment results are presented in Table 1, Table 2, Table 3 and Table 4.

2.5. Summary Measures

The following measures of treatment success were included: absolute values, absolute risk differences, hazard ratio (HR), relative risk, and odds ratio. Unadjusted and adjusted measures were included if available.

3. Results

3.1. Study Selection

A total of 11,740 records were found, and 2225 duplicate records were removed. Then, 9515 records were screened and excluded by title or abstract, and 164 were excluded after reading the full text because they did not meet the eligibility criteria, and 42 articles regarding Gram-positive bacteria were excluded. A total of twenty-two articles were included in the systematic qualitative review (Figure 1). We report the characteristics of the studies in Table 1, Table 2, Table 3 and Table 4. Figure 2 shows the studies with a relevant number of patients.

3.2. ESBL Enterobacterales

We included only one retrospective study regarding ESBL-producing Enterobacterales in neonates [22]. The mortality, clinical success, and microbiological eradication are displayed in Table 1. The study was conducted in an LMIC to observe the treatment and related outcomes of ESBL infections in neonates: a case series of 100 neonates infected with ESBL-producing Klebsiella (bloodstream infection (BSI) or meningitis) described an overall mortality of 30%, with rates of 25% if empirical therapy was undertaken with piperacillin-tazobactam and amikacin, 32% with meropenem, 42% with no empiric therapy or when other antibiotics were used [22].

3.3. Carbapenem-Resistant Enterobacterales (CRE)

We included eight articles regarding CRE (including carbapenemase-producing K. pneumoniae-KPC and XDR Enterobacterales): three case reports, two case series, and three retrospective studies [23,24,25,26,27,28,39,40]
The mortality and clinical and microbiological outcomes are presented in Table 2. The settings were different, but most studies (6/8) were conducted in LMICs.
The selected studies report the following CRE-associated clinical settings: BSI, ventilator-associated pneumonia (VAP), meningitis, and others.
The included antibiotic regimens were colistin (3/8), meropenem in high dose or association (3/8), and ceftazidime-avibactam (2/8). Matma [23] reported five neonates with CRE infection treated with colistin in monotherapy with an 80% success rate. Colistin was used in combination with meropenem (75%), amikacin (18.5%), or ciprofloxacin (10.8%) in a study by Eren Çağan on 40 neonates [25]. Ceftazidime-avibactam showed efficacy and tolerability as a salvage therapy in six patients with KPC from a bloodstream infection. [29].

3.4. Pseudomonas aeruginosa

We included four articles regarding DTR-Pseudomonas aeruginosa (DTR-PA): two case series, one prospective study, and one retrospective study [23,31,32,33]
The mortality, clinical success, and microbiological outcomes are presented in Table 3. All studies were conducted in LMICs. The selected studies report the following infections: BSI, VAP, urine, and others.
Colistin, in association with other antimicrobials, remains the most described antimicrobial for DTR and XDR-PA. However, Belet et al. described the use of ciprofloxacin in 30 infants from a NICU with PA susceptible only to quinolones, with a clinical success of 28 out of 30 [32].
New drugs such as ceftazidime/avibactam or ceftolozane–tazobactam have not been studied in neonates for DTR-PA.

3.5. Acinetobacter Baumannii

We identified nine studies about the treatment of XDR A. baumannii (XDR-AB): one prospective study, one case report, one case series, and six retrospective studies [23,25,31,33,34,35,36,37,38].
The study characteristics and treatment outcomes are summarized in Table 4. All studies were conducted in LMICs. The selected studies reported as the site of infection BSI, VAP, meningitis, and others.
Colistin is the most represented antimicrobial therapy (8/9).
One case report shows its use for CNS infections by XDR-AB, both intravenous and intraventricular [35], while when used by the intravenous route, it is mainly accompanied by other antimicrobials such as carbapenems and amikacin [23,25,36].
Colistin has shown efficacy as a nebulized therapy for MDR and XDR-AB pneumonia in neonates [37]. Cefoperazone/sulbactam was evaluated in different combinations in XDR-AB VAP in a 12-patient series [41].

3.6. Risk of Bias

Data on antimicrobial therapy in children are not conclusive. Our systematic review shows that 31 articles are case reports or case series, and 17 are retrospective studies.

4. Discussion

Our research identified twenty-two articles describing the clinical and microbiological outcomes of different treatment options for the most common MDR Gram-negative bacteria in the neonatal population.
Most studies were retrospective and analyzed the use of a specific treatment for different species and resistance patterns, so they were reported in our results more than once (e.g., colistin for CRE and P. aeruginosa). To outline clear and comparable results, we included only studies where information about the spectrum of susceptibility, antimicrobial treatment, and outcomes were detailed and quantifiable, excluding descriptive studies on the use of antibiotics in infectious syndromes but without precise microbiological isolation. However, data on the neonatal population are scant, and significant conclusions are challenging to achieve.
ESBL-producing bacteria are the most widespread MDROs, with 10% of colonized neonates becoming infected [42]. Further, in addition to cephalosporins, different Enterobacterales species can also share some genetic elements with different patterns susceptible to other antibiotics [43,44].
Extended-spectrum cephalosporins (cefotaxime and ceftriaxone) showed inadequacy in pediatrics, even with low minimum inhibitory concentrations (MICs) in vitro. The same unfavorable response was observed with an aminoglycoside in monotherapy, which is, therefore, discouraged [45].
Carbapenems remain the mainstay of treatment, with most evidence in adults [46,47,48,49] but also a high percentage in pediatrics as well, with evidence of superiority compared to other regimens (piperacillin-tazobactam and amikacin) [50]. In addition, we found a unique article concerning ESBL-producing bacteria in neonates with defined outcomes and treatment in South Africa, describing mortality rates of 25% in the empirical therapy of piperacillin-tazobactam and amikacin and 32% with meropenem; these data must be interpreted with caution since infants receiving meropenem as empiric treatment were more likely to be severely ill [22]. With neonatal sepsis, empirical therapy plays a key role, and local epidemiology must be considered. The NeoMero1 trial in 2020 found no evidence that meropenem as an empiric therapy is superior to the standard of care for neonatal sepsis and that it should be reserved for suspected Gram-negative infections and places with high rates of beta-lactamase circulation [51].
Major challenges come with DTR-Pseudomonas aeruginosa, Acinetobacter baumannii, and CRE. The role of carbapenems in treating carbapenem-resistant strains remains widely heterogeneous according to the available resources.
A previous systematic review on the treatment of carbapenem-resistant bacterial sepsis reported a fatality rate of 19% for neonates treated with monotherapy, pointing out the paucity of data from which to draw conclusions [52].
We found most studies coming from LMICs, where colistin is the prevalent antimicrobial used [53,54], but the use of carbapenem in association with colistin or other antibiotics is reported, trying to use synergistic or strategic combinations to overcome resistance. However, on the other hand, in HICs more recent antibiotics are being studied.
The Infectious Diseases Society of America (IDSA) 2021 guidance for MDRO treatment recommended colistin only as an alternative strategy if first-line options are not available or tolerated (mainly combination beta-lactamase inhibitors, carbapenems, and monobactam); however, these recommendations are tailored for adults and high-income settings [55].
Colistin (Polymyxin E) has been available for clinical use since the 1950s. The intravenous formulation was gradually rejected worldwide in the early 1980s because of nephrotoxicity. However, in the last two decades, the emergence of XDR organisms and the lack of newer drugs, especially in LMICs, led to reconsidering colistin as a therapeutic option [56].
Colistin has also been used in newborns and preterm infants, appearing to be effective in treating Gram-negative MDROs [57]. Due to its narrow therapeutic index and scant PK/PD studies in children, dosage recommendations for colistin in children and neonates are challenging to achieve [58,59].
Lastly, as a global concern, Gram-negative bacteria can gain resistance to colistin by acquiring a mobilized colistin resistance (mcr) gene and other subgroups, which are trending toward worrisome dissemination worldwide [60].
Recently approved beta-lactam agents/beta-lactamase inhibitor combinations (βL-βLI ceftazidime-avibactam, ceftolozane-tazobactam, imipenem-relebactam, and meropenem-vaborbactam) offer activity for CRE, DTR-Pseudomonas aeruginosa, and Acinetobacter baumannii, largely depending on the resistance mechanism and the subtype of carbapenemase [61]. Avibactam is a β-lactamase inhibitor that is not susceptible to hydrolysis by ESBLs, AmpCs, KPCs, or OXA 48-like carbapenemases. The FDA initially approved it in 2015 to treat complicated UTIs and intra-abdominal infections (IAI) in combination with metronidazole. It exerts no activity against A. baumannii. Further studies in HICs revealed its non-inferiority compared to meropenem in pediatric patients and informed the recommended doses. Bradley et al. [62] described ceftazidime-avibactam in combination with metronidazole as non-inferior compared to meropenem in treating intra-abdominal infections in infants older than three months of age, reporting a favorable clinical and microbiological response in ≥90% of cases. The same author reported similar safety and efficacy data on the use of ceftazidime-avibactam in complicated UTIs due to MDR Enterobacterales [63]. Ceftolozane-tazobactam has the most potent antipseudomonal activity compared with other βL-βLIs. It was approved in 2014 by the FDA to treat cUTIs and cIAIs in adult patients; Bradley [62,63] showed its efficacy and safety in children in two recent randomized controlled trials for the same indications. Meropenem-vaborbactam consists of an injectable synthetic carbapenem and a boronic acid β-lactamase inhibitor. To date, it has been approved by the FDA to treat adults with CRE-cUTI. [64]. Pediatric data are limited to a single case report in which a KPC-producing K. pneumoniae bloodstream infection was treated successfully [65]. A phase 1 study is underway evaluating dosing, PK, and safety in children [66]. We could not find studies about other molecules such as imipenem-relebactam or cefiderocol.
However, these molecules are not yet approved in neonates, and in the literature, few recent case reports are described [27]. Iosifidis reported one case series about using Ceftazidime/avibactam in neonates with a complete success in XDR or PDR K. pneumoniae infections [29]. In addition, two case reports (not included in this systematic review because they were published subsequently), showed the efficacy and safety of ceftazidime/avibactam in preterms [67,68].
As already highlighted, the main issue comes with affordability in LMICs. In a recent review, Darlow et al. identified five possible affordable antibiotics for neonatal sepsis in LMICs: amikacin, tobramycin, fosfomycin, flomoxef, and Cefepime [18]. However, their use for MDROs is hindered by widespread resistances in LMICs: 63, 55, 25 and 18% of Gram-negative bacteria (including non-Enterobacterales, e.g., Acinetobacter species) for tobramycin, cefepime, amikacin, and fosfomycin, respectively [14].
Our research has many limitations: the included studies are heterogeneous in terms of study design and outcome definitions, making it difficult to compare results, excluding the capacity for a meta-analysis. Further, the quality of evidence is low for most studies due to their retrospective nature (or case reports). The resistance mechanism was not always clearly reported, and in many cases, we deduced it from the susceptibility tests available in the article; the microbiological characterization of resistance was performed only in a few studies. Treatments have been evaluated according to antimicrobial choices in our review, but dosages and durations have not been addressed in our research and are pretty different in the single studies, making them even more challenging to describe. Furthermore, most patients included in the studies were critically ill, with multiple comorbidities, and were admitted to an intensive care unit; these factors may lead to underestimating the specific effect of a particular antibiotic treatment on mortality. In addition, the studies were often based on small sample sizes, reducing the ability to find any effect difference and to consider confounder adjustment and multivariate regression analysis.

5. Conclusions

Newer drugs are promising against emerging MDROs, but data on the neonatal population are lacking, and more extensive studies are needed. However, older molecules still play a crucial role in LMICs for the treatment of neonatal sepsis, where novel combination regimens must be addressed. Strategies cannot be generalized but will be different according to the country’s epidemiology and resources.
As infections run faster than clinical trials, newer ways to reach evidence in pediatrics need to be arranged, such as using global networks that overcome countries’ differences and real-world-data strategies.

Author Contributions

L.C. and D.D. conceived the presented review. L.C., E.B., M.C. and L.R. contributed to the literature search, study selection, and data collection. L.C., M.C. and L.R. discussed study selection and study details and decided which studies were to be included in the review. L.C. and C.L. conducted the assessments of the titles, abstracts, and full texts independently. L.C. performed the numerical calculations and wrote the manuscript with input from all authors. L.C., C.L. and D.D. aided in interpreting the results and revised the manuscript. D.D. and C.G. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The authors received no specific funding for this work.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Report, G. Antimicrobial Resistance: Global Report on Surveillance; World Health Organization: Geneva, Switzerland, 2014. [Google Scholar]
  2. Yusef, D.; Jahmani, T.; Kailani, S.; Al-Rawi, R.; Khasawneh, W. Community-acquired serious bacterial infections in the first 90 days of life: A revisit in the era of multi-drug-resistant organisms. World J. Pediatr. 2019, 15, 580–585. [Google Scholar] [CrossRef] [PubMed]
  3. Folgori, L.; Bernaschi, P.; Piga, S.; Carletti, M.; Cunha, F.P.; Lara, P.H.R.; De Castro Peixoto, N.C.; Alves Guimarães, B.G.; Sharland, M.; Araujo Da Silva, A.R.; et al. Healthcare-Associated Infections in Pediatric and Neonatal Intensive Care Units: Impact of Underlying Risk Factors and Antimicrobial Resistance on 30-Day Case-Fatality in Italy and Brazil. Infect. Control Hosp. Epidemiol. 2016, 37, 1302–1309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Fleischmann-Struzek, C.; Goldfarb, D.M.; Schlattmann, P.; Schlapbach, L.J.; Reinhart, K.; Kissoon, N. The global burden of paediatric and neonatal sepsis: A systematic review. Lancet Respir. Med. 2018, 6, 223–230. [Google Scholar] [CrossRef]
  5. Oza, S.; Lawn, J.E.; Hogan, D.R.; Mathers, C.; Cousens, S.N. Estimations des causes de décès néonatales pour les périodes néonatales précoces et tardives dans 194 pays: 2000–2013. Bull. World Health Organ. 2015, 93, 19–28. [Google Scholar] [CrossRef] [PubMed]
  6. Seale, A.C.; Blencowe, H.; Manu, A.A.; Nair, H.; Bahl, R.; Qazi, S.A.; Zaidi, A.K.; Berkley, J.A.; Cousens, S.N.; Lawn, J.E.; et al. Estimates of possible severe bacterial infection in neonates in sub-Saharan Africa, south Asia, and Latin America for 2012: A systematic review and meta-analysis. Lancet Infect. Dis. 2014, 14, 731–741. [Google Scholar] [CrossRef] [Green Version]
  7. Collins, A.; Weitkamp, J.H.; Wynn, J.L. Why are preterm newborns at increased risk of infection? Arch. Dis. Child. Fetal Neonatal Ed. 2018, 103, F391–F394. [Google Scholar] [CrossRef]
  8. Patel, S.J.; Saiman, L.; Stanley, M. Antibiotic Resistance in NICU Pathogens: Mechanisms, Clinical Impact, and Prevention including Antibiotic Stewardship. Clin. Perinatol. 2015, 37, 547–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Vergnano, S.; Menson, E.; Kennea, N.; Embleton, N.; Russell, A.B.; Watts, T.; Robinson, M.J.; Collinson, A.; Heath, P.T. Neonatal infections in England: The neonIN surveillance network. Arch. Dis. Child. Fetal Neonatal Ed. 2011, 96, 9–14. [Google Scholar] [CrossRef] [Green Version]
  10. Viswanathan, R.; Singh, A.K.; Basu, S.; Chatterjee, S.; Sardar, S.; Isaacs, D. Multi-drug resistant gram negative bacilli causing early neonatal sepsis in India. Arch. Dis. Child. Fetal Neonatal Ed. 2012, 97, F182–F187. [Google Scholar] [CrossRef] [PubMed]
  11. Zaidi, A.K.M.; Huskins, W.C.; Thaver, D.; Bhutta, Z.A.; Abbas, Z.; Goldmann, D.A. Hospital-acquired neonatal infections in developing countries. Lancet 2005, 365, 1175–1188. [Google Scholar] [CrossRef]
  12. Agarwal, R.; Chaurasia, S.; Jeeva Sankar, M.; Yadav, C.P.; Arya, S.; Kapil, A.; Gaind, R.; Vishnubhatla, S.; Chellani, H.; Ramji, S.; et al. Characterisation and antimicrobial resistance of sepsis pathogens in neonates born in tertiary care centres in Delhi, India: A cohort study. Lancet Glob. Health 2016, 4, e752–e760. [Google Scholar] [CrossRef] [Green Version]
  13. Klein, E.Y.; Van Boeckel, T.P.; Martinez, E.M.; Pant, S.; Gandra, S.; Levin, S.A.; Goossens, H.; Laxminarayan, R. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc. Natl. Acad. Sci. USA 2018, 115, E3463–E3470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sands, K.; Carvalho, M.J.; Portal, E.; Thomson, K.; Dyer, C.; Akpulu, C.; Andrews, R.; Ferreira, A.; Gillespie, D.; Hender, T.; et al. Characterization of antimicrobial-resistant Gram-negative bacteria that cause neonatal sepsis in seven low- and middle-income countries. Nat. Microbiol. 2021, 6, 512–523. [Google Scholar] [CrossRef]
  15. WHO Regional Office for Europe and European Centre for Disease Prevention and Control. Surveillance of Antimicrobial Resistance in Europe; ECDC: Solna, Sweden, 2020; Volume 1244, pp. 1–71. [Google Scholar]
  16. Antibiotic Resistance Threats Report. 2019. CDC. Available online: www.cdc.gov/DrugResistance/Biggest-Threats.html (accessed on 20 February 2022).
  17. Agnew, E.; Dolecek, C.; Hasan, R.; Lahra, M.; Merk, H.; Perovic, O.; Sievert, D.; Smith, R.M.; Taylor, A.; Turner, P. Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report; Antimicrobial Resistance Division; Global Antimicrobial Resistance Surveillance System; Global Antimicrobial Resistance Surveillance System (GLASS); SurveillancePrevention & Control; World Health Organization: Geneva, Switzerland, 2021. [Google Scholar]
  18. Darlow, C.A.; da Costa, R.M.A.; Ellis, S.; Franceschi, F.; Sharland, M.; Piddock, L.; Das, S.; Hope, W. Potential Antibiotics for the Treatment of Neonatal Sepsis Caused by Multidrug-Resistant Bacteria. Pediatr. Drugs 2021, 23, 465–484. [Google Scholar] [CrossRef] [PubMed]
  19. Basco, S.A.; Girotto, J.E. Contemporary Treatment of Resistant Gram-Negative Infections in Pediatric Patients Antimicrobial stewardship Resistance Pharmacotherapy. Infect. Dis. Clin. N. Am. 2022, 36, 5520. [Google Scholar] [CrossRef] [PubMed]
  20. Magiorakos, A.; Srinivasan, A.; Carey, R.B.; Carmeli, Y.; Falagas, M.E.; Giske, C.G.; Harbarth, S.; Hindler, J.F. Bacteria: An international expert proposal for interim standard definitions for acquired resistance. Clin. Microbiol. Infect. 2011, 18, 268–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. The National Heart, Lung, and Blood Institute. Guidance for Quality Assessment Tool for Systematic Reviews and Meta-Analyses. Available online: https://www.nhlbi.nih.gov/health-topics/study-quality-assessment-tools (accessed on 20 February 2022).
  22. Velaphi, S.; Wadula, J.; Nakwa, F. Mortality rate in neonates infected with extended-spectrum β lactamase-producing Klebsiella species and selective empirical use of meropenem. Ann. Trop. Paediatr. 2009, 29, 101–110. [Google Scholar] [CrossRef] [PubMed]
  23. Jajoo, M.; Kumar, V.; Jain, M.; Kumari, S.; Manchanda, V. Intravenous colistin administration in neonates. Pediatr. Infect. Dis. J. 2011, 30, 218–221. [Google Scholar] [CrossRef] [PubMed]
  24. Bonfanti, P.; Bellù, R.; Principe, L.; Caramma, I.; Condò, M.; Giani, T.; Rossolini, G.M.; Luzzaro, F. Mother-to-child transmission of KPC carbapenemase-producing Klebsiella pneumoniae at birth. Pediatr. Infect. Dis. J. 2017, 36, 228–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Çağan, E.; Baş, E.K.; Asker, H.S. Use of colistin in a neonatal intensive care unit: A cohort study of 65 patients. Med. Sci. Monit. 2017, 23, 548–554. [Google Scholar] [CrossRef] [Green Version]
  26. Escobar Pérez, J.A.; Olarte Escobar, N.M.; Castro-Cardozo, B.; Valderrama Márquez, I.A.; Garzón Aguilar, M.I.; De La Barrera, L.M.; Rocio Barrero Barreto, E.; Alejandro Marquez-Ortiz, R.; Victoria Moncada Guayazán, M.; Gómez, N.V. Outbreak of NDM-1-producing Klebsiella pneumoniae in a neonatal unit in Colombia. Antimicrob. Agents Chemother. 2013, 57, 1957–1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Coskun, Y.; Atici, S. Successful Treatment of Pandrug-resistant Klebsiella pneumoniae Infection with Ceftazidime-avibactam in a Preterm Infant: A Case Report. Pediatr. Infect. Dis. J. 2020, 39, 854–856. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, X.Y.; Li, X.P.; Wang, M.; Yue, H.J.; Li, P.L.; Liu, Y.P.; Cao, W.; Yao, D.M.; Liu, L.; Zhou, X.L.; et al. Outbreak of NDM-1-producing Klebsiella pneumoniae causing neonatal infection in a teaching hospital in mainland China. Antimicrob. Agents Chemother. 2015, 59, 4349–4351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Iosifidis, E.; Chorafa, E.; Agakidou, E.; Kontou, A.; Violaki, A.; Volakli, E.; Christou, E.I.; Zarras, C.; Drossou-Agakidou, V.; Sdougka, M.; et al. Use of Ceftazidime-avibactam for the Treatment of Extensively drug-resistant or Pan drug-resistant Klebsiella pneumoniae in Neonates and Children Years < 5 of Age. Pediatr. Infect. Dis. J. 2019, 38, 812–815. [Google Scholar] [CrossRef] [PubMed]
  30. Wu, Y.E.; Xu, H.Y.; Shi, H.Y.; van den Anker, J.; Chen, X.Y.; Zhao, W. Carbapenem-Resistant Enterobacteriaceae Bloodstream Infection Treated Successfully With High-Dose Meropenem in a Preterm Neonate. Front. Pharmacol. 2020, 11, 2018–2021. [Google Scholar] [CrossRef] [PubMed]
  31. Celik, I.H.; Oguz, S.S.; Demirel, G.; Erdeve, O.; Dilmen, U. Outcome of ventilator-associated pneumonia due to multidrug-resistant Acinetobacter baumannii and Pseudomonas aeruginosa treated with aerosolized colistin in neonates: A retrospective chart review. Eur. J. Pediatr. 2012, 171, 311–316. [Google Scholar] [CrossRef] [PubMed]
  32. Belet, N.; Haciömeroǧlu, P.; Küçüködük, Ş. Ciprofloxacin treatment in newborns with multi-drug-resistant nosocomial pseudomonas infections. Biol. Neonate 2004, 85, 263–268. [Google Scholar] [CrossRef]
  33. Celebi, S.; Hacimustafaoglu, M.; Koksal, N.; Ozkan, H.; Çetinkaya, M. Colistimethate sodium therapy for multidrug-resistant isolates in pediatric patients. Pediatr. Int. 2010, 52, 410–414. [Google Scholar] [CrossRef] [PubMed]
  34. Kang, C.H.; Tsai, C.M.; Wu, T.H.; Wu, H.Y.; Chung, M.Y.; Chen, C.C.; Huang, Y.C.; Liu, S.F.; Liao, D.L.; Niu, C.K.; et al. Colistin inhalation monotherapy for ventilator-associated pneumonia of Acinetobacter baumannii in prematurity. Pediatr. Pulmonol. 2014, 49, 381–388. [Google Scholar] [CrossRef]
  35. Pratheep, R.; Ray, S.; Mukhopadhyay, K.; Gautam, V.; Shafiq, N.; Dutta, S.; Saini, S.S.; Bhatia, A. First Case Report of Intraventricular Tigecycline in a Neonate with Extensively Drug-resistant Acinetobacter baumannii Ventriculitis. Pediatr. Infect. Dis. J. 2019, 38, E172–E174. [Google Scholar] [CrossRef]
  36. Al-lawama, M.; Aljbour, H.; Tanash, A.; Badran, E. Intravenous Colistin in the treatment of multidrug-resistant Acinetobacter in neonates. Ann. Clin. Microbiol. Antimicrob. 2016, 15, 1–4. [Google Scholar] [CrossRef] [Green Version]
  37. Nakwan, N.; Wannaro, J.; Thongmak, T.; Pornladnum, P.; Saksawad, R.; Nakwan, N.; Chokephaibulkit, K. Safety in treatment of ventilator-associated pneumonia due to extensive drug-resistant Acinetobacter Baumannii with aerosolized colistin in neonates: A preliminary report. Pediatr. Pulmonol. 2011, 46, 60–66. [Google Scholar] [CrossRef]
  38. Thatrimontrichai, A.; Apisarnthanarak, A.; Chanvitan, P.; Janjindamai, W.; Dissaneevate, S.; Maneenil, G. Risk factors and outcomes of carbapenem-resistant acinetobacter baumannii bacteremia in neonatal intensive care unit: A case-case-control study. Pediatr. Infect. Dis. J. 2013, 32, 140–145. [Google Scholar] [CrossRef]
  39. Iosifidis, E.; Antachopoulos, C.; Ioannidou, M.; Mitroudi, M.; Sdougka, M.; Drossou-Agakidou, V.; Tsivitanidou, M.; Roilides, E. Colistin administration to pediatric and neonatal patients. Eur. J. Pediatr. 2010, 169, 867–874. [Google Scholar] [CrossRef]
  40. He, Z.; Wang, C.; Liu, B.; Feng, M.; Wang, Z. Successful treatment of serious meningitis caused by extremely carbapenem-resistant enterobacter cloacae (MIC ≥ 16 mg/L) with i.v. meropenem and i.v. amikacin plus intraventricular amikacin. Infect. Drug Resist. 2019, 12, 3765–3770. [Google Scholar] [CrossRef] [Green Version]
  41. Chen, J.; Yang, Y.; Xiang, K.; Li, L. Combined Rifampin and Sulbactam Therapy for Multidrug- Resistant Acinetobacter Baumannii Ventilator-Associated Pneumonia in Pediatric Patients. J. Anesth. Perioper. Med. 2018, 5, 176–185. [Google Scholar] [CrossRef] [PubMed]
  42. Milic, M.; Siljic, M.; Cirkovic, V.; Jovicevic, M.; Perovic, V.; Markovic, M.; Martic, J.; Stanojevic, M.; Mijac, V. Colonization with multidrug-resistant bacteria in the first week of life among hospitalized preterm neonates in serbia: Risk factors and outcomes. Microorganisms 2021, 9, 2613. [Google Scholar] [CrossRef]
  43. Silago, V.; Kovacs, D.; Samson, H.; Seni, J.; Matthews, L.; Oravcová, K.; Lupindu, A.M.; Hoza, A.S.; Mshana, S.E. Existence of multiple esbl genes among phenotypically confirmed esbl producing klebsiella pneumoniae and escherichia coli concurrently isolated from clinical, colonization and contamination samples from neonatal units at bugando medical center, mwanza, ta. Antibiotics 2021, 10, 476. [Google Scholar] [CrossRef] [PubMed]
  44. Frenk, S.; Rakovitsky, N.; Temkin, E.; Schechner, V.; Cohen, R.; Kloyzner, B.S.; Schwaber, M.J.; Solter, E.; Cohen, S.; Stepansky, S.; et al. Investigation of outbreaks of extended-spectrum beta-lactamase-producing klebsiella pneumoniae in three neonatal intensive care units using whole genome sequencing. Antibiotics 2020, 9, 705. [Google Scholar] [CrossRef]
  45. Kim, Y.K.; Pai, H.; Lee, H.J.; Park, S.E.; Choi, E.H.; Kim, J.; Kim, J.H.; Kim, E.C. Bloodstream infections by extended-spectrum β-lactamase-producing Escherichia coli and Klebsiella pneumoniae in children: Epidemiology and clinical outcome. Antimicrob. Agents Chemother. 2002, 46, 1481–1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Harris, P.N.A.; Tambyah, P.A.; Lye, D.C.; Mo, Y.; Lee, T.H.; Yilmaz, M.; Alenazi, T.H.; Arabi, Y.; Falcone, M.; Bassetti, M.; et al. Effect of piperacillin-tazobactam vs meropenem on 30-day mortality for patients with e coli or Klebsiella pneumoniae bloodstream infection and ceftriaxone resistance. JAMA J. Am. Med. Assoc. 2018, 320, 984–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Tamma, P.D.; Han, J.H.; Rock, C.; Harris, A.D.; Lautenbach, E.; Hsu, A.J.; Avdic, E.; Cosgrove, S.E. Carbapenem therapy is associated with improved survival compared with piperacillin-tazobactam for patients with extended-spectrum β-lactamase bacteremia. Clin. Infect. Dis. 2015, 60, 1319–1325. [Google Scholar] [CrossRef] [Green Version]
  48. Pneumonia, H. Guidelines for the management of adults with hospital-acquired, ventilator-associated, and healthcare-associated pneumonia. Am. J. Respir. Crit. Care Med. 2005, 171, 388–416. [Google Scholar] [CrossRef] [Green Version]
  49. Lee, N.Y.; Lee, C.C.; Huang, W.H.; Tsui, K.C.; Hsueh, P.R.; Ko, W.C. Carbapenem therapy for bacteremia due to extended-spectrum-β-lactamase-producing Escherichia coli or Klebsiella pneumoniae: Implications of ertapenem susceptibility. Antimicrob. Agents Chemother. 2012, 56, 2888–2893. [Google Scholar] [CrossRef] [Green Version]
  50. Malande, O.O.; Nuttall, J.; Pillay, V.; Bamford, C.; Eley, B. A ten-year review of ESBL and non-ESBL Escherichia coli bloodstream infections among children at a tertiary referral hospital in South Africa. PLoS ONE 2019, 14, e0222675. [Google Scholar] [CrossRef] [Green Version]
  51. Lutsar, I.; Chazallon, C.; Trafojer, U.; De Cabre, V.M.; Auriti, C.; Bertaina, C.; Carducci, F.I.C.; Canpolat, F.E.; Esposito, S.; Fournier, I.; et al. Meropenem vs standard of care for treatment of neonatal late onset sepsis (NeoMero1): A randomised controlled trial. PLoS ONE 2020, 15, e0229380. [Google Scholar] [CrossRef] [Green Version]
  52. Donà, D.; Sharland, M.; Heath, P.T.; Folgori, L. Strategic Trials to Define the Best Available Treatment for Neonatal and Pediatric Sepsis Caused by Carbapenem-resistant Organisms. Pediatr. Infect. Dis. J. 2019, 38, 825–827. [Google Scholar] [CrossRef] [PubMed]
  53. Karli, A.; Paksu, M.S.; Karadag, A.; Belet, N.; Paksu, S.; Guney, A.K.; Akgun, M.; Yener, N.; Sensoy, S.G. Colistin use in pediatric intensive care unit for severe nosocomial infections: Experience of an university hospital. Ann. Clin. Microbiol. Antimicrob. 2013, 12, 1. [Google Scholar] [CrossRef] [Green Version]
  54. Sahbudak Bal, Z.; Kamit Can, F.; Yazici, P.; Berna Anil, A.; Duyu, M.; Yilmaz Ciftdogan, D.; Nisel Yilmaz, O.; Cilli, F.; Karapinar, B. The evaluation of safety and efficacy of colistin use in pediatric intensive care unit: Results from two reference hospitals and review of literature. J. Infect. Chemother. 2018, 24, 370–375. [Google Scholar] [CrossRef]
  55. Tamma, P.D.; Aitken, S.L.; Bonomo, A.R.; Mathers, A.J.; van Duin, D.; Clancy, C.J. Infectious Diseases Society of America Guidance on the Treatment of Extended-Spectrum β-lactamase Producing Enterobacterales (ESBL-E), Carbapenem-Resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with Difficult-to-Treat Resistance (DTR-P. aeruginosa). Clin. Infect. Dis. 2021, 72, e169–e183. [Google Scholar] [CrossRef]
  56. Falagas, M.E.; Kasiakou, S.K. Colistin: The revival of polymyxins for the management of multidrug-resistant gram-negative bacterial infections. Clin. Infect. Dis. 2005, 40, 1333–1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ilhan, O.; Bor, M.; Ozdemir, S.A.; Akbay, S.; Ozer, E.A. Efficacy and Safety of Intravenous Colistin in Very Low Birth Weight Preterm Infants. Pediatr. Drugs 2018, 20, 475–481. [Google Scholar] [CrossRef]
  58. Cheah, S.E.; Wang, J.; Nguyen, V.T.T.; Turnidge, J.D.; Li, J.; Nation, R.L. New pharmacokinetic/pharmacodynamic studies of systemically administered colistin against Pseudomonas aeruginosa and Acinetobacter baumannii in mouse thigh and lung infection models: Smaller response in lung infection. J. Antimicrob. Chemother. 2015, 70, 3291–3297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Forrest, A.; Garonzik, S.M.; Thamlikitkul, V.; Giamarellos-Bourboulis, E.J.; Paterson, D.L.; Li, J.; Silveira, F.P.; Nation, R.L. Pharmacokinetic/Toxicodynamic Analysis of Colistin-Associated Acute Kidney Injury in Critically Ill Patients. Antimicrob. Agents Chemother. 2017, 61, e01367-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Gharaibeh, M.H.; Shatnawi, S.Q. An overview of colistin resistance, mobilized colistin resistance genes dissemination, global responses, and the alternatives to colistin: A review. Vet. World 2019, 12, 1735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Tamma, P.D.; Hsu, A.J. Defining the Role of Novel β-Lactam Agents That Target Carbapenem-Resistant Gram-Negative Organisms. J. Pediatr. Infect. Dis. Soc. 2019, 8, 251–260. [Google Scholar] [CrossRef] [PubMed]
  62. Bradley, J.S.; Broadhurst, H.; Cheng, K.; Mendez, M.; Newell, P.; Prchlik, M.; Stone, G.G.; Talley, A.K.; Tawadrous, M.; Wajsbrot, D.; et al. Safety and Efficacy of Ceftazidime-Avibactam Plus Metronidazole in the Treatment of Children ≥3 Months to <18 Years with Complicated Intra-Abdominal Infection: Results from a Phase 2, Randomized, Controlled Trial. Pediatr. Infect. Dis. J. 2019, 38, 816–824. [Google Scholar] [CrossRef]
  63. Bradley, J.S.; Roilides, E.; Broadhurst, H.; Cheng, K.; Huang, L.M.; Mascasullo, V.; Newell, P.; Stone, G.G.; Tawadrous, M.; Wajsbrot, D.; et al. Safety and Efficacy of Ceftazidime-Avibactam in the Treatment of Children ≥ 3 Months to <18 Years with Complicated Urinary Tract Infection: Results from a Phase 2 Randomized, Controlled Trial. Pediatr. Infect. Dis. J. 2019, 38, 920–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chiotos, K.; Hayes, M.; Gerber, J.S.; Tamma, P.D. Treatment of Carbapenem-Resistant Enterobacteriaceae Infections in Children. J. Pediatr. Infect. Dis. Soc. 2019, 9, 56–66. [Google Scholar] [CrossRef] [PubMed]
  65. Hanretty, A.M.; Kaur, I.; Evangelista, A.T.; Moore, W.S.; Enache, A.; Chopra, A.; Cies, J.J. Pharmacokinetics of the Meropenem Component of Meropenem-Vaborbactam in the Treatment of KPC-Producing Klebsiella pneumoniae Bloodstream Infection in a Pediatric Patient. Pharmacotherapy 2018, 38, e87–e91. [Google Scholar] [CrossRef]
  66. Dose-Finding, Pharmacokinetics, Safety, and Tolerability of VABOMERE (Meropenem-Vaborbactam) in Pediatric Subjects with Serious Bacterial Infections (TANGOKIDS). Available online: https://www.clinicaltrials.gov/ct2/show/record/NCT02687906?term=meropenem-vabo (accessed on 20 February 2022).
  67. Nascimento, A.S.; Passaro, M.F.; Rodriguez, F.; Martins, M.K.; Silva, P.S.D.S.; Visacri, B.; Oliveira, S.C.P.; Moriel, P. Off-Label Use of Ceftazidime-Avibactam in a Premature Infant With Multidrug-Resistant Klebsiella pneumoniae Infection: A Case Report. J. Pharm. Prac. 2022, 1–6. [Google Scholar] [CrossRef] [PubMed]
  68. Asfour, S.S.; Alaklobi, F.A.; Abdelrahim, A.; Taha, M.Y. Intravenous Ceftazidime-Avibactam in Extremely Premature Neonates With Carbapenem-Resistant Enterobacteriaceae: Two Case Reports. J. Pediatr. Pharmacol. Ther. 2022, 27, 192–197. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart of the study selection process.
Figure 1. Flowchart of the study selection process.
Antibiotics 11 01088 g001
Figure 2. Results regarding choice of treatment and clinical success for ESBL-producing Enterobacterales non-UTI infections, Carbapenem-resistant (CR) Enterobacterales, Pseudomonas aeruginosa, and XDR A. baumannii. BSI: blood stream infection; UTI: urinary tract infection; VAP: ventilator-associated pneumonia; CNS: central nervous system.
Figure 2. Results regarding choice of treatment and clinical success for ESBL-producing Enterobacterales non-UTI infections, Carbapenem-resistant (CR) Enterobacterales, Pseudomonas aeruginosa, and XDR A. baumannii. BSI: blood stream infection; UTI: urinary tract infection; VAP: ventilator-associated pneumonia; CNS: central nervous system.
Antibiotics 11 01088 g002
Table 1. ESBL Enterobacterales.
Table 1. ESBL Enterobacterales.
ReferenceStudy TypePublication YearCountryCenterSettingN of Patients (Inc/All)Median Age (Year)ResistanceSite of InfectionAntimicrobial TreatmentRouteEvaluated Outcomes Outcome MeasuresResultsQuality
Assessment
S. Velaphi [22]Retrospective2009South AfricaMonocenterNICU100neonateESBLBSI, CNS, LungEmpirical therapy:
Pip–Taz + Amikacina vs. Meropenem vs. other vs. none
IVMortalityAbsolute ValueClinical success: Pip tazo/amikacina: 51%. Meropenem 39%. Mortality: 40% (Pip–Tazo + Amikacina); 43% (Meropenem)fair
NICU: neonatal intensive care unit; ESBL: extended-spectrum β-lactamase; BSI: blood stream infection; CNS: central nervous system; IV: intravenous.
Table 2. Carbapenem-resistant Enterobacterales.
Table 2. Carbapenem-resistant Enterobacterales.
ReferenceStudy TypePublication YearCountryCenterSettingN of
Patients (Inc/All)
Median Age (Year)BacteriaResistanceSite of
Infection
Antimicrobial TreatmentRouteEvaluated Outcomes Outcome MeasuresResultsQuality Assessment
Mamta Jajoo [23]retrospective2011Indiamonocenternicu5preterm and termKCREpneumonia, bsi, cns, chest empyema ColistinivClinical successAbsolute valueClinical success 4/5 (80%)fair
Bonfanti [24]case report2016Italymonocenternicu1PretermKCREBSIColistinivClinical successAbsolute valueClinical success: 1/1poor
eren cağan [25]retrospective2017Turkeymonocenternicu40PretermECREvap, bsi, intrabdominalColistinivClinical successAbsolute valueClinical success: 30/40 (75%)fair
Escobar Perez JA [26]case series2012Colombiamonocenternicu4PretermKCRE NDM 1bsiImipenem + Ciprofloxacin; Meropenem + RifampicinivClinical successAbsolute valueClinical success: Imipenem + Ciprofloxacin 2/3; Meropenem + Rifampicin 1/1fair
yesim coskun [27]Case report2020Turkeymonocenternicu1PretermKPDRUtiCeftazidim/avibactamivClinical successAbsolute valueClinical success: 1/1poor
Zhang XY [28]retrospective2015Chinamonocenternicu8PretermKCRE NDM 1Pneumoniae, bsiMeropenem + ciprofloxacin; ceftazidime, piperacillina/tazobactam + ceftazidime; Meropenem; Meropenem + Piperacillina/tazobactamivClinical successAbsolute valueClinical success: Meropenem + ciprofloxacin: 0/1; ceftazidime: 2/2; piperacillina/tazobactam + ceftazidime: 0/1, Meropenem: 3/3; Meropenem + Piperacillina/tazobactam 1/1fair
elias Iosifidis [29]case series2019Greecemonocenternicu6neonateKXDRbsiceftazidime/avibactamivClinical success; microbiological eradicationAbsolute valueClinical/Microbiological success: 6/6good
Yue-E Wu [30]case report2020Chinamonocenternicu1pretermKCREbsiMeropenem high doseivClinical successAbsolute valueClinical success: 1/1poor
K: Klebsiaella pneumoniae; E: Enterobacterales spp.; iv: intravenous; bsi: blood stream infection; cns: central nervous system; Uti: urinary tract infection; vap: ventilator-associated pneumonia; XDR: extensively drug-resistant; PDR: pan-drug-resistant; CRE: Carbapenem resistance in Enterobacteriaceae (CRE); NDM: New Delhi metallo-beta-lactamase; nicu: neonatal intensive care unit.
Table 3. Pseudomonas aeruginosa.
Table 3. Pseudomonas aeruginosa.
ReferenceStudy TypePublication YearCountryCenterSettingN of
Patients (Inc/All)
Median Age (Year)ResistanceSite of
Infection
Antimicrobial TreatmentRouteEvaluated Outcomes Outcome MeasuresResultsQuality Assessment
Mamta Jajoo [23]retrospective2011Indiamonocenternicu3preterm and termMDRpneumonia, bsi, cns, empyema thoracisColistinivClinical successAbsolute valueClinical success 1/3fair
İstemi Han Celik [31]case series2012Turkeymonocenternicu1preterm and termMDRVAPColistinaerosolizedClinical successAbsolute valueClinical success: 1/1poor
Nursen Belet [32]case series2004Turkeymonocenternicu30pretermMDRVAP, urine, bsi, pleural fluidCiprofloxacinivClinical successAbsolute valueClinical success 23/30 (77%)fair
Solmaz Celebi [33]prospective2010Turkeymonocenterinpatient10pretermXDRVAP, bsiColistinivClinical successAbsolute valueClinical success 8/10 (80%)fair
nicu: neonatal intensive care unit; MDR: multidrug-resistant; bsi: blood stream infection; cns: central nervous system; VAP: ventilator-associated pneumonia; iv: intravenous; XDR: extensively drug-resistant.
Table 4. XDR Acinetobacter baumannii.
Table 4. XDR Acinetobacter baumannii.
ReferenceStudy TypePublication YearCountryCenterSettingN of
Patients (Inc/All)
Median Age (Year)ResistanceSite of
Infection
Antimicrobial TreatmentRouteEvaluated Outcomes Outcome MeasuresResultsQuality
Assessment
Mamta Jajoo [23]retrospective2011Indiamonocenternicu12preterm and termXDRpneumonia, bsi, cns, empyema thoraciscolistinivClinical success, mortalityAbsolute valueClinical success 10/12 (83%)fair
eren cağan [25]retrospective2017Turkeymonocenternicu20pretermXDRVAP, bsi, intra-abdominalcolistinivClinical success, mortality rateAbsolute valueClinical success: 16/20 (80%)fair
İstemi
Han Celik [31]
retrospective2012Turkeymonocenternicu2preterm and term infantXDRVAPcolistinaerosolizedMortalityAbsolute valueMortality: 2/2poor
Solmaz Celebi [33]prospective2010Turkeymonocenterinpatient7pretermXDRpneumoniae, bsicolistinivMortality, clinical successAbsolute valueClinical success: 6/7 (85%)fair
Chia-Hao Kang [34]case series2013Chinamonocenternicu12pretermXDRVAPcolistinivClinical successAbsolute valueClinical success: 12/12poor
Rathna Pratheep [35]case report2019Indiamonocenternicu1pretermXDRcnscolistiniv + ivtClinical successAbsolute valueClinical success: 1/1poor
Manar
Al-lawama [36]
retrospective2016Jordanmonocenternicu21pretermXDRbsicolistinivClinical and microbiological eradicationAbsolute valueClinical success 19/21 (91%)good
narongsak nakwan [37]retrospective2011Thailandmonocenternicu8pretermXDRVAPcolistinaerosolizedClinical successAbsolute valuesClinical success: 7/8 (87%)good
Thatrimontrichai A [38]retrospective2013Thailandmonocenternicu12NeonateXDRBsiceftazidime; Cefperazone/sulbactam; Imipenem, colistin; Imipenem + cefoperazone/sulbactam; Colistin + Cefoperazone sulbactamivClinical successAbsolute valueClinical success: ceftazidime 0/1; cefperazone/sulbactam: 2/3; Imipenem: 2/4; Colistin:1/2; Imipenem + cefperazone/sulbactam: 1/1; Colistin + Cefperazone/sulbactam: 1/1fair
iv: intravenous; ivt: intraventricular; Bsi: blood stream infection; cns: central nervous system; VAP: ventilator-associated pneumonia; nicu: neonatal intensive care unit.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chiusaroli, L.; Liberati, C.; Caseti, M.; Rulli, L.; Barbieri, E.; Giaquinto, C.; Donà, D. Therapeutic Options and Outcomes for the Treatment of Neonates and Preterms with Gram-Negative Multidrug-Resistant Bacteria: A Systematic Review. Antibiotics 2022, 11, 1088. https://doi.org/10.3390/antibiotics11081088

AMA Style

Chiusaroli L, Liberati C, Caseti M, Rulli L, Barbieri E, Giaquinto C, Donà D. Therapeutic Options and Outcomes for the Treatment of Neonates and Preterms with Gram-Negative Multidrug-Resistant Bacteria: A Systematic Review. Antibiotics. 2022; 11(8):1088. https://doi.org/10.3390/antibiotics11081088

Chicago/Turabian Style

Chiusaroli, Lorenzo, Cecilia Liberati, Maria Caseti, Luigi Rulli, Elisa Barbieri, Carlo Giaquinto, and Daniele Donà. 2022. "Therapeutic Options and Outcomes for the Treatment of Neonates and Preterms with Gram-Negative Multidrug-Resistant Bacteria: A Systematic Review" Antibiotics 11, no. 8: 1088. https://doi.org/10.3390/antibiotics11081088

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