Next Article in Journal
Addressing Consumer Misconceptions on Antibiotic Use and Resistance in the Context of Sore Throat: Learnings from Social Media Listening
Next Article in Special Issue
Bloodstream Infection and Gram-Negative Resistance: The Role for Newer Antibiotics
Previous Article in Journal
Development and Validation of a Prognostic Model for Multi-Drug-Resistant Non-Hospital-Acquired Bloodstream Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

New Antimicrobials for the Treatment of Neonatal Sepsis Caused by Multi-Drug-Resistant Bacteria: A Systematic Review

1
Neonatal Intensive Care Unit, Department of Mother and Child Care, Careggi University Hospital, 50141 Florence, Italy
2
Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, 50141 Florence, Italy
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(6), 956; https://doi.org/10.3390/antibiotics12060956
Submission received: 30 April 2023 / Revised: 19 May 2023 / Accepted: 22 May 2023 / Published: 24 May 2023

Abstract

:
Background: Infections by multi-drug-resistant (MDR) organisms are sharply increasing in newborns worldwide. In low and middle-income countries, a disproportionate amount of neonatal sepsis caused by MDR Gram negatives was recently reported. Newborns with infections by MDR organisms with limited treatment options may benefit from novel antimicrobials. Methods: We performed a literature search investigating the use in newborns, infants and children of novel antimicrobials for the treatment of MDR Gram negatives, namely ceftazidime/avibactam, ceftolozane/tazobactam, cefiderocol, meropenem/vaborbactam, imipenem/relebactam, and Gram positives with resistance of concern, namely ceftaroline and dalbavancin. PubMed, EMBASE, and Web of Science were searched. Results: A total of 50 records fulfilled the inclusion criteria. Most articles were case reports or case series, and ceftazidime/avibactam was the most studied agent. All studies showed favorable efficacy and safety profile in newborns and across different age cohorts. Conclusions: novel antibiotics may be considered in newborns for the treatment of MDR Gram negatives with limited treatment options and for Gram positives with resistance concerns. Further studies are needed to address their effectiveness and safety in newborns.

1. Introduction

Sepsis is among the leading causes of neonatal mortality and morbidity worldwide [1], accounting for about 1.3 (0.8–2.3 95% CI) million cases and 0.23 (0.179–0.276 95% CI) million neonatal deaths each year [2,3,4]. Antimicrobial resistance is globally spreading in the neonatal population [5,6,7], with particular concerns in low-income and middle-income countries (LMICs), while access to effective antimicrobials is still crucially limited in several regions [1].
Early-onset sepsis (EOS) is defined as a positive blood or cerebrospinal fluid culture taken within the first 72 h of life, while late-onset sepsis (LOS) occurs after the first 3 days of life [5,6]. Multi-drug resistance (MDR) bacteria were alarmingly reported in the latest years as causing agents of both EOS and LOS [6,7,8,9]. Moreover, several outbreaks caused by MDR organisms in Neonatal Intensive Care Units (NICUs) recently occurred in different regions, including high-income countries (HICs) [10,11,12,13,14], and consistent colonization of both patients admitted to NICUs [15,16,17] and of pregnant women [18] is reported, threatening the outcomes of both EOS and LOS cases.

1.1. Epidemiology

Among 0.68 million annual neonatal deaths associated with possible severe bacterial infection, an estimated 31% were attributable to resistant pathogens worldwide, with disproportionate risk in India, Pakistan, Nigeria, Congo, and China [1]. Studies from LMICs recently reported a higher amount of EOS in comparison HICs, accounting for 70–80% of all neonatal sepsis [2,9], and a high prevalence of Gram-negative strains, including MDR bacteria, in both EOS and LOS [2,6,8,19]. Gram negatives caused 39–64% of all neonatal sepsis [6,7,8] and the most frequently isolated organisms were Klebsiella spp., S. marcescens, E. coli, Enterobacter, and A. baumannii [6,8]. In recent studies, Gram-negative strains causing neonatal sepsis in LMICs across Africa and Asia were resistant to aminoglycosides in approximately 70% of cases [6,7], to cephalosporins in up to 84% of cases [5,6] and to carbapenems in 16–81% of cases [5,6,7]. In China, E. coli and Klebsiella spp. causing neonatal sepsis were reported as MDR organisms in 42 and 61% of cases, respectively [20,21], while carbapenem resistance was found in up to 31% of cases of LOS [21,22].
In HICs, E. coli is responsible for 35% of all EOS, and 51% of EOS in preterm infants [23], and Gram negatives account for 15–30% of LOS [24]. In the US, E. coli causing EOS displayed resistance to gentamicin in 10% of cases [25,26], while among all E. coli isolates from neonatal sepsis resistance to aminoglycosides was found in 14–17% of cases [25,27]. Carbapenem resistance was <5% [27]; however, 25% of all Gram negatives were resistant to at least one antimicrobial among gentamicin, piperacillin-tazobactam, 3rd–4th generation cephalosporins and carbapenems [26]. In the UK, 41% of Enterobacter spp. causing LOS showed resistance to the recommended combination of amoxicillin and cefotaxime and 18% to the combination of benzylpenicillin and gentamicin, while 15% of E. coli and 12% of Klebsiella spp. were resistant to both [28]. A study from Greece reported that Klebsiella spp. causing LOS were resistant to at least one aminoglycoside in almost half of cases [29]. In Germany, MDR organisms accounted for about 4% of LOS and 8% of EOS, with a predominance of extended-spectrum beta-lactamase (EBSL)-producing E. coli [30].
Despite routine screening for EBSL-producing and carbapenem-resistant Enterobacterales (CRE) is not performed in NICUs, colonization of admitted patients is increasingly documented worldwide [15,17,31,32]. Colonization by CRE at NICU admission was reported in 21–30% by studies from Vietnam and Turkey [15,31], and significant colonization acquisition during NICU stay was observed [15]. Colonization of 4% of admitted patients by MDR organisms was reported in a Belgian NICU [33] and in Italy, CRE were found to be significant colonizing agents of newborns admitted to different intensive care facilities [17,34].
Gram-positive strains, such as Coagulase-negative Staphylococcus (CONS), S. aureus, and Enterococcus are at present the most frequent agents of nosocomial LOS in HICs [28,29] and vancomycin is among the most prescribed drugs in NICU [35]. Vancomycin-resistant Enterococci (VRE) are increasingly reported in neonatal sepsis worldwide [29,36,37] and accounted for 14% of all LOS in a network of Greek NICUs [29]. Resistance to glycopeptides of Gram-positive strains was reported in 13% of neonatal sepsis in India and 45% in Nigeria [5].

1.2. Current Treatment Options

To date, treatment options for MDR organisms in NICU are alarmingly limited [38,39,40], particularly for Gram-negative strains. Colistin is the main used antimicrobial for the treatment of MDR P. aeruginosa, A. baumannii, and CRE in newborns in the two last decades, with 75–100% of clinical success [41]. Meropenem at high doses or as an extended infusion or in association with other antimicrobials is the second most reported agent [40,41], while the use of tigecycline, fluoroquinolones, and polymyxin B is less frequently documented [24,40,42,43]. However, the polymyxins safety profile is not optimal, as nephrotoxicity has been reported in newborns in 10–19% of cases [42] along with significant electrolyte imbalances [42]. Treatment of Gram positives, such as methicillin-resistant S. aureus (MRSA), VRE, and CONS, with unfavorable susceptibility profiles or poor clinical response to oxacillin or vancomycin, is mainly based on linezolid and daptomycin [36,44]. However, linezolid showed variable clinical responses with clinical cure rates ranging from 63 to 100%, while the use of daptomycin might be questionable in the case of pneumonia [44].
Different antimicrobials are approved in adults for the treatment of infections caused by organisms with unfavorable susceptibility profiles. Particularly, beta-lactams/beta-lactamase inhibitors and cefiderocol are currently the cornerstones of the treatment of bloodstream infections (BSIs) and infections of different sites caused by Gram-negative strains with limited treatment options [45,46,47,48,49]. Ceftaroline and dalbavancin are among the main treatment options for Gram-positive strains with resistance concern [50,51], alongside with lipoglycopeptides such as televancin (not approved for use in EU) and oritavancin [52,53], the novel oxazolidinone tedizolid [54] and the 4th generation cephalosporin ceftobiprole [55]. Finally, eravacycline, a novel tetracycline with activity against Gram-positive cocci and Gram-negative bacilli, is considered for the treatment of intra-abdominal infections caused by susceptible strains [56].
Given the shortage of current treatment options in newborns, newly available antimicrobials such as beta-lactams/beta-lactamase inhibitors, namely ceftazidime/avibactam, ceftolozane/tazobactam, meropenem/vaborbactam, imipenem/relebactam, or cefiderocol may represent promising tools for the treatment of MDR Gram negatives in NICU, while ceftaroline and dalbavancin may represent treatment options for Gram-positive strains with resistances of concern. Despite the use of these antimicrobials has been increasingly reported in latest years in newborns and infants, to date, the possibility to extend their use to these populations has not been assessed. Therefore, this study aimed to review the current knowledge of the use of these antimicrobials in newborns.

2. Results

2.1. Study Selection

Among a total of 986 records retrieved, 749 records were removed (736 duplicates); therefore, 237 records were screened and 196 were removed as they did not meet eligibility criteria. Therefore, a total of 68 articles were sought for retrieval and 18 articles were excluded as commentaries or narrative reviews not reporting original data or as studies documenting only antimicrobial susceptibility data. We decided not to include susceptibility studies as they did not provide data directly related to antimicrobials administration in clinical settings. Instead, we decided to include studies on pharmacokinetic simulation models, although not enrolling patients in clinical settings, as they were considered useful for the validation of administration schedules in the age groups of interest. Finally, 50 articles [57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106] were included in the present systematic review (Figure 1).

2.2. Treatment of MDR Gram-Negative Bacteria

We included 35 articles regarding the treatment of MDR Gram negatives with antimicrobials of interest, 16 on ceftazidime/avibactam [57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72], 12 on ceftolozane-tazobactam [72,73,74,75,76,77,78,79,80,81,82], 6 on cefiderocol [84,85,86,87,88,89,90], and 2 on meropenem-vaborbactam [90,91]. No eligible studies were retrieved for imipenem-relebactam. One study reported on both ceftazidime/avibactam and ceftolozane/tazobactam [72] and one [90] on both meropenem/vaborbactam and cefiderocol.

2.2.1. Ceftazidime/Avibactam

Among 16 studies on ceftazidime/avibactam (Table 1), we included 12 case reports or case series, 2 randomized controlled trials (RCTs), and 2 pharmacokinetic studies. Case reports and case series included patients from birth to 16 years of age. In particular, 5/12 case reports or series [59,62,66,69,70] included newborns for a total of 11 newborns infected by carbapenem-resistant or extensively drug-resistant K. pneumoniae, 8 with LOS, 2 with LOS and meningitidis, and 1 with UTI, and 10/11 newborns achieved clinical cure; 4/5 studies on newborns included only preterm newborns [62,66,69,70], while 1 case series included both term and preterm newborns [59]. No significant drug-related AEs were reported; 2 mild AEs with an uncertain relationship with ceftazidime/avibactam were reported.
Included RCTs [60,61] enrolled patients ≥3 months old with urinary tract infections (UTIs) or complicated intra-abdominal infection, clinical cure achieved by ceftazidime/avibactam ranged from 95 to 98%.
Doses and administration schedules for ceftazidime/avibactam in newborns are at present extrapolated from pharmacokinetic data obtained from patients ≥3 months old [57,68].

2.2.2. Ceftolozane/Tazobactam

Among 12 studies on ceftolozane/tazobactam (Table 2), we included 5 case reports or case series, 2 RCTs, and 5 pharmacokinetics studies. One RCT enrolled newborns with complicated UTIs [82] while the other one [83] enrolled patients >2 years of age with complicated intra-abdominal infections.
Case reports and case series and one retrospective study reported the use of ceftolozane/tazobactam in a total of 21 pediatric patients with infections of different sites (BSI, pneumonia, osteomyelitis, intra-abdominal infection) caused by MDR P. aeruginosa [72,73,75,76,79] with age ranging from 3 months to 18 years.
No severe drug-related AEs were reported, mild drug-related AEs included diarrhea, increased transaminases, and neutropenia [82].
Current doses and treatment regimens for ceftolozane/tazobactam were validated by pharmacokinetic studies specifically targeting newborns [74,77,78].

2.2.3. Cefiderocol

Among the 6 included studies on cefiderocol (Table 3), 5 were case reports or series, and 1 was a pharmacokinetic study. In the case reports, 2/6 reported the use of cefiderocol in 2 preterm newborns with carbapenem-resistant K. pneumoniae [88,89], one with LOS [89], and one with LOS and necrotizing enterocolitis [88].
A pharmacokinetic simulation model validated doses and administration regimens for cefiderocol in newborns, providing specific doses basing on post-natal age and gestational age of the patients [84].

2.2.4. Meropenem/Vaborbacatam

We identified 2 case reports on meropenem/vaborbactam regarding patients of 4 and 10 years [90,91] but no reports regarding newborns (Table 3).

2.3. Treatment of Gram-Positive Bacteria with Resistance of Concerns

We included 15 articles regarding the treatment of MDR Gram positives with antimicrobials of interest, 11 on ceftaroline [92,93,94,95,96,97,98,99,100,101,102] and 4 on dalbavancin [103,104,105,106].

2.3.1. Ceftaroline

Among 11 studies on ceftaroline (Table 4), we included 3 RCTs, 1 clinical phase 2 trial, 3 case reports, 3 pharmacokinetics studies, and 1 retrospective study. Ceftaroline was studied in RCTs for the treatment of pneumonia and skin and skin structure infections (SSSIs) mostly caused by S. aureus, including MRSA, in patients ≥2 months of age [92,93,94], achieving clinical cure in 83–88% of cases [92,93,94]. Among case reports, 2/3 documented the use of ceftaroline in preterm newborns of 24–30 weeks of gestation infected by MRSA, one with LOS and pneumonia [96], and one with hepatic abscess and infected thrombus of the portal system [100].
Drug-related AEs were reported in 10–23% of treated patients [92,93]; the most frequently reported AEs were diarrhea, vomiting, dermatitis or rush, increased transaminases; two severe drug-related AEs were reported [92,93], one hypersensitivity event and one case of colitis by C. difficilis [94].
Pharmacokinetic studies specifically targeted to neonatal age [95,101,102] validated ceftaroline administration schedule of 8–10 mg/kg q8h [95,101] and demonstrated no differences in the probability of target attainment between 5 min or 60 min drug infusion [102].

2.3.2. Dalbavancin

We included 4 studies on dalbavancin (Table 4), 3 pharmacokinetic studies, and one RCT. In the included RCT [106] dalbavancin was administered for the treatment of BSI in patients from birth to 3 months and SSSIs from birth to 18 years.
Drug-related AEs were not reported.

3. Discussion

We identified 50 articles regarding the use of the antibiotics of interest, namely ceftazidime/avibactam, ceftolozane/tazobactam, cefiderocol, meropenem/vaborbactam, imipenem/relebactam, ceftaroline and dalbavancin in newborns, infants, and children.
Most articles were case reports or case series or other retrospective studies, while few neonatal patients were enrolled in RCTs; the most studied antimicrobials were ceftazidime/avibactam and ceftolozane/tazobactam. The most frequently isolated MDR organisms in the included studies were K. pneumoniae, P. aeruginosa, and E. coli among Gram negatives, and MSSA, MRSA, and Enterococcus spp. among Gram positives.
Ceftazidime/avibactam is a cephalosporin/beta-lactamase inhibitor, with excellent activity against KPC and OXA-48-like producing CRE and non-carbapenemase-producing CRE, and it is currently approved for use in patients ≥3 months for the treatment of complicated intra-abdominal infections, complicated UTIs, hospital-acquired pneumonia, and BSI associated with those conditions; it is also approved for treatment of infections caused by Gram negatives with limited treatment options [45]. Case reports on preterm infants [62,66,69,70] included newborns of 27–29 weeks, in whom treatment was started at 11–46 days of life and continued for 10 to 14 days in the case of UTI or BSI [66,70] and for 21 to 47 days in the case of meningitidis [62,69]. Among 11 patients treated with ceftazidime/avibactam 10 achieved microbiological and clinical cure, while one died. In 36 infants of 3 months–2 years of age with complicated UTIs, a clinical cure was observed in 99% of patients randomized to ceftazidime/avibactam vs. 90% of patients randomized to comparator cefepime [60]. Similar efficacy of ceftazidime/avibactam and meropenem was observed in 83 patients of 3 months–18 years of age with complicated intra-abdominal infections [61], indicating high efficacy of ceftazidime/avibactam in infants and children. Similarly, in adults, ceftazidime/avibactam showed higher efficacy in the treatment of infections caused by CRE in comparison with different combinations of colistin, tigecycline, fosfomycin, and carbapenems [43]. A recent meta-analysis [107] demonstrated that ceftazidime/avibactam was more effective than comparators to achieve clinical cure of infections caused by carbapenem-resistant K. pneumoniae, and patients treated with ceftazidime/avibactam also presented lower mortality rates at 28–30 days. Ceftazidime/avibactam was recently included in the treatment algorithm for both carbapenemase-positive and negative CRE infections in children, without restrictions of age [43]. It was also suggested to strongly consider the use of beta-lactams/beta-lactamase inhibitors, as ceftazidime/avibactam, imipenem/relebactam and meropenem/vaborbactam, for susceptible CRE isolates with MIC for meropenem ≥4 microg/mL or known to produce KPC based on rapid molecular diagnostic tests [43]. Meropenem/vaborbactam is at present approved for adult use in complicated UTIs, intra-abdominal infections, hospital-acquired pneumonia, or BSI associated with the previous conditions, and in general for infections by Gram negatives with limited treatment options [46]; similarly, imipenem/relebactam is approved for adult use [47]. At present no data are available for newborns.
Ceftolozane/tazobactam is a cephalosporin/beta-lactamase inhibitor with enhanced activity against P. aeruginosa, approved for the treatment of complicated intra-abdominal infection, and complicated UTIs including pyelonephritis with no restrictions of age, provided the patients are ≥ 7 days and ≥ 32 weeks of gestation, and for the treatment of hospital-acquired pneumonia in adults [48]. Among 95 patients with complicated UTIs, including 20 patients with age birth-3 months randomized to ceftolozane/tazobactam or meropenem, a clinical cure was observed in 94% vs. 100%, respectively [82], suggesting that ceftolozane/tazobactam is an effective treatment option in newborns, infants, and children. In patients >2 years with complicated intra-abdominal infections ceftolozane/tazobactam in combination with metronidazole was effective as meropenem and well-tolerated [83]. One case series [79] reported 3 infants with age 3–10 months with comorbidities treated with ceftolozane/tazobactam for pneumonia, 2 of whom were clinically cured. These results are consistent with data from adult patients, indicating the high efficacy of ceftolozane/tazobactam in patients with MDR Gram negatives infections, such as lower and upper UTIs [108] and intra-abdominal infections [109]. A recent meta-analysis [110] showed that ceftolozane/tazobactam was more effective in achieving clinical cure or microbiological eradication in comparison to polymyxin/aminoglycoside and quinolones in adults with Gram negatives infections, including MDR P. aeruginosa.
Cefiderocol is a siderophore cephalosporin with excellent activity against carbapenemases and it is currently approved for the treatment of Gram negatives with limited treatment options in adults [49]. Two case reports documented the successful treatment of preterm newborns of 27–31 weeks of gestation with LOS by VIM metallo-beta-lactamase producing K. pneumoniae [89] and LOS and necrotizing enterocolitis caused by KPC-producing K. pneumoniae [88] after the failure of netilmicin [89] and meropenem and colistin [88]. Treatment with cefiderocol was started at 9 [88] and 20 days of life [89] and continued for 14 and 9 days, respectively. In adults, cefiderocol showed similar efficacy to other available best comparators in the case of infections by MDR Gram negatives [111] and showed non-inferiority in comparison to meropenem for the treatment of hospital-acquired pneumonia [112]. Cefiderocol also showed superiority to the best available therapy and high-dose meropenem for the outcomes of clinical cure, microbiological eradication, and mortality at 28 days in the case of infections caused by metallo-beta-lactamase-producing Gram negatives [113].
Noticeably, all antimicrobial targeting MDR Gram negatives considered in this review were recently included in the guidelines of the Infectious Disease Society of America applying both to adult and pediatric patients, with no further age cohort specification [114], with an indication that all of them may be considered for the treatment of CRE, and, except for meropenem/vaborbactam, for the treatment of P. aeuruginosa with “difficult-to-treat resistance”. Cefiderocol is also recommended for the treatment of carbapenem-resistant A. baumannii [114].
Ceftaroline is a 5th generation cephalosporin with activity against Gram positives including MRSA and MDR S. pneumoniae, and it is currently approved for the treatment of patients of any age, including newborns, with SSSIs or community-acquired pneumonia [50]. In 11 infants of 7–60 days with LOS treated with ceftaroline plus ampicillin and optional aminoglycoside, no treatment failure was observed [98]. In patients ≥2 months with complicated community-acquired pneumonia, ceftaroline showed similar efficacy in comparison to vancomycin plus ceftriaxone, with clinical cure observed in 83% vs. 78% of cases, respectively [93]. Ceftaroline also showed similar efficacy in comparison to ceftriaxone for the treatment of community-acquired pneumonia, with clinical cure observed in 92% vs. 89% of cases, respectively [92]. Likewise, ceftaroline was proved highly effective for the treatment of adult pneumonia, and a recent meta-analysis found a higher probability of clinical cure with ceftaroline in comparison to ceftriaxone [115]. Moreover, ceftaroline recently showed non-inferiority in comparison to daptomycin for the treatment of BSIs caused by MRSA without pulmonary origin [116]. For the treatment of 159 patients ≥2 months of age with SSSIs ceftaroline achieved clinical cure in 96% vs. 88% of comparators, vancomycin or cefazolin [94], in accordance with data from adult patients [117,118,119]. In 2 case reports, ceftaroline was effective in preterm newborns of 24–30 weeks of gestation with BSI by MRSA, one with LOS and pneumonia [96], and one with hepatic abscess and infected thrombus of the portal system [100]. In one case [96], treatment with ceftarolin was started at 43 days of life, after failure of oxacillin, vancomycin, and rifampin, and administered for 21 days [96], while in the other case, treatment was started at 54 days of life, after failure of vancomycin, daptomycin, and linezolid, and continued for 18 days [100]. Successful pharmacokinetic target attainment was also reported with the administration of 8.5 mg/kg q8h [96].
Dalbavancin is a long-acting semisynthetic lipoglycopeptide antibiotic with bactericidal activity against Gram-positive pathogens, including S. aureus including MRSA, S. pneumoniae, S. agalactiae, S. pyogenes, and Enterococcus spp., and it is currently approved for the treatment of SSSIs in patients >3 months [51]. Dalbavancin was administered for the treatment of BSIs in patients from birth to 3 months and SSSIs from birth to 18 years known or suspected to be caused by susceptible Gram positives [106], mainly MSSA; 5 patients in the cohort birth-3 months, including 3 patients with age < 1 month, were treated with single dose dalbavancin, with excellent overall efficacy.
At present, the susceptibility of pediatric and neonatal isolates to novel antimicrobials is excellent in HICs. In studies on pediatric and neonatal isolates, Enterobacterales showed excellent susceptibility of 97–100% to ceftazidime/avibactam [120,121], while P. aeruginosa maintained high susceptibility of 96–100% in the general pediatric population including newborns [121] but showed poor susceptibility of 47% in colonized pediatric patients with cystic fibrosis [122]. At present overall susceptibility of pediatric Gram negatives isolates to ceftolozane/tazobactam is excellent in HICs [123,124,125]; however, slightly lower susceptibility has been shown for K. pneumoniae [125] and resistance to ceftolozane/tazobactam was reported in approximately half of the cases for pediatric cystic fibrosis patients colonized with MDR P. aeruginosa [122]. In 1460 isolates from pediatric respiratory tract infections and SSSIs, including 263 isolates obtained from patients <1 year, susceptibility to ceftaroline was 100% for H. influenzae, S. aureus, and E. coli, 99.6% for S. pneumoniae, and 97% for Klebsiella spp. [126].
No significant safety issues in newborns and infants emerged from the included studies. For the treatment of Gram negatives, the use of ceftazidime/avibactam was not associated with any drug-related AEs, in accordance with the re-assuring safety profile observed in adults [43,107]. In preterm newborns 2 AEs were reported to have an uncertain association with the drug, one case of thrombocytopenia not requiring transfusion [62], and one case of transient glycosuria [66]. Drug-related adverse events were reported for ceftolozane/tazobactam by 2 RCTs and included diarrhea, increased transaminases, and dermatitis or rush, similarly to AEs displayed by adult patients [82,83]. No AEs were reported for preterm newborns. Cefiderocol and meropenem/vaborbactam we not associated with any drug-related AEs. Mild drug-related AEs were reported in 10–23% of patients treated with ceftaroline, including diarrhea, vomiting, dermatitis or rush, and increased transaminases. Two severe ceftaroline-related AEs, one hypersensitivity event and one case of colitis by C. difficilis were observed, both beyond the neonatal period [94], indicating similar safety profile to adult patients [92,93].
This review has some limitations. First, a modest amount of data results from RCTs or clinical studies properly designed to assess the efficacy and safety of new antimicrobials in infants and newborns, while most of the included studies were case reports or case series. Second, included studies presented variable study designs, not allowing direct comparison of the results. Third, most RCTs enrolled infants ≥2 or 3 months of age, thus slightly beyond the neonatal period. However, newborns and particularly preterm newborns may significantly differ from slightly older patients in terms of pharmacokinetic variables [127]. Newborns, particularly if preterm, show a higher volume of distribution of antimicrobial drugs, but lower renal drug clearance, resulting in higher loading dose but lower maintenance dose of the drug [127], with poorly predictable effects on efficacy and toxicity [127]. Finally, most data on the use of novel antimicrobials in newborns were obtained from studies performed in HICs. However, patients who may benefit from new antimicrobials may partially differ across countries, as a particularly high rate of MDR organisms were found in EOS in term or mild preterm newborns in LMICs, while infections with MDR organisms in HICs usually occur in very preterm newborns with healthcare-associated LOS.
Overall, available data indicate that novel antimicrobials against MDR Gram-positive and Gram-negative organisms are effective and safe in the pediatric and neonatal population, and therefore, they can be considered a useful treatment in case of infections caused by MDR organisms in NICU, when other treatment options are limited or absent. However, data on the use of these antimicrobials are still limited for children and newborns; therefore, properly designed RCTs in these populations are warranted, including newborns and preterm newborns, to specifically assess efficacy and safety in these age groups. Finally, limited or no availability of novel antimicrobials in LMICs might represent a significant issue, as those areas would likely mostly benefit from novel agents in consideration of the high rates of MDR Gram negatives reported.

4. Materials and Methods

4.1. Search Strategy

This systematic review was carried out according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines (Figure 1). A literature search was conducted on 25 March 2023, using the following databases: PubMed Medline, EMBASE, and Web of Science.
The search strategy included the following terms and was performed for each considered antibiotic (ceftazidime/avibactam, ceftolozane/tazobactam, cefiderocol, meropenem/vaborbactam, imipenem/relebactam, ceftaroline, dalbavancin): “antibiotic of interest” AND “neonate” OR “newborn” OR “neonat” OR “infant” OR “child” OR “pediatric”. No date restriction was applied. The literature search was limited to the English language. Articles were checked for duplication.

4.2. Eligibility

Two reviewers (C.P., C.D.) independently assessed eligibility. Titles and abstracts of all retrieved articles were screened to identify potentially eligible studies, and all selected articles were analyzed in full text for conclusive evaluation. Eligibility criteria for the present study were as follows: (1) studies investigating the use of new antimicrobials of interest (2) in newborns, infants, and children (3) with BSIs or infection of any site, or (4) pharmacokinetics studies in the same population. Based on the extremely recent introduction of novel antimicrobials in newborns and pediatric age in general, we decided to consider all studies on patients younger than 18 years, to collect all available data for the developmental age. The eligible study design included RCTs, retrospective studies, case reports/series, and pharmacokinetic studies. Reviews, commentaries, or meta-analyses were considered not eligible. Considered outcomes were clinical cure, microbiological eradication, safety issues, and pharmacokinetic target attainment or dose validation, depending on different types of considered studies.

5. Conclusions

In conclusion, robust evidence on the efficacy and effectiveness of novel antimicrobials for the treatment of MDR Gram positives and Gram negatives is lacking. However, all available data suggest high effectiveness and favorable safety profile of the considered novel antimicrobials in the neonatal population, including preterm newborns. Therefore, these drugs might be regarded as useful treatments in newborns and infants with infections caused by MDR organisms with limited treatment options. Further studies are warranted to specifically address indications and safety profiles in infants and newborns.

Author Contributions

Conceptualization, C.P. and C.D.; methodology, C.P.; validation, C.P. and C.D.; formal analysis, C.P.; data curation, C.P.; writing—original draft preparation, C.P.; writing—review and editing, C.P. and C.D.; visualization, C.P.; supervision, C.P. and C.D.; project administration, C.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Laxminarayan, R.; Matsoso, P.; Pant, S.; Brower, C.; Røttingen, J.A.; Klugman, K.; Davies, S. Access to effective antimicrobials: A worldwide challenge. Lancet 2016, 387, 168–175. [Google Scholar] [CrossRef] [PubMed]
  2. Fleischmann, C.; Reichert, F.; Cassini, A.; Horner, R.; Harder, T.; Markwart, R.; Tröndle, M.; Savova, Y.; Kissoon, N.; Schlattmann, P.; et al. Global incidence and mortality of neonatal sepsis: A systematic review and meta-analysis. Arch. Dis. Child. 2021, 106, 745–752. [Google Scholar] [CrossRef] [PubMed]
  3. GBD 2017 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: A systematic analysis for the global burden of disease study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef] [PubMed]
  4. GBD 2017 Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: A systematic analysis for the global burden of disease study 2017. Lancet 2018, 392, 1736–1788. [Google Scholar] [CrossRef]
  5. Li, G.; Bielicki, J.A.; Ahmed AS MN, U.; Islam, M.S.; Berezin, E.N.; Gallacci, C.B.; Guinsburg, R.; da Silva Figueiredo, C.E.; Vieira, R.S.; Silva, A.R.; et al. Towards understanding global patterns of antimicrobial use and resistance in neonatal sepsis: Insights from the NeoAMR network. Arch. Dis. Child. 2020, 105, 26–31. [Google Scholar] [CrossRef]
  6. 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]
  7. Thomson, K.M.; Dyer, C.; Liu, F.; Sands, K.; Portal, E.; Carvalho, M.J.; Barrell, M.; Boostrom, I.; Dunachie, S.; Farzana, R.; et al. Effects of antibiotic resistance, drug target attainment, bacterial pathogenicity and virulence, and antibiotic access and affordability on outcomes in neonatal sepsis: An international microbiology and drug evaluation prospective substudy (BARNARDS). Lancet Infect. Dis. 2021, 21, 1677–1688. [Google Scholar] [CrossRef]
  8. Investigators of the Delhi Neonatal Infection Study (DeNIS) Collaboration. Characterization 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]
  9. Wang, J.; Lv, Y.; Yang, W.; Zhao, P.; Yin, C. Epidemiology and clinical characteristics of infection/colonization due to carbapenemase-producing Enterobacterale in neonatal patients. BMC Microbiol. 2022, 22, 177. [Google Scholar] [CrossRef]
  10. Pathak, A.; Tejan, N.; Dubey, A.; Chauhan, R.; Fatima, N.; Jyoti Singh, S.; Bhayana, S.; Sahu, C. Outbrteak of colistin resistant, carbapenemase (blaNDM, blaOXA-232) producing Klebsiella pneumoniae causing blood stream infection among neonates at a tertiary care hospital in India. Front. Cell. Infect. Microbiol. 2023, 13, 1051020. [Google Scholar] [CrossRef]
  11. Alanezi, G.; Almulhem, A.; Aldriwesh, M.; Bawazeer, M. A triple antimicrobial regimen for multi-drug resistant Klebsiella pneumoniae in a neonatal intensive care unit outbreak: A case series. J. Infect. Public Health 2022, 15, 138–141. [Google Scholar] [CrossRef] [PubMed]
  12. Manandhar, S.; Nguyen, Q.; Pham, D.; Amatya, P.; Rabaa, M.; Dongol, S.; Basnyat, B.; Dixit, S.; Baker, S.; Karkey, A. A fatal outbreak of neonatal sepsis caused by mcr-10-carrying Enterobacter kobei in a tertiary care hospital in Nepal. J. Hosp. Infect. 2022, 125, 60–66. [Google Scholar] [CrossRef] [PubMed]
  13. Labi, A.-K.M.; Enweronu-Laryea, C.C.M.; Nartey, E.T.; Bjerrum, S.M.; Ayibor, P.K.M.; Andersen, L.P.; Newman, M.J.M.; Kurtzhals, J.A.L.M. Bloodstream Infections at Two Neonatal Intensive Care Units in Ghana: Multidrug Resistant Enterobacterales Undermine the Usefulness of Standard Antibiotic Regimes. Pediatr. Infect. Dis. J. 2021, 40, 1115–1121. [Google Scholar] [CrossRef]
  14. Agosta, M.; Bencardino, D.; Argentieri, M.; Pansani, L.; Sisto, A.; Degli Atti, M.L.C.; D’amore, C.; Bagolan, P.; Iacobelli, B.D.; Magnani, M.; et al. Clonal Spread of Hospital-Acquired NDM-1-Producing Klebsiella pneumoniae and Escherichia coli in an Italian Neonatal Surgery Unit: A Retrospective Study. Antibiotics 2023, 12, 642. [Google Scholar] [CrossRef] [PubMed]
  15. Berglund, B.; Hoang, N.T.B.; Lundberg, L.; Le, N.K.; Tärnberg, M.; Nilsson, M.; Bornefall, E.; Khu, D.T.K.; Welander, J.; Le, H.T.; et al. Clonal spread of carbapenem-resistant Klebsiella pneumoniae among patients at admission and discharge at a Vietnamese neonatal intensive care unit. Antimicrob. Resist. Infect. Control 2021, 10, 162. [Google Scholar] [CrossRef]
  16. Villinger, D.; Schultze, T.G.; Musyoki, V.M.; Inwani, I.; Aluvaala, J.; Okutoyi, L.; Ziegler, A.-H.; Wieters, I.; Stephan, C.; Museve, B.; et al. Genomic transmission analysis of multidrug-resistant Gram-negative bacteria within a newborn unit of a Kenyan tertiary hospital: A four-month prospective colonization study. Front. Cell. Infect. Microbiol. 2022, 12, 892126. [Google Scholar] [CrossRef]
  17. Agosta, M.; Bencardino, D.; Argentieri, M.; Pansani, L.; Sisto, A.; Degli Atti, M.L.C.; D’amore, C.; Putignani, L.; Bagolan, P.; Iacobelli, B.D.; et al. Prevalence and Molecular Typing of Carbapenemase-Producing Enterobacterales among Newborn Patients in Italy. Antibiotics 2022, 11, 431. [Google Scholar] [CrossRef]
  18. Carvalho, M.J.; Sands, K.; Thomson, K.; Portal, E.; Mathias, J.; Milton, R.; Gillespie, D.; Dyer, C.; Akpulu, C.; Boostrom, I.; et al. Antibiotic resistance genes in the gut microbiota of mothers and linked neonates with or without sepsis from low- and middle-income countries. Nat. Microbiol. 2022, 7, 1337–1347. [Google Scholar] [CrossRef]
  19. Okomo, U.; Akpalu, E.N.K.; Le Doare, K.; Roca, A.; Cousens, S.; Jarde, A.; Sharland, M.; Kampmann, B.; E Lawn, J. Aetiology of invasive bacterial infection and antimicrobial resistance in neonates in sub-Saharan Africa: A systematic review and meta-analysis in line with the STROBE-NI reporting guidelines. Lancet Infect. Dis. 2019, 3099, 1219–1234. [Google Scholar] [CrossRef]
  20. Gu, S.; Lai, J.; Kang, W.; Li, Y.; Zhu, X.; Ji, T.; Feng, J.; Zhao, L.; Li, Z.; Dong, L.; et al. Drug resistance characteristics and molecular typing of Escherichia coli isolates from neonates in class A tertiary hospitals: A multicentre study across China. J. Infect. 2022, 85, 499–506. [Google Scholar] [CrossRef]
  21. Ren, Z.; Yang, S.; Han, J.; Nie, C.; Wang, C.; Wang, J.; Zheng, X.; Yang, H.; Zhang, Q.; Pei, J.; et al. Reduction of antibiotic use and multi-drug resistance bacteria infection in neonates after improvement of antibiotics use strategy in a level 4 neonatal intensive care unit in southern China. Eur. J. Clin. Microbiol. Infect. Dis. 2023, 42, 87–98. [Google Scholar] [CrossRef] [PubMed]
  22. Li, J.-Y.; Chen, S.-Q.; Yan, Y.-Y.; Hu, Y.-Y.; Wei, J.; Wu, Q.-P.; Lin, Z.-L.; Lin, J. Identification and antimicrobial resistance of pathogens in neonatal septicemia in China—A meta-analysis. Int. J. Infect. Dis. 2018, 71, 89–93. [Google Scholar] [CrossRef] [PubMed]
  23. Stoll, B.J.; Puopolo, K.M.; Hansen, N.I.; Sánchez, P.J.; Bell, E.F.; Carlo, W.A.; Cotten, C.M.; D’Angio, C.T.; Kazzi, S.N.J.; Poindexter, B.B.; et al. Early-Onset Neonatal Sepsis 2015 to 2017, the Rise of Escherichia coli, and the Need for Novel Prevention Strategies. JAMA Pediatr. 2020, 174, e200593. [Google Scholar] [CrossRef] [PubMed]
  24. Flannery, D.D.; Chiotos, K.; Gerber, J.S.; Puopolo, K.M. Neonatal multidrug-resistant gram-negative infection: Epidemiology, mechanisms of resistance, and management. Pediatr. Res. 2022, 91, 380–391. [Google Scholar] [CrossRef]
  25. Flannery, D.D.; Akinboyo, I.C.; Mukhopadhyay, S.; Tribble, A.C.; Song, L.; Chen, F.; Li, Y.; Gerber, J.S.; Puopolo, K.M. Antibiotic Susceptibility of Escherichia coli among Infants Admitted to Neonatal Intensive Care Units across the US from 2009 to 2017. JAMA Pediatr. 2021, 175, 168–175. [Google Scholar] [CrossRef]
  26. Patel, S.J.; Green, N.; Clock, S.A.; Paul, D.A.; Perlman, J.M.; Zaoutis, T.; Ferng, Y.-H.; Alba, L.; Jia, H.; Larson, E.L.; et al. Gram-Negative Bacilli in Infants Hospitalized in the Neonatal Intensive Care Unit. J. Pediatr. Infect. Dis. Soc. 2017, 6, 227–230. [Google Scholar] [CrossRef]
  27. Daniels, K.; Arrieta, A.; Nieves, D.J.; Bhakta, K.; Tran, M.T.; Osborne, S.; Morphew, T. Ampicillin and Gentamicin Treatment for Early Onset Neonatal Sepsis: When One Size Does Not Fit All. Clin. Pediatr. 2023, 24, 99228221150612. [Google Scholar] [CrossRef]
  28. Cailes, B.; Kortsalioudaki, C.; Buttery, J.; Pattnayak, S.; Greenough, A.; Matthes, J.; Russell, A.B.; Kennea, N.; Heath, P. Antimicrobial resistance in UK neonatal units: neonIN infection surveillance network. Arch. Dis. Child. Fetal Neonatal Ed. 2018, 103, F474–F478. [Google Scholar] [CrossRef]
  29. Gkentzi, D.; Kortsalioudaki, C.; Cailes, B.C.; Zaoutis, T.; Kopsidas, J.; Tsolia, M.; Spyridis, N.; Siahanidou, S.; Sarafidis, K.; Heath, P.T.; et al. Epidemiology of infections and antimicrobial use in Greek Neonatal Units. Arch. Dis. Child. Fetal. Neonatal Ed. 2019, 104, F293–F297. [Google Scholar] [CrossRef]
  30. Köstlin-Gille, N.; Härtel, C.; Haug, C.; Göpel, W.; Zemlin, M.; Müller, A.; Poets, C.F.; Herting, E.; Gille, C. Epidemiology of Early and Late Onset Neonatal Sepsis in Very Low Birthweight Infants: Data from the German Neonatal Network. Pediatr. Infect. Dis. J. 2021, 40, 255–259. [Google Scholar] [CrossRef]
  31. Orman, A.; Celik, Y.; Evik, G.; Ersöz, G.; Kuyucu, N.; Ozmen, B.O. Should Perirectal Swab Culture Be Performed in Cases Admitted to the Neonatal Intensive Care Unit? Lessons Learned from the Neonatal Intensive Care Unit. Children 2023, 10, 187. [Google Scholar] [CrossRef] [PubMed]
  32. Odoyo, E.; Matano, D.; Tiria, F.; Georges, M.; Kyanya, C.; Wahome, S.; Mutai, W.; Musila, L. Environmental contamination across multiple hospital departments with multidrug-resistant bacteria pose an elevated risk of healthcare-associated infections in Kenyan hospitals. Antimicrob. Resist. Infect. Control 2023, 12, 22. [Google Scholar] [CrossRef] [PubMed]
  33. Van der Hoeven, A.; Bekker, V.; Jansen, S.J.; Saccoccia, B.; Berkhout, R.J.; Lopriore, E.; Veldkamp, K.E.; van der Beek, M.T. Impact of transition from open bay to single room design neonatal intensive care unit on multidrug-resistant organism colonization rates. J. Hosp. Infect. 2022, 120, 90–97. [Google Scholar] [CrossRef]
  34. Saporito, L.; Graziano, G.; Mescolo, F.; Amodio, E.; Insinga, V.; Rinaudo, G.; Aleo, A.; Bonura, C.; Vitaliti, M.; Corsello, G.; et al. Efficacy of a coordinated strategy for containment of multidrug-resistant Gram-negative bacteria carriage in a Neonatal Intensive Care Unit in the context of an active surveillance program. Antimicrob. Resist. Infect. Control 2021, 10, 30. [Google Scholar] [CrossRef] [PubMed]
  35. Litz, J.E.; Goedicke-Fritz, S.; Härtel, C.; Zemlin, M.; Simon, A. Management of early- and late-onset sepsis: Results from a survey in 80 German NICUs. Infection 2019, 47, 557–564. [Google Scholar] [CrossRef] [PubMed]
  36. Matrat, L.; Plaisant, F.; Barreto, C.; Claris, O.; Butin, M. Increasing use of linezolid in a tertiary NICU during a 10-year period: Reasons and concerns for the future. Antimicrob. Resist. Infect. Control 2020, 9, 156. [Google Scholar] [CrossRef]
  37. Marom, R.; Mandel, D.; Haham, A.; Berger, I.; Ovental, A.; Raskind, C.; Grisaru-Soen, G.; Adler, A.; Lellouche, J.; Schwartz, D.; et al. A silent outbreak of vancomycin-resistant Enterococcus faecium in a neonatal intensive care unit. Antimicrob. Resist. Infect. Control 2020, 9, 87. [Google Scholar] [CrossRef]
  38. 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. Paediatr. Drugs 2021, 23, 465–484. [Google Scholar] [CrossRef]
  39. Williams, P.C.; Qazi, S.A.; Agarwal, R.; Velaphi, S.; Bielicki, J.A.; Nambiar, S.; Giaquinto, C.; Bradley, J.; Noel, G.J.; Ellis, S.; et al. Antibiotics needed to treat multidrug-resistant infections in neonates. Bull. World Health Organ. 2022, 100, 797–807. [Google Scholar] [CrossRef]
  40. Tzialla, C.; Borghesi, A.; Pozzi, M.; Stronati, M. Neonatal infections due to multi-resistant strains: Epidemiology, current treatment, emerging therapeutic approaches and prevention. Clin. Chim. Acta 2015, 451 Pt A, 71–77. [Google Scholar] [CrossRef]
  41. Chiusaroli, L.; Liberati, C.; Vaseti, M.; Rulli, L.; Barbieri, E.; Giacquinto, 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. [Google Scholar] [CrossRef] [PubMed]
  42. Thomas, R.; Velaphi, S.; Ellis, S.; Walker, A.S.; Standing, J.F.; Heath, P.; Sharland, M.; Donà, D. The use of polymyxins to treat carbapenem resistant infections in neonates and children. Expert Opin. Pharmacother. 2019, 20, 415–422. [Google Scholar] [CrossRef] [PubMed]
  43. Chiotos, K.; Hayes, M.; Gerber, J.S.; Tamma, P.D. Treatment of Carbapenem-Resistant Enterobacteriaceae Infections in Children. J. Pediatr. Infect. Dis. Soc. 2020, 9, 56–66. [Google Scholar] [CrossRef] [PubMed]
  44. Chiusaroli, L.; Liberati, C.; Rulli, L.; Barbieri, E.; De Pieri, M.; Di Chiara, C.; Mengato, D.; Giacquinto, 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. [Google Scholar] [CrossRef]
  45. Zavicefta. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zavicefta (accessed on 25 March 2023).
  46. Vaborem. Available online: https://www.ema.europa.eu/en/documents/product-information/vaborem-epar-product-information_it.pdf (accessed on 25 March 2023).
  47. Recarbrio. Available online: https://www.ema.europa.eu/en/documents/product-information/recarbrio-epar-product-information_it.pdf (accessed on 25 March 2023).
  48. Zerbaxa. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zerbaxa (accessed on 25 March 2023).
  49. Fetcroja. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/fetcroja (accessed on 25 March 2023).
  50. Zinforo. Available online: https://www.ema.europa.eu/en/documents/product-information/zinforo-epar-product-information_en.pdf (accessed on 25 March 2023).
  51. Xydalba. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/xydalba (accessed on 25 March 2023).
  52. Tenkasi (previously Orbactiv). Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/tenkasi-previously-orbactiv (accessed on 18 May 2023).
  53. VIBATIV (telavancin) for Injection. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/022407_vibativ_toc.cfm (accessed on 18 May 2023).
  54. Sivextro. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/sivextro (accessed on 18 May 2023).
  55. Zeftera. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zeftera-previously-zevtera (accessed on 18 May 2023).
  56. Xerava. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/xerava (accessed on 18 May 2023).
  57. Bradley, J.S.; Armstrong, J.; Arrieta, A.; Bishai, R.; Das, S.; Delair, S.; Edeki, T.; Holmes, W.C.; Li, J.; Moffett, K.S.; et al. Phase I Study Assessing the Pharmacokinetic Profile, Safety, and Tolerability of a Single Dose of Ceftazidime-Avibactam in Hospitalized Pediatric Patients. Antimicrob. Agents Chemother. 2016, 60, 6252–6259. [Google Scholar] [CrossRef]
  58. Tamma, P.D.; Fan, Y.; Bergman, Y.; Sick-Samuel, A.C.; Hsu, A.J.; Timp, W.; Simner, P.J.; Prokesch, B.C.; Greenberg, D.E. Successful Treatment of Persistent Burkholderia cepacia Complex Bacteremia with Ceftazidime-Avibactam. Antimicrob. Agents Chemother. 2018, 62, e02213-17. [Google Scholar] [CrossRef]
  59. 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 <5 Years of Age. Pediatr. Infect. Dis. J. 2019, 38, 812–815. [Google Scholar] [CrossRef] [PubMed]
  60. Bradley, J.S.; Roilides, E.; Broadhurst, H.; Cheng, K.; Huang, L.; 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]
  61. 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]
  62. Esposito, P.; Sbrana, F.; Di Toro, A.; Gombos, S.; Tascini, C. Ceftazidine-avibactam salvage therapy in newborn with KPC-producing Klebsiella pneumoniae invasive infections. Minerva Anestesiol. 2019, 85, 804–805. [Google Scholar] [CrossRef]
  63. Vargas, M.; Buonomo, A.R.; Buonanno, P.; Iacovazzo, C.; Servillo, G. Successful treatment of KPC-MDR septic shock with ceftazidime-avibactam in a pediatric critically ill patient. IDCases 2019, 18, e00634. [Google Scholar] [CrossRef]
  64. Nguyen, T.T.; Condren, M.; Walter, J. Ceftazidime-avibactam for the treatment of multidrug resistant Burkholderia cepacia complex in a pediatric cystic fibrosis patient. Pediatr. Pulmonol. 2020, 55, 283–284. [Google Scholar] [CrossRef] [PubMed]
  65. Hobson, C.A.; Bonacorsi, S.; Fahd, M.; Baruchel, A.; Cointe, A.; Poey, N.; Jacquier, H.; Doit, C.; Monjault, A.; Tenaillon, O.; et al. Successful Treatment of Bacteremia Due to NDM-1-Producing Morganella morganii with Aztreonam and Ceftazidime-Avibactam Combination in a Pediatric Patient with Hematologic Malignancy. Antimicrob. Agents Chemother. 2019, 63, e02463-18. [Google Scholar] [CrossRef] [PubMed]
  66. Coskun, I.; 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]
  67. Ji, Z.; Sun, K.; Li, Z.; Cheng, W.; Yang, J. Carbapenem-Resistant Klebsiella pneumoniae Osteomyelitis Treated with Ceftazidime-Avibactam in an Infant: A Case Report. Infect. Drug Resist. 2021, 14, 3109–3113. [Google Scholar] [CrossRef]
  68. Franzese, R.C.; McFadyen, L.; Watson, K.J.; Ricobene, T.; Carrothers, T.J.; Vourvahis, M.; Chan, P.L.S.; Raber, S.; Bradley, J.S.; Lovern, M. Population Pharmacokinetic Modeling and Probability of Pharmacodynamic Target Attainment for Ceftazidime-Avibactam in Pediatric Patients Aged 3 Months and Older. Clin. Pharmacol. Ther. 2022, 111, 635–645. [Google Scholar] [CrossRef]
  69. Asfour, S.; Alaklobi, F.A.; Abdelrahim, A.; Taha, M.Y.; Asfour, R.A.; Khalil, T.M.; Al-Mouqdad, M.M. Intravenous Ceftazidime-Avibactam in Extremely Premature Neonates with Carbapenem-Resistant Enterobacteriaceae: Two Case Report. J. Pediatr. Pharmacol. Ther. 2022, 27, 192–197. [Google Scholar] [CrossRef]
  70. Nascimento, A.; Passaro, M.F.; de S Silva, P.S.; Rodriguez, S.; Martins, M.K.; Oliveira, S.C.; Moriel, P.; Visacri, M.B. Off-Label Use of Ceftazidime-Avibactam in a Premature Infant with Multidrug-Resistant Klebsiella pneumoniae Infection: A Case Report. J. Pharm. Pract. 2022. [Google Scholar] [CrossRef] [PubMed]
  71. Almangour, T.A.; Alsubaie, S.; Ghonem, L.; Almohaini, H.A.; Bakheet, H.M.; Altweijri, I. Ceftazidime-avibactam for the Treatment of Multidrug-resistant Pseudomonas aeruginosa Central Nervous System Infection in Pediatric Patient: A Case Report. Pediatr. Infect. Dis. J. 2022, 41, 436–438. [Google Scholar] [CrossRef]
  72. Perruccio, K.; D’Amico, M.R.; Baretta, V.; Onofrillo, D.; carraro, F.; Calore, E.; Muggeo, P.; Colombini, A.; Zama, D.; Meazza, C.; et al. Ceftolozane/Tazobactam and Ceftazidime/Avibactam: An Italian Multi-center Retrospective Analysis of Safety and Efficacy in Children with Hematologic Malignancies and Multi-drug Resistant Gram-negative Bacteria Infections. Pediatr. Infect. Dis. J. 2022, 41, 994–996. [Google Scholar] [CrossRef]
  73. Aitken, S.L.; Kontoyiannis, D.P.; DePombo, A.M.; Bhatti, M.M.; Tverdek, F.P.; Gettys, S.C.; Nicolau, D.P.; Nunez, C.A. Use of Ceftolozane/Tazobactam in the Treatment of Multidrug-resistant Pseudomonas aeruginosa Bloodstream Infection in a Pediatric Leukemia Patient. Pediatr. Infect. Dis. J. 2016, 35, 1040–1042. [Google Scholar] [CrossRef] [PubMed]
  74. Bradley, J.S.; Ang, J.Y.; Arrieta, A.C.; Larson, K.B.; Rizk, M.L.; Caro, L.; Yang, S.; Yu, B.; Johnson, M.G.; Rhee, E.G. Pharmacokinetics and Safety of Single Intravenous Doses of Ceftolozane/Tazobactam in Children with Proven or Suspected Gram-Negative Infection. Pediatr. Infect. Dis. J. 2018, 37, 1130–1136. [Google Scholar] [CrossRef]
  75. Martin-Cazana, M.; Grau, S.; Epalza, C.; Branas, P.; Flores, M.; Olmedilla, M.; Blazquez-Gamero, D. Successful ceftolozane-tazobactam rescue therapy in a child with endocarditis caused by multidrug-resistant Pseudomonas aeruginosa. J. Pediatr. Child Health 2019, 55, 985–987. [Google Scholar] [CrossRef] [PubMed]
  76. Zikri, A.; el Masri, K. Use of Ceftolozane/tazobactam for the Treatment of Multidrug-resistant Pseudomonas aeruginosa Pneumonia in a Pediatric Patient with Combined Immunodeficiency (CID): A Case Report from a Tertiary Hospital in Saudi Arabia. Antibiotics 2019, 8, 67. [Google Scholar] [CrossRef]
  77. Ang, J.Y.; Arrieta, A.; Bradley, J.S.; Zhang, Z.; Yu, B.; Rizk, M.L.; Johnson, M.G.; Rhee, E.G. Ceftolozane/Tazobactam in Neonates and Young Infants: The Challenges of Collecting Pharmacokinetics and Safety Data in This Vulnerable Patient Population. Am. J. Perinatol. 2021, 38, 804–809. [Google Scholar] [CrossRef] [PubMed]
  78. Larson, K.B.; Patel, Y.T.; Willavize, S.; Bradley, J.S.; Rhee, E.G.; Caro, L.; Rizk, M.L. Ceftolozane-Tazobactam Population Pharmacokinetics and Dose Selection for Further Clinical Evaluation in Pediatric Patients with Complicated Urinary Tract or Complicated Intra-abdominal Infections. Antimicrob. Agents Chemother. 2019, 63, e02578-18. [Google Scholar] [CrossRef]
  79. Molloy, L.; Abdulhamid, I.; Srivastava, R.; Ang, J.Y. Ceftolozane/Tazobactam Treatment of Multidrug-resistant Pseudomonas aeruginosa Infections in Children. Pediatr. Infect. Dis. J 2020, 39, 419–420. [Google Scholar] [CrossRef]
  80. Arrieta, A.C.; Ang, J.Y.; Zhang, Z.; Larson, K.B.; Yu, B.; Johnson, M.G.; Rhee, E.G.; Feng, E.H.; Rizk, M.L. Plasma pharmacokinetics of ceftolozane/tazobactam in pediatric patients with cystic fibrosis. Pediatr. Pulmonol. 2020, 55, 2025–2032. [Google Scholar] [CrossRef]
  81. Butragueno-Laiseca Troconiz, I.F.; Grau, S.; Campillo, N.; Garcia, X.; Padilla, B.; Fernandez, S.N.; Santiago, M.J. Finding the Dose for Ceftolozane-Tazobactam in Critically Ill Children with and without Acute Kidney Injury. Antibiotics 2020, 9, 887. [Google Scholar] [CrossRef]
  82. Roilides, E.; Ashouri, N.; Bradley, J.S.; Johnson, M.G.; Lonchar, J.M.; Su, F.H.M.; Huntington, J.A.; Popejoy, M.W.; Bensaci, M.; De Anda, C.; et al. Safety and Efficacy of Ceftolozane/Tazobactam Versus Meropenem in Neonates and Children with Complicated Urinary Tract Infection, Including Pyelonephritis: A Phase 2, Randomized Clinical Trial. Pediatr. Infect. Dis. J. 2023, 42, 292–298. [Google Scholar] [CrossRef]
  83. Jackson, C.A.; Newland, J.; Demetieva, N.; Lonchar, J.; Su, F.; Huntington, J.A.; Bensaci, M.; Popejoy, M.; Johnson, M.G.; De Anda, C.; et al. Safety and Efficacy of Ceftolozane/Tazobactam Plus Metronidazole Versus Meropenem from a Phase 2, Randomized Clinical Trial in Pediatric Participants with Complicated Intra-abdominal Infection. Pediatr. Infect. Dis. J. 2021, 8, 668–669. [Google Scholar] [CrossRef]
  84. Katsube, T.; Echols, R.; Wajima, T. Prediction of Cefiderocol Pharmacokinetics and Probability of Target Attainment in Pediatric Subjects for Proposing Dose Regimen. Open Forum Infect. Dis. 2019, 6 (Suppl. S2), S330–S331. [Google Scholar]
  85. Alamarat, Z.; Babic, J.; Tran, T.T.; Wootton, S.H.; Dinh, A.Q.; Miller, W.R.; Hanson, B.; Wanger, A.; Gary, J.L.; Arias, C.A.; et al. Long-Term Compassionate Use of Cefiderocol to Treat Chronic Osteomyelitis Caused by Extensively Drug-Resistant Pseudomonas aeruginosa and Extended-Spectrum-β-Lactamase-Producing Klebsiella pneumoniae in a Pediatric Patient. Antimicrob. Agents Chemother. 2020, 64, e01872-19. [Google Scholar] [CrossRef] [PubMed]
  86. Warner, C.; Bartelt, L.A.; Lachiewicz, A.M.; Tompkins, K.M.; Miller, M.B.; Alby, K.; Jones, M.B.; Carr, a.L.; Alexander, J.; Gainey, A.B.; et al. Cefiderocol for the Treatment of Adult and Pediatric Patients with Cystic Fibrosis and Achromobacter xylosoxidans Infections. Clin. Infect. Dis. 2021, 73, e1754–e1757. [Google Scholar] [CrossRef]
  87. Grasa, C.D.; Gomez-Gil, M.R.; Pacheco, S.S.R.; Del Rosal, T.; Moreno, F.; Gerig, N.; Fernandez, B.O.; Calvo, C.; Baquero-Artigao, F. Compassionate use of cefiderocol for VIM metallo-β-lactamase-producing Pseudomonas aeruginosa infection in a toddler with Burkitt lymphoma. J. Glob. Antimicrob. Resist. 2021, 26, 91–92. [Google Scholar] [CrossRef] [PubMed]
  88. Bawankule, S.; Nabar, N.; Joshi, P.; Singhal, T. Cefiderocol as Salvage Therapy for Carbapenem-Resistant Klebsiella pneumoniae Sepsis in an Extremely Preterm Neonate. Indian. J. Pediatr. 2023, 90, 310. [Google Scholar] [CrossRef]
  89. Monari, C.; Spagnolo, F.; Piasaturo, M.; Ascione, S.; Donnarumma, G.; Calò, F.; Caredda, E.; Montella, F.; Maietta, A.; Montaldo, P.; et al. Bloodstream Infection Due to a VIM-Metallo-β-Lactamase-Producing Klebsiella pneumoniae Treated with Cefiderocol in a Preterm Newborn. Infect. Dis. Ther. 2023, 12, 727–734. [Google Scholar] [CrossRef]
  90. 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]
  91. Gainey, A.B.; Burch, A.K.; Brownstein, M.J.; Brown, D.E.; Fackler, J.; Horne, B.; Biswas, B.; Bivens, B.N.; Malagon, F.; Daniels, R. Combining bacteriophages with cefiderocol and meropenem/vaborbactam to treat a pan-drug resistant Achromobacter species infection in a pediatric cystic fibrosis patient. Pediatr. Pulmonol. 2020, 55, 2990–2994. [Google Scholar] [CrossRef]
  92. Cannavino, C.R.; Nemeth, A.; Korczowski, B.; Bradley, J.S.; O’Neal, T.; Jandourek, A.; Friedland, H.D.; Kaplan, S.L. A Randomized, Prospective Study of Pediatric Patients with Community-acquired Pneumonia Treated with Ceftaroline Versus Ceftriaxone. Pediatr. Infect. Dis. J. 2016, 35, 752–759. [Google Scholar] [CrossRef] [PubMed]
  93. Blumer, J.; Ghonghadze, T.; Cannavino, C.; O’Neal, T.; Jandourek, A.; Friedland, H.D.; Bradley, J.S. A Multicenter, Randomized, Observer-blinded, Active-controlled Study Evaluating the Safety and Effectiveness of Ceftaroline Compared with Ceftriaxone Plus Vancomycin in Pediatric Patients with Complicated Community-acquired Bacterial Pneumonia. Pediatr. Infect. Dis. J. 2016, 35, 760–766. [Google Scholar] [CrossRef] [PubMed]
  94. Korczowski, B.; Antadze, T.; Giorgobani, 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]
  95. Riccobene, T.A.; Khariton, T.; Knebel, W.; Das, S.; Li, J.; Jandourek, A.; carrothers, T.J.; Bradley, J.S. Population PK Modeling and Target Attainment Simulations to Support Dosing of Ceftaroline Fosamil in Pediatric Patients with Acute Bacterial Skin and Skin Structure Infections and Community-Acquired Bacterial Pneumonia. J. Clin. Pharmacol. 2017, 57, 345–355. [Google Scholar] [CrossRef] [PubMed]
  96. Salerno, S.N.; Bernhardt, J.; laughon, M.; Jhaveri, R.; Massaro, M.; Gonzales, 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]
  97. Branstetter, J.; Searcy, H.; benner, K.; Yarbroug, 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] [PubMed]
  98. Bradley, J.S.; Stone, G.G.; Chan, P.L.S.; Raber, S.R.; Riccobene, T.; Mas Casullo, V.; Yan, J.L.; Hendrick, V.M.; Hammond, J.; Leister-Tebbe, H. 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]
  99. Ferguson, C.L.; Cowart, M.C.; Jordan-Villegas, A.; Laham, F. High-level ceftaroline resistance in a paediatric patient with invasive methicillin-resistant Staphylococcus aureus infection without previous ceftaroline exposure. J. Clin. Pharm. Ther. 2021, 46, 524–527. [Google Scholar] [CrossRef] [PubMed]
  100. Heger, M.L.; Al-Sayyad, B. Ceftaroline and Daptomycin Combination Antibiotic Therapy for a Methicillin-Resistant Staphylococcus Aureus Liver Abscess in a Premature Infant. J. Pediatr. Pharmacol. Ther. 2022, 27, 754–759. [Google Scholar] [CrossRef]
  101. Chan, P.L.S.; McFadyen, L.; Quaye, A.; Leister-Tebbe, H.; Hendrick, V.M.; Hammond, J.; Raber, S. The use of extrapolation based on modeling and simulation to support high-dose regimens of ceftaroline fosamil in pediatric patients with complicated skin and soft-tissue infections. CPT Pharmacomet. Syst. Pharmacol. 2021, 10, 551–563. [Google Scholar] [CrossRef]
  102. Riccobene, T.A.; Carrothers, T.J.; Knebel, W.; Raber, s.; Chan, P.L.S. Pharmacokinetic and Pharmacodynamic Target Attainment in Adult and Pediatric Patients Following Administration of Ceftaroline Fosamil as a 5-Minute Infusion. Clin. Pharmacol. Drug Dev. 2021, 10, 420–427. [Google Scholar] [CrossRef]
  103. Bradley, J.S.; Puttagunta, S.; Rubino, C.M.; Blumer, J.L.; Dunne, M.; Sullivan, J.E. Pharmacokinetics, Safety and Tolerability of Single Dose Dalbavancin in Children 12–17 Years of Age. Pediatr. Infect. Dis. J. 2015, 34, 748–752. [Google Scholar] [CrossRef]
  104. Gonzalez, D.; Bradley, J.S.; Blumer, J.; Yogev, R.; Watt, K.V.; James, L.P.; Palazzi, D.L.; Bhatt-Metha, V.; Sullivan, J.E.; Zhang, L.; et al. Dalbavancin Pharmacokinetics and Safety in Children 3 Months to 11 Years of Age. Pediatr. Infect. Dis. J. 2017, 36, 645–653. [Google Scholar] [CrossRef]
  105. Carrothers, T.J.; Lagraauw, H.M.; Lindbom, L.; Riccobene, T.A. Population Pharmacokinetic and Pharmacokinetic/Pharmacodynamic Target Attainment Analyses for Dalbavancin in Pediatric Patients. Pediatr. Infect. Dis. J. 2023, 42, 99–105. [Google Scholar] [CrossRef] [PubMed]
  106. Giorgobiani, M.; Burroughgs, M.H.; antadze, T.; carrothers, T.J.; Riccobene, T.A.; Patel, R.; Lin, T.; Stefanova, P. The Safety and Efficacy of Dalbavancin and Active Comparator in Pediatric Patients with Acute Bacterial Skin and Skin Structure Infections. Pediatr. Infect. Dis. J. 2023, 42, 199–205. [Google Scholar] [CrossRef]
  107. Karampatakis, T.; Tsergouli, K.; Lowrie, K. Efficacy and safety of ceftazidime-avibactam compared to other antimicrobials for the treatment of infections caused by carbapenem-resistant Klebsiella pneumoniae strains, a systematic review and meta-analysis. Microb. Pathog. 2023, 179, 106090. [Google Scholar] [CrossRef] [PubMed]
  108. Wagenlehner, F.M.; Umeh, O.; Steenbergen, J.; Yuan, G.; Darouiche, R.O. Ceftolozane tazobactam compared with levofloxacin in the treatment of complicated urinary-tract infections including pyelonephritis: A randomised, double-blind, phase 3 trial (ASPECTcUTI). Lancet 2015, 385, 1949–1956. [Google Scholar] [CrossRef] [PubMed]
  109. Solomkin, J.; Hershberger, E.; Miller, B.; Popejoy, M.; Friedland, I.; Steenbergen, J.; Yoon, M.; Collins, S.; Yuan, G.; Barie, P.S.; et al. Ceftolozane/tazobactam plus metronidazole for complicated intra-abdominal infections in an era of multidrug resistance: Results from a randomized, double-blind, phase 3 trial (ASPECT-cIAI). Clin. Infect. Dis. 2015, 60, 1462–1471. [Google Scholar] [CrossRef]
  110. Chi, Y.; Xu, J.; Bai, N.; Liang, B.; Cai, Y. The efficacy and safety of ceftolozane-tazobactam in the treatment of GNB infections: A systematic review and meta-analysis of clinical studies. Expert Rev. Anti-Infect. Ther. 2023, 21, 189–201. [Google Scholar] [CrossRef]
  111. Bassetti, M.; Echols, R.; Matsungnaga, Y.; Ariyasu, M.; Doi, Y.; Ferrer, R.; Lodise, T.P.; Naas, t.; Niki, Y.; Paterson, d.L.; et al. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant Gram-negative bacteria (CREDIBLE-CR): A randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect. Dis. 2021, 21, 226–240. [Google Scholar] [CrossRef] [PubMed]
  112. Wunderink, R.G.; Matsunaga, Y.; Ariyasu, M.; Clevenbergh, P.; Echols, R.; Kaye, K.S.; Kollef, M.; Menon, A.; Pogue, J.M.; Shorr, A.F.; et al. Cefiderocol versus high-dose, extended-infusion meropenem for the treatment of Gram-negative nosocomial pneumonia (APEKS-NP): A randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect. Dis. 2021, 21, 213–225. [Google Scholar] [CrossRef]
  113. Timsit, J.F.; Paul, M.; Shields, R.; Echols, R.; Baba, T.; Yamano, Y.; Portsmouth, S. Cefiderocol for the Treatment of Infections Due to Metallo-B-lactamase-Producing Pathogens in the CREDIBLE-CR and APEKS-NP Phase 3 Randomized Studies. Clin. Infect. Dis. 2022, 75, 1081–1084. [Google Scholar] [CrossRef] [PubMed]
  114. Tamma, P.D.; Aitken, S.L.; Bonomo, r.A.; Mathers, a.J.; van Duin, D.; Clancy, C.J. Infectious Diseases Society of America 2022 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). Clin. Infect. Dis. 2022, 75, 187–212. [Google Scholar] [CrossRef]
  115. Lan, S.H.; Chang, S.P.; Lai, C.C.; Lu, L.C.; Chao, C.M. Efficacy and Safety of Ceftaroline for the Treatment of Community-Acquired Pneumonia: A Systemic Review and Meta-Analysis of Randomized Controlled Trials. J. Clin. Med. 2019, 8, 824. [Google Scholar] [CrossRef] [PubMed]
  116. Zasowski, E.J.; Trinh, T.D.; Claeys, K.C.; Lagnf, A.M.; Bathia, s.; Klinker, K.P.; Veve, M.P.; Estrada, S.J.; Johns, S.T.; Sawyer, A.T.; et al. Multicenter cohort study of ceftaroline versus daptomycin for the treatment of methicicllin-resistant Staphylococcus aureus bloodstream infection. Open Forum Infect Dis. 2021, 9, ofab606. [Google Scholar] [CrossRef]
  117. Wilcox, M.H.; Corey, G.R.; Talbot, G.H.; Thye, D.; Friedland, D.; Baculik, t.; CANVAS 2 investigators. CANVAS 2: The second Phase III, randomized, double-blind study evaluating ceftaroline fosamil for the treatment of patients with complicated skin and skin structure infections. J. Antimicrob. Chemother. 2010, 65 (Suppl. S4), iv53–iv65. [Google Scholar] [CrossRef] [PubMed]
  118. Corrado, M.L. Integrated safety summary of CANVAS 1 and 2 trials: Phase III, randomized, double-blind studies evaluating ceftaroline fosamil for the treatment of patients with complicated skin and skin structure infections. Antimicrob. Chemother. 2010, 65 (Suppl. S4), iv67–iv71. [Google Scholar] [CrossRef]
  119. Corey, G.R.; Wilcox, M.H.; Talbot, G.H.; Thye, D.; Friedlan, D.; Baculik, T.; CANVAS 1 investigators. CANVAS 1: The first Phase III, randomized, double-blind study evaluating ceftaroline fosamil for the treatment of patients with complicated skin and skin structure infections. J. Antimicrob. Chemother. 2010, 65 (Suppl. S4), iv41–iv51. [Google Scholar] [CrossRef]
  120. Al-Sweih, N.; Jamal, W.; Mokaddas, E.; Habashy, N.; Kurdi, A.; Mohamed, N. Evaluation of the in vitro activity of ceftaroline, ceftazidime/avibactam and comparator antimicrobial agents against clinical isolates from paediatric patients in Kuwait: ATLAS data 2012-19. JAC Antimicrob. Resist. 2021, 3, dlab159. [Google Scholar] [CrossRef] [PubMed]
  121. Lin, L.Y.; Riccobene, T.; Debabov, D. Antimicrobial Activity of Ceftazidime-Avibactam Against Contemporary Pathogens from Urinary Tract Infections and Intra-abdominal Infections Collected from US Children During the 2016–2019 INFORM Surveillance Program Pediatr. Infect. Dis. J. 2021, 40, 338–343. [Google Scholar] [CrossRef]
  122. Marner, M.; Kolberg, L.; Horst, J.; Bohringer, N.; Hubner, J.; Kresna, I.D.M.; Liu, Y.; Mettal, U.; Wang, L.; Meyer-Buhn, M.; et al. Antimicrobial Activity of Ceftazidime-Avibactam, Ceftolozane-Tazobactam, Cefiderocol, and Novel Darobactin Analogs against Multidrug-Resistant Pseudomonas aeruginosa Isolates from Pediatric and Adolescent Cystic Fibrosis Patients. Microbiol. Spectr. 2023, 11, e0443722. [Google Scholar] [CrossRef] [PubMed]
  123. Shortbridge, D.; Duncan, L.; Pfaller, R.D.; Pfaller, M.A.; Flamm, R.K. Activity of ceftolozane-tazobactam and comparators when tested against Gram-negative isolates collected from paediatric patients in the USA and Europe between 2012 and 2016 as part of a global surveillance programme. Int. J. Antimicrob. Agents 2019, 53, 637–643. [Google Scholar] [CrossRef]
  124. Kanwar, N.; Harrison, C.J.; Pence, M.A.; Qin, X.; Selvarangan, R. Comparative in vitro antipseudomonal activity of ceftolozane/tazobactam against Pseudomonas aeruginosa isolates from children with cystic fibrosis. Diagn. Microbiol. Infect. Dis. 2023, 105, 115904. [Google Scholar] [CrossRef]
  125. Kanwar, N.; Banerjee, D.; Harrison, C.J.; Newland, J.G.; Qin, X.; Zerr, D.M.; Zaotis, T.; Selvarangan, R. Comparative in vitro effectiveness of ceftolozane/tazobactam against pediatric gram-negative drug-resistant isolates. J. Chemother. 2021, 33, 288–293. [Google Scholar] [CrossRef] [PubMed]
  126. Pfaller, M.A.; Mendes, R.E.; Castanheira, M.; Flamm, R.K.; Jones, R.N.; Sader, H.S. Ceftaroline Activity Tested Against Bacterial Isolates Causing Community-acquired Respiratory Tract Infections and Skin and Skin Structure Infections in Pediatric Patients from United States Hospitals: 2012–2014. Pediatr. Infect. Dis. J. 2017, 36, 486–491. [Google Scholar] [CrossRef] [PubMed]
  127. Touw, D.J.; van den Anker, J.N. Therapeutic Drug Monitoring of Antimicrobial Drugs in Neonates: An Opinion Article. Ther. Drug Monit. 2022, 44, 65–74. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA 2020 diagram for study selection.
Figure 1. PRISMA 2020 diagram for study selection.
Antibiotics 12 00956 g001
Table 1. Studies on ceftazidime/avibactam.
Table 1. Studies on ceftazidime/avibactam.
Study TypeCountryPatient CharacteristicsNOrganismsStudy InterventionOutcomes
Bradley 2016
[57]
Phase I
Pharmacokinetics
US3 month–2 years8n.p.50 mg/kg
Single 2 h infusion
Pharmacokinetics model validation
1/8 drug-related mild AEs
(sinus tachycardia)
Tamma 2018
[58]
Case reportUS2 month
BSI
1Burkholderia cepacia50 mg/kg q8h
Continuous infusion
Duration: 6 weeks
1/1 BC sterilization
1/1 clinical recovery
No drug-related AEs
Iosifidis 2019 [59]Case seriesGreeceNewborns including preterm,
LOS
6XDR or PDR
Klebsiella pneumoniae
50 mg/kg q8h
Duration: 4–38 days (median 14 d)
6/6 BC sterilization
6/6 clinical recovery
No drug-related AEs
Bradley 2019 [60]Phase II RCTUS3 month–2 years
UTI
95EnterobacteralesRandomization 3:1 to C/A 40–50 mg/kg q8h or cefepime
Duration: ≥72 h
In C/A group:
17/22 urine sterilization
21/22 clinical recovery
No drug-related AEs
Bradley 2019 [61]Phase II RCTUS3 month–18 years
complicated intra-abdominal infection
83Escherichia coli
Pseudomonas aeruginosa
Randomization 3:1 to C/A 40–50 mg/kg q8h + metronidazole or MEM
Duration: ≥72 h
In C/A group:
57/61 clinical recovery
No drug-related AEs
Esposito 2019 [62]Case reportItalyPreterm infant BW 680 g,
LOS + meningitidis (30 DOL)
1KPC-producing Klebsiella pneumoniae75 mg/kg q8h
Duration: 47 days
1/1 BC sterilization
1/1 clinical recovery
1/1 uncertain mild drug-related AE (thrombocytopenia)
Vargas 2019 [63]Case reportItaly14 years
BSI and pneumonia
1MDR Klebsiella pneumoniae2.5 g q8h
Duration: 14 days
1/1 Clinical cure
No drug-related AEs
Nguyem 2019 [64]Case reportUS16 years
CF
Pulmonary exacerbation
1Burkholderia cepacia2.5 g q8h
Duration: 14 days
1/1 Sputum sterilization
1/1 Clinical cure
No drug-related AEs
Hobson 2019 [65]Case reportFrance3 years
BSI in acute leukemia
1MDR Morganella morganii150 mg/kg/d
Duration: n.p.
1/1 BC sterilization
1/1 Clinical cure
No drug-related AEs
Coskum 2020 [66]Case reportTurkeyPreterm infant GA 27 weeks,
UTI (25 DOL)
1PDR Klebsiella pneumoniae40 mg/kg q8h
Duration: 10 days
1/1 urine sterilization
1/1 clinical recovery
1/1 uncertain mild drug-related AE (glycosuria)
Ji 2021 [67]Case reportChina2 month
Shoulder osteomyelitis
1CR Klebsiella pneumoniae200 mg q8h
Duration: 14 days
1/1 clinical recovery
No drug-related AEs
Franzese 2021 [68]PharmacokineticsUS3 month–18 years153n.p.40 mg/kg for infants < 6 month
Single 2 h infusion
Pharmacokinetic model validation
Asfour 2022 [69]Case seriesSaudi ArabiaPreterm infants GA 27–28 weeks LOS + meningitidis (DOL 11)
LOS (DOL 37)
2CR Klebsiella pneumoniae50 mg/kg q8h
Duration: 5–21 days
2/2 BC sterilization
1/2 clinical recovery
1/2 death
No drug-related AEs
Nascimento 2022 [70]Case reportBrazilPreterm infant GA 29 weeks
LOS (46 DOL)
1MDR Klebsiella pneumoniae40 mg/kg q8h
Duration: 14 days
1/1 BC sterilization
1/1 clinical recovery
No drug-related AEs
Almangour 2022 [71]Case reportSaudi Arabia2 years
Ventriculoperitoneal shunt infection
1MDR Pseudomonas aeruginosa62.5 mg/kg q8h
Duration: 21 days
1/1 CSF sterilization
1/1 Clinical cure
No drug-related AEs
Perruccio 2022 [72]Case seriesItaly7 month–17 years
Malignancy
BSI, pneumonia, appendicitis
21
(+4 C/T)
MDR Enterobacterales50mg/kg q8h
Duration: 6–19 days
23/25 Clinical cure
2/25 Death
No drug-related AEs
AEs: adverse events, BC: blood culture, BSI: bloodstream infection, CSF: cerebrospinal fluid, CF: cystic fibrosis, C/A: ceftazidime/avibactam, C/T: ceftolozane/tazobactam, CR: carbapenem-resistant, DOL: days of life, GA: gestational age, LOS: late-onset sepsis, MDR: multi-drug resistant, MEM: meropenem, PDR: pan-drug resistant, UTI: urinary tract infection.
Table 2. Studies on ceftolozane/tazobactam.
Table 2. Studies on ceftolozane/tazobactam.
Study TypeCountryPatient CharacteristicsNOrganismsStudy InterventionOutcomes
Aitken 2016 [73]Case reportUS9 years
BSI in acute leukemia
1MDR P. aeruginosa1st course: 50 mg/kg q8h Duration: 3 weeks
2nd course: 40 mg 7 Kg q6h
Duration: 3 weeks
1/1 BC sterilization (relapse after 1st course)
1/1 Clinical cure (relapse after 1st course)
No drug-related AEs
Bradley 2018 [74]Phase I
Pharmacokinetics
US>7 days <18 years34Proven or suspected Gram neg infection 20 mg/kg for pts 7d–3 month
Single 1 h infusion
Pharmacokinetics model validation
No drug-related AEs
Martin-Cazana 2019 [75]Case reportSpain5 years
BSI and endocarditis in congenital heart disease
1MDR P. aeruginosa50 mg/kg q8h
Extended infusion (3 h)
Duration: 6 weeks
1/1 BC sterilization
1/1 Clinical cure
No drug-related AEs
Zikri 2019 [76]Case reportSaudi Arabia14 years
BSI and pneumonia in immunodeficiency
1MDR P. aeruginosa1.5 g q8h
Duration: n.p.
1/1 Clinical cure
No drug-related AEs
Ang 2019 [77]Phase I
Pharmacokinetics
US>7 days <3 month13Proven or suspected Gram neg infection 20 mg/kg
Single 1 h infusion
Pharmacokinetics model validation
No drug-related AEs
Larson 2020 [78]PharmacokineticsUSsimulation model (0–18 years)Recommended doses:
20 mg/kg q8h (birth–12 years)
Molloy 2020 [79]Case seriesUS3 month–19 years13MDR P. aeruginosa (7 pneumonia, 3 CF, 2 abdominal infections, 1 osteomyelitis)20 mg/kg q8h
Duration: up to 8 weeks
12/13 clinical cure
2/13 uncertain drug-related AEs (transaminitis, neutropenia)
No drug-related AEs in pts < 1 years
Arrieta 2020 [80]PharmacokineticsUS2–18 years
CF
Respiratory disease
18n.p.18–30 mg/kg (2–7 years)100% target attainment probability
No differences CF vs. non-CF
Butragueno-Laiseca 2020 [81]PharmacokineticsUS9–19 month
CF
3MDR P. aeruginosa30–40 mg/kg q8hRecommended doses:
35 mg/kg q8h if normal renal function
10 mg/kg q8h if acute renal injury
30 mg/kg if renal replacement therapy
Perruccio 2022 [72]Case seriesItaly7 month–17 years
Malignancy
BSI, pneumonia, appendicitis
4
(+21C/A)
MDR Enterobacterales1g q8h
Duration: 14–20 days
23/25 Clinical cure
2/25 Death
No drug-related AEs
Roiledes 2023 [82]Phase II RCTUS/Europe7d–18 years
Complicated UTI
95E. coli
K. pneumoniae
P. aeruginosa
Randomization 3:1 to C/T 20 mg/kg q8h or MEM
Duration: 7–14 days
94% vs. 80% clinical cure
14% mild drug-related AEs (diarrhea, increased transaminases, neutropenia)
No severe drug-related AEs
Jackson 2023 [83]Phase II RCTUS/Europe0–18 years
Complicated intra-abdominal infection
91E. coliRandomization to C/T 20 mg/kg q8h + metronidazole or MEMClinical cure: 80 vs. 95%
In C/T group: 13/70 mild drug-related AEs (diarrhea, increased transaminases, increased alkaline phosphatase, vaginal mycosis, dysgeusia)
No sere drug-related AEs
AEs: adverse events, BC: blood culture, BSI: bloodstream infection, CF: cystic fibrosis, C/T: ceftolozane/tazobactam, DOL: days of life, GA: gestational age, MDR: multi drug resistant, MEM: meropenem, UTI: urinary tract infection.
Table 3. Studies on cefiderocol and meropenem/vaborbactam.
Table 3. Studies on cefiderocol and meropenem/vaborbactam.
AntibioticStudy TypeCountryPatient CharacteristicsNOrganismsStudy InterventionOutcomes
Katsube 2019 [84]CefiderocolPharmacokineticsJapansimulation model (0–18 years)Recommended doses:
GA < 32 weeks: <2 month 30 mg/kg q8h
>2 month 40 mg/kg q8h
GA ≥ 32 weeks: <2 month 40 mg/kg q8h
>2 month 60 mg/kg q8h
Alamarat 2020 [85]CefiderocolCase reportUS15 years
Chronic osteomyelitis
1XDR P. aeruginosa +
ESBL producing
K. pneumoniae
2g q8h
Extended infusion (3 h)
Duration: 14 weeks (+surgery)
1/1 Bone biopsy specimen sterilization
1/1 Clinical cure
No drug-related AEs
Warner 2021 [86]CefiderocolCase seriesUS0–18 years
CF
Pulmonary exacerbation
2Achromobacter xylosoxidans60 mg/kg q8h2/2 clinical recovery
1/2 relapse
No drug-related AEs
Grasa 2021 [87]CefiderocolCase reportSpain2 years
BSI in Burkitt lymphoma
1Carbapenemase-producing P. aeruginosa60 mg/kg q8h
Duration: 7 days
1/1 Clinical cure
No drug-related AEs
Bawankule 2022 [88]CefiderocolCase reportIndiaPreterm 27 weeks GA
DOL 9
LOS + NEC
1KPC-producing
K. pneumoniae
30 mg/kg q6h
Duration: 14 days
1/1 BC sterilization
1/1 clinical recovery
No drug-related AEs
Monari 2023 [89]CefiderocolCase reportItalyPreterm 31 weeks GA
DOL 20
LOS
1KPC-producing
K. pneumoniae
60 mg/kg loading dose
40 mg/kg q8h
Extended infusion (3–4 h)
Duration: 9 days
1/1 BC sterilization
1/1 clinical recovery
No drug-related AEs
Henretty 2018 [90]Meropenem/VaborvactamCase report
Pharmacokinetics
US4 years
CLABSI
1KPC-producing
K. pneumoniae
40 mg/kg q6h, 3 h infusion
Duration: 14 days
1/1 BC sterilization
1/1 Clinical cure
100% MEM concentration > MIC
Gainey 2020 [91]Meropenem/Vaborvactam
+Cefiderocol
+bacteriophage
Case reportUS10 years
CF
1Achromobacter spp. resistant to FDC and M/VDuration: 14 days1/1 Sputum sterilization
1/1 Clinical cure
AEs: adverse events, CLABSI: central line-associated bloodstream infection, CF: cystic fibrosis, DOL: days of life, ESBL: extended spectrum beta-lactamase, KPC: Klebsiella pneumoniae carbapenemase; FDC: cefiderocol, GA: gestational age, M/V: meropenem/vaborbactam, NEC: necrotizing enterocolitis, UTI: urinary tract infection, XDR: extensively drug-resistant.
Table 4. Studies on ceftaroline and dalbavancin.
Table 4. Studies on ceftaroline and dalbavancin.
AntibioticStudy TypeCountryPatient CharacteristicsNOrganismsStudy InterventionOutcomes
Cannavino 2016 [92]CeftarolineRCTUS, Europe2 month–17 years,
CABP
160 8 mg/kg q8h (2–6 month)
1 h infusion
Duration:
92% clinical cure
10% drug-related AEs (diarrhea, vomiting)
Blumer 2016 [93]CeftarolineRCTUS2 month–18 years
Complicated CABP
38
2 month–2 years: 6
3/29 MSSA
1/29 MRSA
Others: S. pneumonia, S. pyogenes, H. Influenzae, P. aeruginosa
Randomization 3:1 to ceftaroline 10 mg/kg q8h (2–6 month) or ceftriaxone+ vancomycin
Duration: 3–19 days
In ceftaroline group:
24/29 clinical cure
7/30 drug-related AEs (vomiting, diarrhea, increased AST/ALT, dermatitis, rush)
Korczowski 2016 [94]CeftarolineRCTUS, Europe, South America, Africa2 month–17 years
SSSI
159Mostly S. aureus
(42% MRSA)
Randomization 2:1 to ceftaroline 8 mg/kg q8h (2–6 month) or comparator (cefazolin or vancomycin) Duration: 5–14 days96% clinical cure
94% microbiological eradication
MRSA 89% microbiological eradication
Riccobene 2017 [95]CeftarolinePharmacokineticsUS— simulation model (0–18 years)8 mg/kg q8h (2 month–2 years) has >97% probability of target attainment
Salerno 2018 [96]CeftarolineCase reportUSPreterm 24 weeks GA DOL 43
LOS and pneumonia
1MRSA8.5 mg/kg q8h
Duration: 21 days
1/1 BC sterilization
1/1 clinical cure
No drug-related AEs
Pharmacokinetics target attained
Branstetter 2020 [97]CeftarolineRetrospective
US0–21 years
CF pulmonary exacerbation
90*71/90 MRSA
21/90 Pseudomonas coinfection
Randomization 1:1 to ceftaroline or vancomycin
Doses and duration not reported
No differences in lung function and readmission rate
Bradley 2020 [98]CeftarolinePhase II
US7–60 d,
LOS
11E. coli
Staphyloccocus spp.
4–6 mg/kg q8h, 1h infusion
Duration: 2–14 days
0/11 clinical failure
1/11 mild AE (diarrhea)
Pharmacokinetic target attainment probability > 95%
Ferguson 2020 [99]CeftarolineCase report 20 month
BSI, wound infection, endocarditis, septic pulmonary emboli
1MRSA8 mg/kg q6h
Duration: 1 days (shifted to vancomycin)
Resistance to ceftaroline despite no previous exposure
Heger 2022 [100]CeftarolineCase reportUSPreterm GA 30 weeks
DOL 54
Hepatic abscess and infected portal thrombus
1MRSA8 mg/kg q8h
Duration: 18 days
1/1 clinical cure
No drug-related AEs
Chan 2021 [101]CeftarolinePharmacokineticsUSsimulation model (0–18 years)10 mg/kg q8h (2 month–2 years) has 99% probability of target attainment
Riccobene 2021 [102]CeftarolinePharmacokineticsUSsimulation model 2 month–18 years5 min or 60 min infusion have >99% probability of target attainment
Bradley 2015 [103]DalbavancinPharmacokineticsUS12–17 years10n.p.15 mg/kg single doseSlightly lower exposure than adults given 1 g
Gonzalez 2017 [104]DalbavancinPhase I
Pharmacokinetics
US3 month–11 years
Suspected or confirmed bacterial infection
43n.p.Recommended regimens 3 month–6 years:
15 mg/kg day 1 + 7.5 mg/kg day 8 OR
22.5 mg/kg day 1
5 probable drug-related AEs (rash, dermatitis, urticaria, elevated liver enzymes); no drug-related severe AEs
Carrothers 2023 [105]DalbavancinPharmacokineticsUS— simulation model 0–18 years
SSSI, neonatal sepsis
211n.p.22.5 mg/kg 30 min single infusion has probability of target attainment >94%
Giorgobiani 2023 [106]DalbavancinPhase III RCTUS0–18 years SSSI
0–3 month BSI
191 (0–3 month: 5)S. aureus, S. pyogenes, S. mitis/S. oralis, E. faecalisRandomization 3:3:1 to DAL 1 dose or DAL 2 doses or comparator (<3 month: DAL 1 dose 18 mg/kg or comparator)
30 min infusion
Duration
Clinical cure 97 vs. 99 vs. 89%
No drug-related AEs
BSI: bllodstream infection, CABP: community-acquired bacterial pneumonia, CF: cystic fibrosis, CARTI: community-acquired respiratory infection, DOL: days of life, GA: gestational age, LOS: late-onset sepsis, MRSA: methicillin resistant S. aureus, MSSA: methicillin sensitive S. aureus, SSSI: skin and skin structure infection.
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

Poggi, C.; Dani, C. New Antimicrobials for the Treatment of Neonatal Sepsis Caused by Multi-Drug-Resistant Bacteria: A Systematic Review. Antibiotics 2023, 12, 956. https://doi.org/10.3390/antibiotics12060956

AMA Style

Poggi C, Dani C. New Antimicrobials for the Treatment of Neonatal Sepsis Caused by Multi-Drug-Resistant Bacteria: A Systematic Review. Antibiotics. 2023; 12(6):956. https://doi.org/10.3390/antibiotics12060956

Chicago/Turabian Style

Poggi, Chiara, and Carlo Dani. 2023. "New Antimicrobials for the Treatment of Neonatal Sepsis Caused by Multi-Drug-Resistant Bacteria: A Systematic Review" Antibiotics 12, no. 6: 956. https://doi.org/10.3390/antibiotics12060956

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