Next Article in Journal
Ethnopharmacology, Antimicrobial Potency, and Phytochemistry of African Combretum and Pteleopsis Species (Combretaceae): A Review
Next Article in Special Issue
Editorial for the Special Issue “Antibiotic Prescribing and Antimicrobial Resistance Patterns in Pediatric Patients”
Previous Article in Journal
Diagnosis by Microbial Culture, Breath Tests and Urinary Excretion Tests, and Treatments of Small Intestinal Bacterial Overgrowth
Previous Article in Special Issue
Antibiotics Self Medication among Children: A Systematic Review
 
 
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 Children with Gram-Positive Bacteria with Resistances of Concern: A Systematic Review

1
Pediatric Infectious Diseases, Department for Women’s and Child’s Health, University Hospital of Padova, 35128 Padua, Italy
2
Hospital Pharmacy Department, University Hospital of Padova, 35128 Padua, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Antibiotics 2023, 12(2), 261; https://doi.org/10.3390/antibiotics12020261
Submission received: 7 December 2022 / Revised: 14 January 2023 / Accepted: 26 January 2023 / Published: 28 January 2023

Abstract

:
Background: Methicillin-resistant Staphylococcus aureus (MRSA), methicillin-resistant coagulase-negative Staphylococci (MR-CoNS), and vancomycin-resistant Enterococci (VRE) are increasing worldwide and represent a threat for the limited treatment options in pediatric patients and neonates compared to adults. Recommendations in pediatrics are mainly extrapolated from adults’ studies. Methods: A literature search for the treatment of these pathogens in children (<18 years old) was conducted in Embase, MEDLINE, and Cochrane Library. Studies reporting data on single-patient-level outcomes related to a specific antibiotic treatment for multidrug resistant (MDR) Gram-positive bacterial infection in children were included. Studies reporting data from adults and children were included if single-pediatric-level information could be identified (PROSPERO registration: CRD42022383867). Results: The search identified 11,740 studies (since January 2000), of which 48 fulfilled both the inclusion and the exclusion criteria and were included in the analysis: 29 for MRSA, 20 for VRE, and seven for MR-CoNS. Most studies were retrospective studies. Vancomycin was mainly used as a comparator, while linezolid and daptomycin were the most studied antimicrobials showing good efficacy. Conclusions: Linezolid showed a safety and efficacy profile in a neonatal setting; daptomycin is increasingly used for MRSA, but the evidence is scarce for VRE.

1. Introduction

Gram-positive multidrug-resistant organisms (GP-MDROs) significantly cause hospital-related infections in neonatal and pediatric populations. The increased life expectancy for chronically ill individuals is accompanied by the increased use of invasive devices and access to medical services, raising the risk of colonization and infection with MDR organisms.
Due to their clinical and public health impact, the World Health Organization (WHO) and Centers for Disease Control and Prevention (CDC) have listed both methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE) as high-priority pathogens in urgent need of drug research and development [1,2]. In Europe, in 2019, 18.6% of Staphylococcus aureus (SA) isolates showed single or combined resistances with significant geographical variability; methicillin resistance (MR) can be isolated (2.7%) or combined with quinolones (9.6%) or quinolones and rifampicin (0.6%) [3]. The burden of MRSA, overall, increased from 2007 and was higher in infants younger than 12 months, compared to other age groups [3,4].
The WHO surveillance net in low–middle-income countries (LMICs) reports relatively higher rates of MR for SA compared to high-income countries (HICs): 33.3% in LMICs and 15% in HICs [5].
Reports from HICs (USA) showed that MRSA infections occur mainly as healthcare-associated infections in predisposed individuals, but community-acquired (CA) infections are increasing [6]. In African countries, it is estimated that the majority of MRSA infections are hospital-acquired, but significantly under-recorded cases make the CA-MRSA reservoir underrated [7,8].
In Europe, infections and deaths attributable to VRE doubled between 2007 and 2015 [3,9]. Proportions of bloodstream infections caused by VR Enterococcus faecium (VREFm) increased from 8.1% in 2012 to 19.0% in 2018. The ECDC reported a significant increase in the percentage of VRE isolated in Europe, from 11.6% in 2016 to 16.8% in 2020, in the overall population [3]. However, children and adolescents showed lower VRE proportions than older age groups [10].
Compared to other MDR organisms, a relationship between increased resistance rates for E. faecium and country income status is not observed [11], although the presence of VRE has been widely described in Africa and South America [12,13].
Coagulase-negative staphylococci (CoNS) are typically resistant to methicillin and multiple drugs due to the selective pressure of antibiotic exposure, with an increasing trend [14,15,16]. Although CoNS harbor fewer virulence factors and do not correlate with high morbidity and mortality, they are a significant cause of sepsis in neonatal intensive care units, representing a challenge for the limited antibiotic options in this population [17,18].
To date, the management of GP-MDROs is based on a few indications extrapolated from data on adults, with scant evidence in pediatrics.
This study aims to critically appraise the current antimicrobial treatment options and the relative outcomes for infections caused by the most common Gram-positive bacteria harboring resistances of concern for treatment in the pediatric and neonatal populations: MRSA, VRE, and MR-CoNS.

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 “children”, “Gram-positive bacteria”, “resistant”, “methicillin-resistant Staphylococcus aureus”, “vancomycin-resistant Enterococci”, and “outcome assessment” (see complete search strategy in Supplementary File S1. The search strategy involved restrictions on the date (from 1 January 2000 to 1 November 2022) but not on language. All studies on children younger than 18 were considered.
This study was registered with the International Prospective Register of Systematic Reviews (PROSPERO) under record number CRD42022383867.
We included studies with any method of diagnosing infections with pathogens of interest in children, neonates and preterms. All sites of infection were 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., C.L., and L.R.). Discussion with a fourth senior 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 the following bacteria were included: MRSA, VRE, and MR-CoNS.
The populations of interest were children, as well as term and preterm newborns, with confirmed GP-MDROs infections receiving antimicrobial treatment and presenting clinical and/or microbiological outcomes.
The outcomes of interest we collected from the selected studies were infection-related mortality from the initiation of treatment until discharge, clinical success (defined as complete resolution or a substantial improvement in 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 30 October 2022 were included. Further details are reported in the PICOS format (P: problem/patient/population; I: intervention; C: comparison/control; O: outcome; S: study design).
P (Participants/population): Children, as well as term and preterm newborns, with confirmed GP-MDROs infections that were receiving antimicrobial treatment and presenting clinical and/or microbiological outcomes clearly specified.
I (Intervention/Exposure): Any antimicrobial treatment clearly defined.
C: (Comparator/Control): Standard of care at the time and place where the study was conducted. Not applicable.
O: (outcomes): The primary outcome was mortality-related GP-MDRO infection. Secondary outcomes were clinical success, defined as complete resolution or substantial improvement of signs and symptoms of the index infection, microbiological success measured by suppression, eradication, or relapses of bacterial growth, and treatment-related adverse effects.

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 [19]. The quality assessment results are presented in Table 1, Table 2 and Table 3. Each study was quality-rated according to one of the following categories on the basis of the proportion of yes answers to all relevant questions: poor quality, 0–40%; fair quality, 41–80%; good quality, 81–100% [19].

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, followed by 138 exclusions after reading the full text as they did not meet the eligibility criteria. Forty-two articles regarding Gram-negative bacteria were also excluded. Therefore, 48 articles were included in the systematic qualitative review (Figure 1). We report the characteristics of the studies in Table 1, Table 2 and Table 3. Figure 2 shows the studies with a relevant number of patients.

3.2. Methicillin-Resistant Staphylococcus aureus (MRSA)

We included 29 studies regarding MRSA: 11 randomized controlled trials, two prospective studies, six retrospective studies, two case series, and nine case reports [20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48].
Clinical and microbiological success is reported in Table 1.
Most relevant studies were conducted in high-income countries (19/30), and three were multicenter international studies. The selected studies reported the following MRSA clinical infections: skin and skin structure infections (SSSIs), bloodstream infections (BSIs), central line-associated bloodstream infections (CLABSIs), endocarditis, community and hospital pneumonia, bone infections, and others.
The included antibiotic regimens were linezolid (13/30), daptomycin (10/30), ceftaroline (3/30), and other drugs 4/30 (TMP-SMX, cephalexin, and quinupristin/dalfopristin). Vancomycin was considered the comparator in 4/30 studies.

3.3. Vancomycin-Resistant Enterococcus (VRE)

We included 20 studies regarding VRE: one randomized controlled trial, two prospective studies, three retrospective studies, and nine case reports [22,23,25,27,36,46,49,50,51,52,53,54,55,56,57,58,59,60,61,62].
Clinical and microbiological success and antimicrobial safety are reported in Table 2.
The settings were different, but most studies (11/20) were conducted in HICs. The selected studies reported the following infections: BSIs, central nervous system (CNS), pneumonia, and other sites.
The included antibiotic regimens were linezolid (11/20), daptomycin (6/20), and other drugs 5/20 (tigecycline, quinupristin/dalfopristin). Linezolid as monotherapy was the more described antibiotic. A case report described the use of phage therapy for VRE [54].

3.4. Methicillin-Resistant Coagulase-Negative Staphylococci (MR-CoNS)

We included seven studies regarding MR-CoNS: one randomized controlled trial, one retrospective study, one case series, and three case reports [25,27,46,63,64,65,66].
Clinical and microbiological success and antimicrobial safety are reported in Table 3.
Only two out of six studies were conducted in LMICs. The selected studies reported the following infections: BSIs, CNS, pneumonia, and other sites.
Described antimicrobials were linezolid, vancomycin, and daptomycin.
Table 1. Treatment choices and outcomes for methicillin-resistant Staphylococcus aureus (MRSA).
Table 1. Treatment choices and outcomes for methicillin-resistant Staphylococcus aureus (MRSA).
ReferenceStudy TypePublication YearCountryCenterSettingN of Patients (Inc/All)Median Age (Year)ResistanceSite of InfectionAntimicrobial TreatmentRouteOutcomes MeasuresOutcomes MeasuresResultsQuality Assessment
Korczowski, Bartosz [20]Randomized, observer-blinded, active-controlled2016WorldwideMulticenterInpatient25Not indicated #MRSASSSIsCeftarolineivClinical success, microbiological eradicationAbsolute valueClinical success: ceftaroline 17/18 (94%) vs. vancomycin 6/7 (86%) Microbiological response: 16/18 (89%) ceftaroline vs. 4/7 (57%) vancomycinGood
John S. Bradley [21]Randomized, controlled, double-blind2020WorldwideMulticenterInpatient, outpatient89MRSABoneDaptomycinivClinical success, microbiological eradicationAbsolute valueClinical success: daptomycin 2/4 (50.0%) vs. vancomycin1/4 (25.0%) Microbiological eradication: daptomycin 2/4 (50.0%) vs. vancomycin 3/4 (75.05)Good
Monica I. Ardura [22]Retrospective2007USAMonocenterInpatient, outpatient146MRSABSIs, endocarditis, pneumonia, pyomyositis, and osteoarthritisDaptomycinivClinical successAbsolute valueClinical success: daptomycin 13/14 (92%)Fair
Maria Moschovi [23]Prospective2010GreeceMonocenterOncoematological44MRSACLABSIsLinezolidivClinical successAbsolute valueClinical success: 4/4 (100%)Fair
Sheldon L. Kaplan [25]Randomized trial, comparator-controlled2003USA e MexicoMulticenterInpatient, outpatient296MRSAHAP, SSSIs, BSIsLinezolid vs. vancomycinivClinical successAbsolute valueClinical success
Linezolid 16/17 (94%) vs. vancomycin 9/12 (90%)
Good
Glenn Isaacson [26]Retrospective2008USAMonocenterOutpatient72MRSAEarLinezolidOralClinical successAbsolute valueClinical success: 100% (7/7)Poor
Adem YılmAz [27]Retrospective2010TurkeyMonocenterInpatient111MRSACNSLinezolidivClinical successAbsolute valueClinical success: 1/1Fair
Tae-Jung Sung [28]Case report2008KoreaMonocenterInpatient1PrematureMRSAEndocarditisLinezolidivClinical successAbsolute valueClinical response: 0/1Poor
Joshua I. Chan [29]Case report2020USAMonocenterInpatient1PrematureMRSABSIsDaptomycinivClinical successAbsolute valueClinical success: 1/1Poor
Zakaria Jalal [30]Case report2013TurkeyMonocenterInpatient112MRSAEndocarditis, BSIsDaptomycinivClinical successAbsolute valueClinical success: 1/1Poor
Aaron e. Chen [31]Randomized trial2010USAMonocenterOutpatient133Not indicated #MRSASSSIsCephalexin Vs ClindamycinOralClinical successAbsolute valueClinical success:
cephalexin 63/63 (100%) vs. clindamycin 66/70 (94%)
Good
Stefan Borgmann [32]Case report2016GermanyMonocenterInpatient19MRSABSIsCeftarolineivClinical successAbsolute valueClinical success: BSIs resolvedPoor
Al Zabem [33]Case series2016JordanMonocenterInpatient, outpatient55.8MRSABoneVancomycin + rifampicin, then Tmp/Smx + rifampiciniv/oralClinical successAbsolute valueClinical success: 4/5 (80%)Poor
Lucy Holmes [34]Randomized trial2015USAMonocenterInpatient137Not indicated #MRSASSSIsTMP-SMXOralClinical successAbsolute valueClinical success:
61/68 (89%) 3 days of therapy vs. 68/69 (98%) 10 days of therapy
Good
Satoshi Iwata [39]Open-label, single-arm phase 2 study2021JapanMulticenterInpatient, outpatient87MRSAcSSIs, BSIsDaptomycinivClinical successAbsolute valueClinical success: cSSIs 5/7 (71%), BSI 1/1 (100%)Good
Nicholas M. Fusco [37]Retrospective2019USAMonocenterCystic fibrosis122 *18MRSAPneumoniaLinezolid vs. vancomycinivClinical success, adverse effectsAbsolute valueClinical success: vancomycin 53/66 (80.3%) vs. linezolid 50/66 (76%)
Adverse effect: vancomycin 10/66 (15.2%) vs. linezolid 2/66 (3%)
Good
John Bradley [38]Randomized trial2017WorldwideMulticenterInpatient, outpatient97Not indicated #MRSASSSIsDaptomycin vs. standard of careivClinical success, adverse effectsAbsolute valueClinical success: daptomycin 82/97 (85%) and SOC 41/46 (89%)
Adverse effect: 14% of daptomycin vs. 17% of SOC
Good
Aaron Cook [40]Case report2005USAMonocenterInpatient14MRSACNSLinezolid + rifampicinivClinical and microbiological successAbsolute valueClinical success and microbiological success: 1/1Poor
masayoshi shinjoh [35]Retrospective2012JapanMonocenterInpatient164MRSABSIs, skin, lung, CNSLinezolidiv/oralClinical successAbsolute valueClinical success: 10/16 (63%)Fair
Loeffler A. [36] Prospective2002USAMulticenterNot specified87MRSABSIs, skin, pneumonia, joints, bone, CLABSIQuinupristin/dalfopristinivClinical successAbsolute valueClinical success: 5/8 (62%)Fair
Antonio c. Arrieta [48]Randomized multicenter2018WorldwideMulticenterNot specified108MRSABone, joints, BSIs, CLABSIs, intrabdominalDaptomycin vs. SOCiv/oralClinical successAbsolute valueClinical success: daptomycin 6/7 (85%) vs. 2/3 (67%) SOCGood
Kenneth Wible [24]Randomized controlled trial2003USAMulticenterNot specified2010MRSASSSIsLinezolidoralClinical successAbsolute valueClinical success: linezolid 12/13 (92%) vs. cefadroxil 6/7 (85%)Good
Ram Yogev [41]Open label, randomized2003USAMulticenterInpatient183MRSASSSIsLinezolid vs. vancomyciniv/oralClinical successAbsolute valueClinical success: linezolid 9/10 (90%) vs. vancomycin 6/8 (75%)Good
Gallagher [42]Case report2008UKMonocenterInpatient14MRSA-VISACNSRifampicin, linezolidiv/oralClinical and microbiological successAbsolute valueClinical success: 1/1Poor
Chih-Jung Chen [43]Retrospective2007TaiwanMonocenterNot specified116MRSABoneLinezolid iv/osiv/oralClinical and microbiological successAbsolute valueClinical success: 9/11 (81%)Fair
Lara Jacobson [44]Case report2009USAMonocenterInpatient115MRSABSIsDaptomycinivClinical successAbsolute valueClinical success: 0/1Poor
Salerno [45]Case report2017USAMonocenterInpatient1PrematureMRSAPneumoniaCeftaroline + rifampicinivClinical and microbiological successAbsolute valueClinical and microbiological success: 1/1Poor
Hussain [47]Case report2011United KingdomMonocenterNICU1PrematureMRSABSIsDaptomycinivClinical successAbsolute valueClinical success: 1/1Poor
Palma [46]Case series2013ItalyMonocenterPICU3Not indicatedMRSABSIs, SSSIsDaptomycinivClinical successAbsolute valueClinical success: 3/3Poor
USA: United States of America; BSI: bloodstream infection; SSSI: skin and skin structure infection; cSSI: complicated skin and skin structure infection; SOC: standard of care; CNS: central nervous system, iv: intravenous, VISA: vancomycin intermediate-resistant Staphylococcus aureus, CLABSI: central line-associated bloodstream infection, HAP: hospital-acquired pneumonia, TMP-SMX: trimethoprim–sulfamethoxazole, NICU: neonatal intensive care unit. * Study reported 122 episodes of acute pulmonary exacerbations in 49 patients with cystic fibrosis. We considered the total of episodes because etiology, treatment, and outcome were defined in each one. # Study regarding pediatric population. The mean age was not clearly defined.
Table 2. Choice of treatment and outcomes for vancomycin-resistant Enterococcus (VRE).
Table 2. Choice of treatment and outcomes for vancomycin-resistant Enterococcus (VRE).
ReferenceStudy TypePublication YearCountryCenterSettingN of Patients (Inc/All)Median Age (Year)ResistanceBacteriaSite of InfectionAntimicrobial TreatmentRouteOutcomes MeasuresOutcoMes MeasuresResultsQuality Assessment
Monica I. Ardura [22]Retrospective2007USAMonocenterInpatient110VREE. faeciumUTIsDaptomycinivClinical successAbsolute valueClinical success: daptomycin 0/1Fair
Maria Moschovi [23]Prospective2010GreeceMonocenterOncoematological102.8VREEnterococcus spp.BSIs, stoolLinezolidivClinical successAbsolute valueClinical success: 10/10 (100%)Fair
Ayse Şahin [49]Case report2019TurkeyMonocenterInpatient12 monthsVREE. faeciumCNSLinezolid iv + daptomycin iviv + ivtClinical successAbsolute valueClinical success: 1/1
Poor
Ayse Sahina [50]Case report2019TurkeyMonocenterInpatient15 monthsVREE. faeciumCNSTigecyclineiv + ivtMicrobiological eradicationAbsolute valueMicrobiological eradication: 1/1Poor
Heather B. Jaspan [51]Case report2010USAMonocenterInpatient121 monthsVREEnterococcus faeciumCNSLinezolid + daptomycin + tigecycline + daptomycin IVTiv + ivtClinical successAbsolute valueClinical success: 1/1Poor
Rene Hoehn [52]Case report2006GermanyMonocenterNICU2PretermVREEnterococcus spp.BSIsLinezolidivClinical successAbsolute valueClinical and success 2/2Poor
Adem YılmAz [27]Retrospective2010TurkeyMonocenterInpatient111VREE. faeciumCNSLinezolidivClinical successAbsolute valueClinical success: 1/1Fair
Sheldon L. Kaplan [25]Trial randomized2003USA and central AmericaMulticenterInpatient33VREE. faeciumBSIsLinezolidivClinical successAbsolute valueClinical success: 2/3 (66%)Good
Marco Fossati [53]Case report2010ItalyInpatientOncoematological111VREE. faeciumBSIsDaptomycinivClinical successAbsolute valueClinical success 0/1Poor
Loeffler A. [36]Prospective2002USAMulticenterNot specified1017VREEnterococcus spp.BSIs, skin, pneumonia, joint, bone, CLABSIQuinupristin/dalfopristinivClinical successAbsolute valueClinical success: 71/101 (70%)Fair
Kevin Paul [54]Case report2021GermanyMonocenterInpatient110 monthsVRE AbdominalBacteriophage therapyivClinical successAbsolute valueClinical success: 1/1Poor
James W. Gray [55]Case Series2000UKMonocenterInpatient and outpatient87VREEnterococcus spp.BSIs, abdominalQuinupristin/dalfopristinivClinical successAbsolute valueClinical success: 7/8 (87%)Fair
Jocelyn Ang [56]Case report2003USAMonocenterNICU1PrematureVREE. faeciumEndocarditisLinezolidivClinical and microbiological successAbsolute valueClinical and microbiological success: 1/1Poor
Mehmet Baysallar [57]Case report2006TurkeyMonocenterInpatient17 monthsVREE. faeciumCNSChloramphenicol, rifampin, and meropenemivClinical and microbiological successAbsolute valueClinical and microbiological success: 1/1Poor
M. Travaglianti [58]Retrospective2007ArgentinaMonocenterInpatient157 yearsVREEnterococcus spp.BSIs, UTIs, abdominal, endocarditisLinezolidiv/oralClinical and microbiological successAbsolute valueClinical and microbiological success: 13/15 (87%)Poor
Graham [59]Case report2002USAMonocenterNICU1PretermVREE. faeciumCNSLinezolidivClinical successAbsolute valueClinical success: 1/1Poor
Beneri [60]Case report2008USAMonocenterInpatient1NeonateVREE. faeciumBSIsDaptomycin + doxyxyxlineIvClinical successAbsolute valueClinical success: 1/1Poor
Maranich [61]Case report2008USAMonocenterInpatient117 monthsVREE. faeciumCNSLinezolidIvClinical successAbsolute valueClinical success: 1/1Poor
Palma [46]Case series2013ItalyMonocenterPICU1Not indicatedVREE. faeciumBSIs, SSSIsDaptomycinivClinical successAbsolute valueClinical success: 1/1Poor
Ergaz [62]Case report2009IsraelMonocenterNICU3PretermVREE. faeciumBSIs, CNSLinezolidIvClinical successAbsolute valueClinical success: 3/3Poor
CNS: central nervous system. IV: intravenous; IVT: intraventricular USA: United States of America. VRE: Vancomycin-resistant enterococci. UTI: urinary tract infection, CLABSI: central line-associated bloodstream infection, BSI: bloodstream infection, VAP: ventilator-associated pneumonia, NICU: neonatal intensive care unit, SSSI: skin and skin structure infection, UTI: urinary tract infection.
Table 3. Choice of treatment and outcomes for methicillin-resistant coagulase-negative staphylococci (MR-CoNs).
Table 3. Choice of treatment and outcomes for methicillin-resistant coagulase-negative staphylococci (MR-CoNs).
ReferenceStudy TypePublication YearCountryCenterSettingN of Patients (Inc/All)Median Age (Year)ResistanceBacteriaSite of InfectionAntimicrobial TreatmentRouteOutcomes MeasuresOutcomes MeasuresResultsQuality Assessment
Shanti [63]Case report2009MalesiaMonocenterInpatient11MRS. epidermisCNSiv teicoplanin + IVT teicoplanin 10 mg dailyiv+ivtMicrobiological eradicationAbsolute valueMicrobiological eradication: 1/1Poor
Sheldon L. Kaplan [25]Trial randomized2003USA e central AmericaMulticenterInpatient462MRS. epidermisBSILinezolid vs. vancomycinivClinical successAbsolute valueClinical success: linezolid 29/34 (85.3) vs. vancomycin 10/12 (83.3)Good
Adem YılmAz [27]Retrospective2010TurkeyMonocenterInpatient411MRS. epidermidisCNSLinezolidivClinical successAbsolute valueClinical success: 4/4Fair
C. Minotti [64]Case report2022ItalyMonocenterNICU1PretermMRS. epidermidisCLABSIDaptomycinivClinical successClinical successClinical success: 1/1Poor
Fumihiro ochi [65]Case series2018JapanMonocenterInpatient22MRS. epidermidisCNSLinezolidivClinical successAbsolute valueClinical success: 2/2Fair
Palma [46]Case series2013ItalyMonocenterPICU3Not indicatedMRS. epidermidisBSI, SSSIsDaptomycinivClinical successAbsolute valueClinical success: 3/3Poor
Gawronski [66]Case report2015OhioMonocenterNICU1PretermMRS. epidermidisBSIDaptomycinivClinical successAbsolute valueClinical success: 1/1Poor
CLABSI: central line-associated bloodstream infection; SOC: standard of care, MR: methicillin-resistant, CNS: central nervous system, iv: intravenous, IVT: intraventricular, CoNS: coagulase-negative staphylococci, CLABSI: central line-associated bloodstream infection, NICU: neonatal intensive care unit.

4. Discussion

Gram-positive bacteria may harbor several types of resistance to one or more antimicrobial class agents, with specific criteria to be fulfilled for being defined as MDR [67]. This research focused on the treatment of the most common drug-resistant Gram-positive pathogen infections.

4.1. MRSA, VRE

We found most studies (21/27) on MRSA, predominantly analyzing its treatment in invasive infections (SSSIs, bloodstream, bone, pneumonia, and CLABSIs).
Only one retrospective study on uncomplicated SSSIs caused by MRSA was included, in which cephalexin was as effective as clindamycin. However, as cephalexin does not exert any activity against MRSA, the authors concluded that wound care and drainage are more important than the antimicrobial choice [31].
For invasive infections, vancomycin, which is usually considered and recommended as first-line treatment for MRSA infections in pediatrics [68,69], was mainly used in our research as a comparator to assess the efficacy of other molecules: daptomycin, ceftaroline, and linezolid. We found no studies (except a case report) on teicoplanin, as it is not approved by the FDA and is used mainly in European countries.
Ceftaroline is currently approved by FDA and EMA for children and newborns to treat cSSSIs and community-acquired pneumonia [70,71]; however, efficacy data for MR organisms in pediatrics remain scarce. In our research, ceftaroline was evaluated for community-acquired MRSA (CA-MRSA) in children with SSSIs compared with vancomycin in 25 patients, resulting in a similar efficacy rate [20]. However, children with complicated infections or bacteremia were excluded from the study. In a case report, ceftaroline was successfully used (in combination therapy with rifampicin) to treat MRSA bloodstream infection and pneumonia in a preterm infant [45]. Ceftaroline showed a favorable efficacy and safety profile in newborns with late-onset sepsis in a case series, but MRSA was not considered, and antibiograms of CoNs were not provided [72]. Ceftaroline showed efficacy similar to vancomycin in children with cystic fibrosis and pulmonary exacerbations, which are often colonized (and infected) with MRSA [73]. Extensive studies and trials evaluating ceftaroline for treating MRSA/MRSE bacteremia and other invasive infections are lacking. In addition, children and newborns with sepsis often suffer from renal failure and are exposed to other nephrotoxic drugs; therefore, a vancomycin-sparing regimen may ease the management avoiding therapeutic drug monitoring and protecting the renal function. In this view, ceftaroline is a promising agent due to its safety and broad activity spectrum [74]. Further studies may explore its applications in special populations such as oncohematological patients and neonates, as well as for device-related infections (endocarditis and CLABSIs).
Daptomycin is a novel lipopeptide approved by FDA and EMA for the treatment of cSSSIs and bacteremia caused by Gram-positive bacteria in children older than 1 year, due to its rapid bactericidal effect [75,76]. It is active against MRSA, VRE, and MR-CoNS. Bradley et al. evaluated daptomycin for complicated SSSI in a randomized trial, including 97 MRSA infections treated with daptomycin and 46 with the standard of care, with a similar safety profile. However, children with bacteremia (or other invasive infections), renal insufficiency, or any clinical or laboratory findings suggestive of potential daptomycin toxicity were excluded from this study. The study was not designed to statistically compare the efficacy of daptomycin with the standard of care; however, overall, the efficacy rate was similar between arms [48]. The same authors described results from a randomized trial including eight children with hematogenous MRSA osteomyelitis treated with daptomycin or vancomycin; however, those cases were insufficient to achieve conclusions [21]. Seven children (neonates and infants excluded) with MRSA cSSSIs and one with bacteremia were successfully treated with daptomycin in a prospective Japanese study [39]. On the other hand, we found no trials or prospective studies evaluating the safety and efficacy of daptomycin for VRE infections in children, with publications limited to case series or case reports [22,46,53,60].
Linezolid is the first agent of oxazolidinones and is approved by FDA for adults and children with SSSI or pneumonia, including MRSA or VRE etiologies [77]. In pediatrics, linezolid was studied in a randomized trial, showing clinical efficacy comparable to vancomycin in 20 MRSA severe infections, and showing microbiological eradication for three cases of VRE [78]. In a prospective series of immunocompromised children, linezolid was effective as a single therapy for MRSA and VRE infections without a concomitant worsening of chemotherapy-induced myelotoxicity [23]. Furthermore, linezolid has been described in many pediatric case reports to treat ventriculitis, bloodstream infections, endocarditis, and others [27,49,51,52,56,59,61].
Despite its bacteriostatic activity, linezolid has a favorable efficacy for bacteremia in neonates, comparable to vancomycin [79,80]. Linezolid has the advantages of tolerability for a prolonged course, possible oral shift, and less concern for resistance, particularly for MRSA, compared to vancomycin.
Two recent meta-analyses showed an increase in the last years of vancomycin-intermediate, resistant, and heterogenous intermediate SA (VISA, VRSA, and hVISA), particularly in the USA and Asia [81,82]. The prevalence of VRSA increased from 1.2% before 2010 to 2.4% after 2010, with the highest frequency in the USA (3.6%) [81]. We found only one case of SA in our research with a MIC of 4 mg/dL for vancomycin [42]. VRE resistance to linezolid is rare but possible. Two case reports described a combination of antibiotics to treat severe VRE infections: linezolid with daptomycin [49], and daptomycin with tigecycline [51]. To date, no recommendations of combination therapy for VRE or other Gram-positive MDR infections have been provided. Concerns exist because the in vitro inhibition of the growth induced by a bacteriostatic drug may reduce efficacy when combined with a bactericidal drug [83]. Clinical data in vivo are missing.
Tedizolid is a newer oxazolidinone approved by EMA for treating SSSIs in children older than 12 years [84]. In the pediatric population, the safety and efficacy of tedizolid were evaluated in a phase III trial in adolescents with SSSIs caused by multiple pathogens, comparing tedizolid with the standard of care. The study included two MRSA in the tedizolid group and one in the SOC group, but specific outcomes were not reported, and the study was then excluded from our collection [85].
The use of quinupristin/dalfopristin has been described in a retrospective series of children with concomitant comorbidities and serious Gram-positive MDR infection without other drug possibilities [36]. It resulted in clinical success for 71 out of 101 VRE infections (in combination with other antimicrobials). As limitations, quinupristin/dalfopristin is ineffective against E. faecalis, requires a central line because it is highly irritant for peripheral veins, and has several adverse effects: metabolic interactions, severe myalgias, arthralgias, nausea, and hyperbilirubinemia [86]. In adults, studies comparing quinupristin to linezolid showed similar efficacy with a better versatility for linezolid [86,87]. In the pediatric population, the paucity of data does not allow us to conclude, but safety concerns led to its progressive desertion in favor of linezolid and daptomycin.
Novel antibiotics are currently approved by the FDA and/or EMA to treat Gram-positive MDROs in adults [88]. Oritavancin is a new-generation lipoglycopeptide, currently approved by the FDA and EMA for acute bacterial soft skin and skin structures infections due to MSSA, MRSA, Streptococcus spp., and E. faecalis [89]. Despite its potential activity, oritavancin has not been clinically evaluated against VRE. A phase I clinical trial is recruiting pediatric patients with Gram-positive bacterial infections to evaluate the safety and tolerability of oritavancin [90].
Dalbavancin is a semisynthetic lipoglycopeptide with a mechanism of action similar to vancomycin, active against MRSA but not against VRE. It is approved by the EMA and FDA to treat adults with cSSSI [91]. Its long half-life (8.5 days) allows a weekly administration, which would be a massive advantage in pediatrics, as prolonged admissions and vascular catheter management are uncomfortable and expensive. A phase III trial (dalbavancin versus standard of care) recruiting patients aged 3 months to 17 years with SSSI is currently ongoing [92].

4.2. MR-CoNS

Compared to other Gram-positive bacteria, there are fewer studies regarding the treatment of MR-CoNS in pediatrics. However, MR-CoNS are widely diffused and poses a challenge, particularly for the limited possibilities in the neonatal population. In our research, neonates were under-represented compared to children. We included only one randomized trial where linezolid was successfully used in 13 neonatal MRSE infections with an 85% eradication rate, compared with vancomycin (100% eradication on six patients). However, no specific information on clinical outcomes was provided in this study [25]. There is a paucity of data regarding daptomycin’s safety and efficacy in neonates, which seems to need higher doses to match the clinically effective exposure [93]. A retrospective case series by Mohzari et al. described 15 preterm infants with MRSE infections treated with daptomycin after vancomycin failure, with a success rate of 11 out of 15 [94].

4.3. Neonatal Safety

Safety concerns and the lack of pharmacokinetics data in neonates and preterm neonates explain why antimicrobial research often leaves this population behind in drug approvals. This study was not designed to assess antimicrobial safety in pediatrics; however, due to the paucity of data, we looked at safety outcomes in neonates included in the research. Deville performed a sub-analysis on neonatal patients included in the randomized trial by Kaplan et al., comparing vancomycin and linezolid [79]; the two groups of neonates were homogeneous except for postnatal age (newborns were younger in the linezolid group), and adverse effects were overall similar. Of note, the percentage of patients with abnormal hematology or serum chemistry values was not statistically different in linezolid-treated patients and vancomycin-treated patients. In the case series by Ergaz, three preterm newborns infected with VRE were successfully treated with linezolid without abnormalities in white blood cell count and liver enzymes [62]; the same was observed for the two preterms treated with linezolid presented by Hoehn [52]. A review by Garazzino on the use of linezolid in pediatric and neonates described an excellent safety profile [95]. Daptomycin use was described in the series by Mohzari (median weight 870 g) without reporting muscular or neurologic toxicity in preterms treated for MR-CoNS invasive infections [94]. In the neonatal case reports included in our study, daptomycin was used with good tolerability [64,66].
In the case series by Bradley, 11 neonates treated with ceftaroline for LOS experienced mild adverse effects, except for one serious effect (salmonellosis), but only a case of diarrhea was related to the study treatment [72].

4.4. Limitations

This review had some limitations, which were intrinsic to the object of our research. The main limiting factor was the paucity of articles exploring the treatment of Gram-positive in children and neonates; in particular, data about MR-CoNS, albeit widely diffused, are very few. Furthermore, the included studies had different study designs, making it difficult to compare results and excluding the capacity for a meta-analysis. We extrapolated information on MDR pathogens from larger studies that were not designed ad hoc to study them but often described the experience with a single antibiotic. In addition, the quality of evidence was low for most studies due to their retrospective nature (or case reports); the definition of the outcomes (clinical and microbiological success) was not detailed in every study, and it could vary, impairing the strength of the results.

5. Conclusions

In conclusion, robust evidence on the treatment of MRSA, VRE, and MR CoNS is lacking in the pediatric and neonatal populations. However, a trend toward newer and safer molecules is observed (ceftaroline and linezolid) compared to the standard of care (vancomycin) for MRSA. Further studies are needed to investigate their effectiveness in particular settings (immunocompromised children, device-related infections, and critical care settings). Linezolid seems promising in the neonatal population for its activity against MRSA and MR CoNS, safety profile, and optimal bioavailability. Daptomycin and linezolid remain the cornerstones of VRE treatment. Novel molecules (oritavancin and dalbavancin) are currently being studied in pediatrics, with promising applications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antibiotics12020261/s1, File S1: Search strategy; File S2: PICOS format.

Author Contributions

L.C. and D.D. conceptualized the presented review; L.C., E.B., C.L. and L.R. contributed to the literature search, study selection, and data collection; L.C., C.L. and L.R. discussed the 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. and C.L. performed the numerical calculations and wrote the manuscript with input from all authors; L.C., C.L., M.D.P., D.M., C.D.C. and D.D. aided in interpreting the results and revised the manuscript; D.D., E.B., M.D.P., D.M., C.D.C. and C.G. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in article and in Supplementary Material.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tacconelli, E.; Carrara, E.; Savoldi, A.; Harbarth, S.; Mendelson, M.; Monnet, D.L.; Pulcini, C.; Kahlmeter, G.; Kluytmans, J.; Carmeli, Y.; et al. Discovery, research, and development of new antibiotics: The WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect. Dis. 2018, 18, 318–327. [Google Scholar] [CrossRef] [PubMed]
  2. Center for Disease Control and Prevention (CDC). Antibiotic Resistance Threats in the United States; Department of Health and Human Services: Washington, DC, USA, 2019; pp. 1–140. [CrossRef] [Green Version]
  3. ECDC. Antimicrobial resistance in the EU/EEA (EARS-Net) Annual Epidemiological Report for 2019. Epidemiolog. Antimicrob. Resist EU/EEA 2020, 174, 341. [Google Scholar]
  4. McMullan, B.J.; Bowen, A.; Blyth, C.C.; Van Hal, S.; Korman, T.M.; Buttery, J.; Voss, L.; Roberts, S.; Cooper, C.; Tong, S.Y.C.; et al. Epidemiology and mortality of Staphylococcus aureus Bacteremia in Australian and New Zealand children. JAMA Pediatr. 2016, 170, 979–986. [Google Scholar] [CrossRef]
  5. World Health Organization. Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report 2021; Word Health Organization: Geneva, Switzerland, 2021. [Google Scholar]
  6. Dantes, R.; Mu, Y.; Belflower, R.; Aragon, D.; Dumyati, G.; Harrison, L.H.; Lessa, F.C.; Lynfield, R.; Nadle, J.; Petit, S.; et al. National burden of invasive methicillin-resistant Staphylococcus aureus infections, United States, 2011. JAMA Intern. Med. 2013, 173, 1970–1979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Fortuin-de Smidt, M.C.; Singh-Moodley, A.; Badat, R.; Quan, V.; Kularatne, R.; Nana, T.; Lekalakala, R.; Govender, N.P.; Perovic, O. Staphylococcus aureus bacteraemia in Gauteng academic hospitals, South Africa. Int. J. Infect. Dis. 2015, 30, e41–e48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Falagas, M.E.; Karageorgopoulos, D.E.; Leptidis, J.; Korbila, I.P. MRSA in Africa: Filling the Global Map of Antimicrobial Resistance. PLoS ONE 2013, 8, e68024. [Google Scholar] [CrossRef] [Green Version]
  9. Cassini, A.; Högberg, L.D.; Plachouras, D.; Quattrocchi, A.; Hoxha, A.; Simonsen, G.S.; Colomb-Cotinat, M.; Kretzschmar, M.E.; Devleesschauwer, B.; Cecchini, M.; et al. Attributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the European Economic Area in 2015: A population-level modelling analysis. Lancet Infect. Dis. 2018, 19, 56–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Ayobami, O.; Willrich, N.; Reuss, A.; Eckmanns, T.; Markwart, R. The ongoing challenge of vancomycin-resistant Enterococcus faecium and Enterococcus faecalis in Europe: An epidemiological analysis of bloodstream infections. Emerg. Microbes Infect. 2020, 9, 1180–1193. [Google Scholar] [CrossRef] [PubMed]
  11. Savoldi, A.; Carrara, E.; Gladstone, B.P.; Azzini, A.M.; Göpel, S.; Tacconelli, E. Gross national income and antibiotic resistance in invasive isolates: Analysis of the top-ranked antibiotic-resistant bacteria on the 2017 WHO priority list. J. Antimicrob. Chemother. 2019, 74, 3619–3625. [Google Scholar] [CrossRef]
  12. Panesso, D.; Reyes, J.; Rincón, S.; Díaz, L.; Galloway-Peña, J.; Zurita, J.; Carrillo, C.; Merentes, A.; Guzmán, M.; Adachi, J.A.; et al. Molecular epidemiology of vancomycin-resistant Enterococcus faecium: A prospective, multicenter study in South American hospitals. J. Clin. Microbiol. 2010, 48, 1562–1569. [Google Scholar] [CrossRef]
  13. Alemayehu, T.; Hailemariam, M. Prevalence of vancomycin-resistant enterococcus in Africa in one health approach: A systematic review and meta-analysis. Sci. Rep. 2020, 10, 20542. [Google Scholar] [CrossRef] [PubMed]
  14. Marincola, G.; Liong, O.; Schoen, C.; Abouelfetouh, A.; Hamdy, A.; Wencker, F.D.R.; Marciniak, T.; Becker, K.; Köck, R.; Ziebuhr, W. Antimicrobial Resistance Profiles of Coagulase-Negative Staphylococci in Community-Based Healthy Individuals in Germany. Front. Public Health 2021, 9, 684456. [Google Scholar] [CrossRef] [PubMed]
  15. May, L.; Klein, E.Y.; Rothman, R.E.; Laxminarayan, R. Trends in antibiotic resistance in coagulase-negative staphylococci in the United States, 1999 to 2012. Antimicrob. Agents Chemother. 2014, 58, 1404–1409. [Google Scholar] [CrossRef] [Green Version]
  16. Becker, K.; Heilmann, C.; Peters, G. Coagulase-negative staphylococci. Clin. Microbiol Rev. 2014, 27, 870–926. [Google Scholar] [CrossRef] [Green Version]
  17. Black, C.G.; Tavares, L.; Stachel, A.; Ratner, A.J.; Randis, T.M. Distribution of Late-Onset Neonatal Sepsis Pathogens Differs in Inpatient and Outpatient Settings. Am. J Perinatol. 2019, 36, 1136–1141. [Google Scholar] [CrossRef] [PubMed]
  18. Shane, A.L.; Sánchez, P.J.; Stoll, B.J. Neonatal sepsis. Lancet 2017, 390, 1770–1780. [Google Scholar] [CrossRef] [PubMed]
  19. National Heart, Lung and Blood Institute. Study Quality Assessment Tools. Available online: https://www.nhlbi.nih.gov/health-topics/study-quality-assessment-tools (accessed on 20 November 2022).
  20. Korczowski, B.; Antadze, T.; Giorgobiani, M.; Stryjewski, M.; Jandourek, A.; Smith, A.; O’Neal, T.; Bradley, J.S. A Multicenter, Randomized, Observer-Blinded, Active-Controlled Study to Evaluate the Safety and Efficacy of Ceftaroline versus Comparator in Pediatric Patients with Acute Bacterial Skin and Skin Structure Infection. Pediatr. Infect. Dis. J. 2016, 35, e239–e247. [Google Scholar] [CrossRef]
  21. Bradley, J.S.; Arrieta, A.C.; Digtyar, V.A.; Popejoy, M.W.; Grandhi, A.; Bokesch, P.; Hershberger, E.; Dorr, M.B.; Tan, C.M.; Murata, Y.; et al. Daptomycin for Pediatric Gram-Positive Acute Hematogenous Osteomyelitis. Pediatr. Infect. Dis. J. 2020, 39, 814–823. [Google Scholar] [CrossRef]
  22. Ardura, M.I.; Mejías, A.; Katz, K.S.; Revell, P.; McCracken, G.H.; Sánchez, P.J. Daptomycin Therapy for Invasive Gram-Positive Bacterial Infections in Children. Pediatr. Infect. Dis. J. 2007, 26, 1128–1132. [Google Scholar] [CrossRef]
  23. Moschovi, M.; Trimis, G.; Tsotra, M.; Chatzi, F.; Karamolegou, K.; Santou, A.; Tourkantoni, N.; Chrousos, G. Efficacy and safety of linezolid in immunocompromised children with cancer. Pediatr. Int. 2010, 52, 694–698. [Google Scholar] [CrossRef] [PubMed]
  24. Wible, K.; Tregnaghi, M.; Bruss, J.; Fleishaker, D.; Naberhuis-Stehouwer, S.; Hilty, M. Linezolid versus cefadroxil in the treatment of skin and skin structure infections in children. Pediatr. Infect. Dis J. 2003, 22, 315–322. [Google Scholar] [CrossRef] [PubMed]
  25. Kaplan, S.L.; Deville, J.G.; Yogev, R.; Morfin, M.R.; Wu, E.; Adler, S.; Edge-Padbury, B.; Naberhuis-Stehouwer, S.; Bruss, J.B. Linezolid versus vancomycin for treatment of resistant Gram-positive infections in children. Pediatr. Infect. Dis. J. 2003, 22, 677–685. [Google Scholar] [CrossRef] [PubMed]
  26. Isaacson, G.; Aronoff, S.C. Linezolid for tympanostomy tube otorrhea caused by methicillin-resistant Staphylococcus aureus and multiple drug-resistant Streptococcus pneumoniae. Int. J. Pediatr Otorhinolaryngol. 2008, 72, 647–651. [Google Scholar] [CrossRef]
  27. Yılmaz, A.; Dalgic, N.; Müslüman, M.; Sancar, M.; Çolak, I.; Aydın, Y. Linezolid treatment of shunt-related cerebrospinal fluid infections in children. J. Neurosurg. Pediatr. 2010, 5, 443–448. [Google Scholar] [CrossRef] [PubMed]
  28. Sung, T.-J.; Kim, H.-M.; Kim, M.-J. Methicillin-Resistant Staphylococcus aureus Endocarditis in an Extremely Low-Birth-Weight Infant Treated With Linezolid. Clin. Pediatr. 2008, 47, 504–506. [Google Scholar] [CrossRef] [PubMed]
  29. Chan, J.I.; Noor, A.; Clauss, C.; Aggarwal, R.; Nayak, A. Methicillin-Resistant Staphylococcus aureus Endovascular Infection in a Neonate: Prolonged, Safe, and Effective Use of Daptomycin and Enoxaparin. J. Pediatr. Pharmacol. Ther. 2020, 25, 68–74. [Google Scholar] [CrossRef]
  30. Jalal, Z.; Camou, F.; Roubertie, F.; Iriart, X.; Thambo, J.-B. Pediatric case of MRSA right-sided infective endocarditis with septic pulmonary embolism successfully treated by daptomycin in combination with gentamicin and rifampin. J. Pediatr. Infect. Dis. 2013, 8, 139–142. [Google Scholar] [CrossRef]
  31. Chen, A.E.; Carroll, K.C.; Diener-West, M.; Ross, T.; Ordun, J.; Goldstein, M.A.; Kulkarni, G.; Cantey, J.B.; Siberry, G.K. Randomized Controlled Trial of Cephalexin Versus Clindamycin for Uncomplicated Pediatric Skin Infections. Pediatrics 2011, 127, 573–580. [Google Scholar] [CrossRef] [Green Version]
  32. Borgmann, S.; Rieß, B.; Von Wernitz-Keibel, T.; Bühler, M.; Layer, F.; Strommenger, B. Recovery of a 10-year-old girl from methicillin- resistant Staphylococcus aureus sepsis in response to low-dose ceftaroline treatment. Ther. Clin. Risk Manag. 2016, 12, 749–753. [Google Scholar] [CrossRef] [Green Version]
  33. Al-Zaben, R.M.; Al Majali, A.; Rahayma, J.; Al-Zaben, A. The treatment of inappropriately treated Methicillin Resistant Staph Aureus osteomyelitis in pediatric age group. Rawal Med. J. 2016, 41, 212–215. [Google Scholar]
  34. Holmes, L.; Ma, C.; Qiao, H.; Drabik, C.; Hurley, C.; Jones, D.; Judkiewicz, S.; Faden, H. Trimethoprim-Sulfamethoxazole Therapy Reduces Failure and Recurrence in Methicillin-Resistant Staphylococcus aureus Skin Abscesses after Surgical Drainage. J. Pediatr. 2015, 169, 128–134.e1. [Google Scholar] [CrossRef] [PubMed]
  35. Shinjoh, M.; Iketani, O.; Watanabe, K.; Kudo, M.; Sugita, K.; Mori, T.; Hasegawa, N.; Iwata, S.; Yamagishi, H.; Shimada, H.; et al. Safety and efficacy of linezolid in 16 infants and children in Japan. J. Infect. Chemother. 2012, 18, 591–596. [Google Scholar] [CrossRef] [PubMed]
  36. Loeffler, A.M.; Drew, R.H.; Perfect, J.R.; Grethe, N.I.; Stephens, J.W.; Gray, S.L.; Talbot, G.H. Safety and efficacy of quinupristin/dalfopristin for treatment of invasive Gram-positive infections in pediatric patients. Pediatr. Infect. Dis. J. 2002, 21, 950–956. [Google Scholar] [CrossRef] [PubMed]
  37. Fusco, N.M.; Meaney, C.J.; Frederick, C.A.; Prescott, W.A. Comparative Effectiveness of Vancomycin Versus Linezolid for the Treatment of Acute Pulmonary Exacerbations of Cystic Fibrosis. Ann. Pharmacother. 2020, 54, 197–204. [Google Scholar] [CrossRef] [PubMed]
  38. Bradley, J.; Glasser, C.; Patino, H.; Arnold, S.R.; Arrieta, A.; Congeni, B.; Daum, R.S.; Kojaoghlanian, T.; Yoon, M.; Anastasiou, D.; et al. Daptomycin for Complicated Skin Infections: A Randomized Trial. Pediatrics 2017, 139, e20162477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Iwata, S.; Koyama, H.; Murata, Y. Efficacy and safety of daptomycin in Japanese pediatric participants with complicated skin and soft tissue infections or bacteremia caused by gram-positive cocci. J. Infect. Chemother. 2022, 28, 406–412. [Google Scholar] [CrossRef]
  40. Cook, A.M.; Ramsey, C.N.; Martin, C.A.; Pittman, T. Linezolid for the Treatment of a Heteroresistant Staphylococcus aureus Shunt Infection. Pediatr. Neurosurg. 2005, 41, 102–104. [Google Scholar] [CrossRef]
  41. Yogev, R.; Patterson, L.E.; Kaplan, S.L.; Adler, S.; Morfin, M.R.; Martin, A.; Edge-Padbury, B.; Naberhuis-Stehouwer, S.; Bruss, J.B. Linezolid for the treatment of complicated skin and skin structure infections in children. Pediatr. Infect. Dis J. 2003, 22, 172–177. [Google Scholar] [CrossRef]
  42. Gallagher, R.; Pizer, B.; Ellison, J.; Riordan, F. Glycopeptide insensitive Staphylococcus aureus subdural empyema treated with linezolid and rifampicin. J. Infect. 2008, 57, 410–413. [Google Scholar] [CrossRef]
  43. Chen, C.-J.; Chiu, C.-H.; Lin, T.-Y.; Lee, Z.-L.; Yang, W.-E.; Huang, Y.-C. Experience With Linezolid Therapy in Children With Osteoarticular Infections. Pediatr. Infect. Dis. J. 2007, 26, 985–988. [Google Scholar] [CrossRef]
  44. Jacobson, L.M.; Milstone, A.; Zenilman, J.; Carroll, K.C.; Arav-Boger, R. Daptomycin therapy failure in an adolescent with methicillin-resistant Staphylococcus aureus bacteremia. Pediatr. Infect. Dis. J. 2009, 28, 445–447. [Google Scholar] [CrossRef] [PubMed]
  45. Salerno, S.N.; Bernhardt, J.; Laughon, M.; Jhaveri, R.; Massaro, M.; Gonzalez, D. Pharmacokinetics of Ceftaroline in a Preterm Infant with Methicillin-Resistant Staphylococcus aureus Pneumonia. J. Pediatr. Infect. Dis. Soc. 2018, 7, 342–345. [Google Scholar] [CrossRef] [PubMed]
  46. Palma, D.M.; Giordano, S.; Cracchiolo, A.N.; Zangara, V.; Coffaro, G.; Tetamo, R. Daptomycin in the treatment of invasive Gram-positive bacterial infections in children: Personal experience. Minerva Pediatr. 2013, 65, 173–178. [Google Scholar]
  47. Hussain, A.; Kairamkonda, V.; Jenkins, D.R. Successful treatment of meticillin-resistant Staphylococcus aureus bacteraemia in a neonate using daptomycin. J. Med. Microbiol. 2011, 60, 381–383. [Google Scholar] [CrossRef] [PubMed]
  48. Arrieta, A.C.; Bradley, J.S.; Popejoy, M.W.; Bensaci, M.; Grandhi, A.; Bokesch, P.; Glasser, C.; Du, L.; Patino, H.; Kartsonis, N.A. Randomized Multicenter Study Comparing Safety and Efficacy of Daptomycin Versus Standard-of-care in Pediatric Patients With Staphylococcal Bacteremia. Pediatr. Infect. Dis. J. 2018, 37, 893–900. [Google Scholar] [CrossRef]
  49. Şahin, A.; Dalgic, N. Intravenous and intraventricular daptomycin plus intravenous linezolid treatment of an infant with Vancomycin-Resistant Enterococci induced ventriculoperitoneal shunt infection. World Neurosurg. 2019, 124, 328–330. [Google Scholar] [CrossRef]
  50. Şahin, A.; Dalgic, N. Intraventricular plus Intravenous Tigecycline for the Treatment of Daptomycin Nonsusceptible Vancomycin-Resistant Enterococci in an Infant with Ventriculoperitoneal Shunt Infection. World Neurosurg. 2019, 130, 470–473. [Google Scholar] [CrossRef]
  51. Jaspan, H.B.; Brothers, A.W.; Campbell, A.J.P.; McGuire, J.K.; Browd, S.R.; Manley, T.J.; Pak, D.; Weissman, S.J. Multidrug-Resistant Enterococcus faecium Meningitis in a Toddler: Characterization of the Organism and Successful Treatment with Intraventricular Daptomycin and Intravenous Tigecycline Heather. Pediatr. Infect. Dis. J. 2016, 29, 379–381. [Google Scholar] [CrossRef] [Green Version]
  52. Hoehn, R.; Groll, A.; Schaefer, V.; Bauer, K.; Schloesser, R. Linezolid treatment of glycopeptide-resistant Enterococcus faecium in very low birth weight premature neonates. Int. J. Antimicrob. Agents 2006, 27, 256–258. [Google Scholar] [CrossRef]
  53. Fossati, M.; Cappelli, B.; Biral, E.; Chiesa, R.; Biffi, A.; Ossi, C.; Moro, M.; Cirillo, D.M.; Clementi, M.; Soliman, C.; et al. Case Report Fatal vancomycin- and linezolid-resistant Enterococcus faecium sepsis in a child undergoing allogeneic haematopoietic stem cell transplantation for beta-thalassaemia major. J. Med. Microbiol. 2010, 59, 839–842. [Google Scholar] [CrossRef] [Green Version]
  54. Paul, K.; Merabishvili, M.; Hazan, R.; Christner, M.; Herden, U.; Gelman, D.; Khalifa, L.; Yerushalmy, O.; Coppenhagen-Glazer, S.; Harbauer, T.; et al. Bacteriophage Rescue Therapy of a Vancomycin-Resistant Enterococcus faecium Infection in a One-Year-Old Child following a Third Liver Transplantation. Viruses 2021, 13, 1785. [Google Scholar] [CrossRef] [PubMed]
  55. Gray, J.W.; Darbyshire, P.J.; Beath, S.V.; Kelly, D. Experience with quinupristin/dalfopristin in treating infections with vancomycin-resistant Enterococcus faecium in children. Pediatr. Infect. Dis. J. 2000, 19, 234–238. [Google Scholar] [CrossRef] [PubMed]
  56. Ang, J.Y.; Lua, J.L.; Turner, D.R.; Asmar, B.I. Vancomycin-resistant Enterococcus faecium endocarditis in a premature infant successfully treated with linezolid. Pediatr. Infect. Dis. J. 2003, 22, 1101–1103. [Google Scholar] [CrossRef] [PubMed]
  57. Baysallar, M.; Izci, Y.; Kilic, A.; Avci, I.Y.; Senses, Z.; Doganci, L.; Tamma, P.D.; Hsu, A.J.; Wang, J.S.; Muzevich, K.; et al. A Case of Ventricular Drainage Infection with a Rare Pathogen in Cerebrospinal Fluid: Vancomycin-Resistant Enterococcus faecium. Microbiol. Drug Resist. 2006, 12, 59–62. [Google Scholar] [CrossRef] [PubMed]
  58. Travaglianti, M.; Pérez, M.; Sberna, N.; Rousseau, M.; Calle, G.; Gómez, S. Tratamiento de infecciones por Enterococcus resistente a vancomicina con linezolid en un hospital pediátrico. Farm. Hosp. 2007, 31, 43–47. [Google Scholar] [CrossRef] [PubMed]
  59. Graham, P.L., III; Ampofo, K.; Saiman, L. Linezolid treatment of vancomycin-resistant Enterococcus faecium ventriculitis. Pediatr. Infect. Dis. J. 2002, 21, 798–800. [Google Scholar] [CrossRef] [PubMed]
  60. Beneri, C. Successful treatment of a neonate with persistent vancomycin-resistant enterococcal bacteremia with a daptomycin-containing regimen. Infect. Drug Resist. 2008, 1, 9–11. [Google Scholar] [CrossRef] [Green Version]
  61. Maranich, A.M.; Rajnik, M. Successful treatment of vancomycin-resistant enterococcal ventriculitis in a pediatric patient with linezolid. Mil. Med. 2008, 173, 927–929. [Google Scholar] [CrossRef] [Green Version]
  62. Ergaz, Z.; Arad, I.; Bar-Oz, B.; Peleg, O.; Benenson, S.; Minster, N.; Moses, A.; Block, C. Elimination of vancomycin-resistant enterococci from a neonatal intensive care unit following an outbreak. J. Hosp. Infect. 2010, 74, 370–376. [Google Scholar] [CrossRef]
  63. Shanti, R.; Ic, S.; Ariffin, H. Intraventricular teicoplanin. J. Health Transl. Med. 2009, 12, 35–38. [Google Scholar]
  64. Minotti, C.; Zuccon, I.; Priante, E.; Bonadies, L.; Di Chiara, C.; Donà, D.; Baraldi, E.; Costenaro, P. Daptomycin for Treatment of S. epidermidis Endocarditis in an Extremely Preterm Neonate—Outcome and Perspectives. Children 2022, 9, 457. [Google Scholar] [CrossRef] [PubMed]
  65. Ochi, F.; Tauchi, H.; Nagai, K.; Moritani, K.; Tezuka, M.; Jogamoto, T.; Aibara, K.; Motoki, T.; Ishii, E. Therapeutic Effect of Linezolid in Children With Health Care—Associated Meningitis or Ventriculitis. Clin. Pediatr. 2018, 57, 1672–1676. [Google Scholar] [CrossRef] [PubMed]
  66. Gawronski, K.M. Successful Use of Daptomycin in a Preterm Neonate With Persistent Methicillin-Resistant Staphylococcus epidermidis Bacteremia. J. Pediatr. Pharmacol. Ther. 2015, 20, 61–65. [Google Scholar] [CrossRef] [PubMed]
  67. Magiorakos, A.-P.; Srinivasan, A.; Carey, R.B.; Carmeli, Y.; Falagas, M.E.; Giske, C.G.; Harbarth, S.; Hindler, J.F.; Kahlmeter, G.; Olsson-Liljequist, B.; et al. 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]
  68. Liu, C.; Bayer, A.; Cosgrove, S.E.; Daum, R.S.; Fridkin, S.K.; Gorwitz, R.J.; Kaplan, S.L.; Karchmer, A.W.; Levine, D.P.; Murray, B.E.; et al. Clinical practice guidelines by the infectious diseases society of america for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clin. Infect. Dis. 2011, 52, e18–e55. [Google Scholar] [CrossRef] [Green Version]
  69. McMullan, B.J.; Campbell, A.J.; Blyth, C.C.; McNeil, J.C.; Montgomery, C.P.; Tong, S.Y.; Bowen, A.C. Clinical Management of Staphylococcus aureus Bacteremia in Neonates, Children, and Adolescents. Pediatrics 2020, 146, e20200134. [Google Scholar] [CrossRef]
  70. FDA Rationale for Recognition Decision: Ceftaroline Fosamil. Published 17 April 2020. Available online: https://www.fda.gov/drugs/development-resources/fda-rationale-recognition-decision-ceftaroline-fosamil (accessed on 25 November 2022).
  71. European Medicines Agency (EMA). Zinforo (Ceftaroline Fosamil). 2022. Last update 27 July 2022. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zinforo (accessed on 25 November 2022).
  72. Bradley, J.S.; Stone, G.G.; Chan, P.L.S.; Raber, S.R.; Riccobene, T.; Casullo, V.M.; Yan, J.L.; Hendrick, V.M.; Hammond, J.; Leister-Tebbe, H.K. Phase 2 study of the safety, pharmacokinetics and efficacy of ceftaroline fosamil in neonates and very young infants with late-onset sepsis. Pediatr. Infect. Dis. J. 2020, 39, 411–418. [Google Scholar] [CrossRef]
  73. Branstetter, J.; Searcy, H.; Benner, K.; Yarbrough, A.; Crowder, C.; Troxler, B. Ceftaroline vs vancomycin for the treatment of acute pulmonary exacerbations in pediatric patients with cystic fibrosis. Pediatr. Pulmonol. 2020, 55, 3337–3342. [Google Scholar] [CrossRef]
  74. Yim, J.; Molloy, L.M.; Newland, J.G. Use of Ceftaroline Fosamil in Children: Review of Current Knowledge and its Application. Infect. Dis. Ther. 2017, 6, 57–67. [Google Scholar] [CrossRef] [Green Version]
  75. Fowler, V.G., Jr.; Boucher, H.W.; Corey, G.R.; Abrutyn, E.; Karchmer, A.W.; Rupp, M.E.; Levine, D.P.; Chambers, H.F.; Tally, F.P.; Vigliani, G.A.; et al. Daptomycin versus Standard Therapy for Bacteremia and Endocarditis Caused by Staphylococcus aureus. N. Engl. J. Med. 2006, 355, 653–665. [Google Scholar] [CrossRef] [Green Version]
  76. Arbeit, R.D.; Maki, D.; Tally, F.P.; Campanaro, E.; Eisenstein, B.I.; Daptomycin 98-01 and 99-01 Investigators. The safety and efficacy of daptomycin for the treatment of complicated skin and skin-structure infections. Infect. Dis. Clin. Pract. 2004, 38, 1673–1681. [Google Scholar] [CrossRef] [PubMed]
  77. U.S. Food and Drug Administration. Drug Approval Package. Zyvox (Linezolid). 2002. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2002/21-130s003_21131s003_21132s003_ZyvoxTOC.cfm (accessed on 23 November 2022).
  78. Kaplan, S.L.; Afghani, B.; Lopez, P.; Wu, E.; Fleishaker, D.; Edge-Padbury, B.; Naberhuis-Stehouwer, S.; Bruss, J.B. Linezolid for the treatment of methicillin-resistant Staphylococcus aureus infections in children. Pediatr. Infect. Dis. J. 2003, 22 (Suppl. 9), 178–185. [Google Scholar] [CrossRef] [PubMed]
  79. Deville, J.G.; Adler, S.; Azimi, P.H.; Jantausch, B.A.; Morfin, M.R.; Beltran, S.; Edge-Padbury, B.; Naberhuis-Stehouwer, S.; Bruss, J.B. Linezolid versus vancomycin in the treatment of known or suspected resistant Gram-positive infections in neonates. Pediatr. Infect. Dis. J. 2003, 22 (Suppl. 9), 158–163. [Google Scholar] [CrossRef] [PubMed]
  80. Minotti, C.; Bonadies, L.; Liberati, C.; De Pieri, M.; Giaquinto, C.; Baraldi, E.; Donà, D. Enteral Linezolid as an Effective Option to Treat an Extremely Preterm Infant with Bacillus cereus Sepsis. Children 2022, 9, 415. [Google Scholar] [CrossRef]
  81. Shariati, A.; Dadashi, M.; Moghadam, M.T.; van Belkum, A.; Yaslianifard, S.; Darban-Sarokhalil, D. Global prevalence and distribution of vancomycin resistant, vancomycin intermediate and heterogeneously vancomycin intermediate Staphylococcus aureus clinical isolates: A systematic review and meta-analysis. Sci. Rep. 2020, 10, 12689. [Google Scholar] [CrossRef]
  82. Wu, Q.; Sabokroo, N.; Wang, Y.; Hashemian, M.; Karamollahi, S.; Kouhsari, E. Systematic review and meta-analysis of the epidemiology of vancomycin-resistance Staphylococcus aureus isolates. Antimicrob. Resist. Infect. Control. 2021, 10, 101. [Google Scholar] [CrossRef] [PubMed]
  83. Ocampo, P.S.; Lázár, V.; Papp, B.; Arnoldini, M.; Wiesch, P.A.Z.; Busa-Fekete, R.; Fekete, G.; Pál, C.; Ackermann, M.; Bonhoeffer, S. Antagonism between bacteriostatic and bactericidal antibiotics is prevalent. Antimicrob. Agents Chemother. 2014, 58, 4573–4582. [Google Scholar] [CrossRef] [Green Version]
  84. European Medicines Agency (EMA). Sivextro, Tedizolid Phosphate. 2015. Last update 16 December 2022. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/sivextro (accessed on 9 January 2023).
  85. Bradley, J.S.; Antadze, T.; Ninov, B.; Tayob, M.S.; Broyde, N.; Butterton, J.R.; Chou, M.Z.; De Anda, C.S.; Kim, J.Y.; Sears, P.S. Safety and Efficacy of Oral and/or Intravenous Tedizolid Phosphate from a Randomized Phase 3 Trial in Adolescents with Acute Bacterial Skin and Skin Structure Infections. Pediatr. Infect. Dis. J. 2021, 40, 238–244. [Google Scholar] [CrossRef] [PubMed]
  86. Raad, I.; Hachem, R.; Hanna, H.; Afif, C.; Escalante, C.; Kantarjian, H.; Rolston, K. Prospective, randomized study comparing quinupristin-dalfopristin with linezolid in the treatment of vancomycin-resistant Enterococcus faecium infections. J. Antimicrob. Chemother. 2004, 53, 646–649. [Google Scholar] [CrossRef] [Green Version]
  87. Chong, Y.P.; Lee, S.-O.; Song, E.H.; Lee, E.J.; Jang, E.-Y.; Kim, S.-H.; Choi, S.-H.; Kim, M.-N.; Jeong, J.-Y.; Woo, J.H.; et al. Quinupristin-dalfopristin versus linezolid for the treatment of vancomycin-resistant Enterococcus faecium bacteraemia: Efficacy and development of resistance. Scand. J. Infect. Dis. 2010, 42, 491–499. [Google Scholar] [CrossRef]
  88. Koulenti, D.; Xu, E.; Yin Sum Mok, I.; Song, A.; Karageorgopoulos, D.E.; Armaganidis, A.; Lipman, J.; Tsiodras, S. Novel Antibiotics for Multidrug-Resistant Gram-Positive Microorganisms. Microorganisms 2019, 7, 270. [Google Scholar] [CrossRef] [Green Version]
  89. Rosenthal, S.; Decano, A.G.; Bandali, A.; Lai, D.; E Malat, G.; E Bias, T. Oritavancin (Orbactiv) a new-generation lipoglycopeptide for the treatment of acute bacterial skin and skin structure infections. Pharm. Ther. 2018, 43, 143–147. [Google Scholar]
  90. Open-Label, Dose-Finding, Pharmacokinetics, Safety and Tolerability Study of Oritavancin in Pediatric Patients with Suspected or Confirmed Bacterial Infections (NCT02134301). ClinicalTrials.gov. Last update 17 January 2023. Available online: https://clinicaltrials.gov/ct2/show/NCT02134301 (accessed on 21 January 2023).
  91. European Medicines Agency (EMA). Xydalba, Dalbavancin. 2015. Last update 14 December 2022. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/xydalba (accessed on 9 January 2023).
  92. Dalbavancin for the Treatment of Acute Bacterial Skin and Skin Structure Infections in Children, Known or Suspected to Be Caused by Susceptible Gram-Positive Organisms, Including MRSA-Full Text View-ClinicalTrials.gov. Last update 17 October 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT02814916 (accessed on 25 November 2022).
  93. Bradley, J.S.; Benziger, D.; Bokesch, P.; Jacobs, R. Single-dose pharmacokinetics of daptomycin in pediatric patients 3-24 months of age. Pediatr. Infect. Dis. J. 2014, 33, 936–939. [Google Scholar] [CrossRef]
  94. Mohzari, Y.; Aljobair, F.; Alrashed, A.; Asdaq, S.; Alshuraim, R.; Asfour, S.; Al-Mouqdad, M.; Bamogaddam, R.; Al-Anazi, D.; Zeilinger, C.; et al. Safety and efficacy of daptomycin in neonates with coagulase-negative staphylococci: Case series analysis. Antibiotics 2021, 10, 168. [Google Scholar] [CrossRef]
  95. Garazzino, S.; Tovo, P.-A. Clinical experience with linezolid in infants and children. J. Antimicrob. Chemother. 2011, 66 (Suppl. 4), iv23–iv41. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the study selection process.
Figure 1. Flowchart of the study selection process.
Antibiotics 12 00261 g001
Figure 2. Results regarding the choice of treatment and clinical success for methicillin-resistant Staphylococcus aureus (MRSA) infections. Clinical success was defined as complete resolution or substantial improvement of the signs and symptoms of the index infection. BSI: bloodstream infection; SSSI: skin and structure skin infections, CLABSI: central line-associated bloodstream infection, HAP: hospital-acquired pneumonia.
Figure 2. Results regarding the choice of treatment and clinical success for methicillin-resistant Staphylococcus aureus (MRSA) infections. Clinical success was defined as complete resolution or substantial improvement of the signs and symptoms of the index infection. BSI: bloodstream infection; SSSI: skin and structure skin infections, CLABSI: central line-associated bloodstream infection, HAP: hospital-acquired pneumonia.
Antibiotics 12 00261 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chiusaroli, L.; Liberati, C.; Rulli, L.; Barbieri, E.; De Pieri, M.; Di Chiara, C.; Mengato, D.; Giaquinto, C.; Donà, D. Therapeutic Options and Outcomes for the Treatment of Children with Gram-Positive Bacteria with Resistances of Concern: A Systematic Review. Antibiotics 2023, 12, 261. https://doi.org/10.3390/antibiotics12020261

AMA Style

Chiusaroli L, Liberati C, Rulli L, Barbieri E, De Pieri M, Di Chiara C, Mengato D, Giaquinto C, Donà D. Therapeutic Options and Outcomes for the Treatment of Children with Gram-Positive Bacteria with Resistances of Concern: A Systematic Review. Antibiotics. 2023; 12(2):261. https://doi.org/10.3390/antibiotics12020261

Chicago/Turabian Style

Chiusaroli, Lorenzo, Cecilia Liberati, Luigi Rulli, Elisa Barbieri, Marica De Pieri, Costanza Di Chiara, Daniele Mengato, Carlo Giaquinto, and Daniele Donà. 2023. "Therapeutic Options and Outcomes for the Treatment of Children with Gram-Positive Bacteria with Resistances of Concern: A Systematic Review" Antibiotics 12, no. 2: 261. https://doi.org/10.3390/antibiotics12020261

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