Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2022 Jun 28;24(4):e13881. doi: 10.1111/tid.13881

Extended‐spectrum β‐lactamase‐producing and carbapenem‐resistant Enterobacterales bloodstream infection after solid organ transplantation: Recent trends in epidemiology and therapeutic approaches

Elena Pérez‐Nadales 1,2,3,4,, Mario Fernández‐Ruiz 1,5, Belén Gutiérrez‐Gutiérrez 1,6, Álvaro Pascual 1,6, Jesús Rodríguez‐Baño 1,6, Luis Martínez‐Martínez 1,2,3,4, José María Aguado 1,5, Julian Torre‐Cisneros 1,2,3,4
PMCID: PMC9540422  PMID: 35691028

Abstract

Background

Infections caused by multidrug‐resistant gram‐negative bacilli (MDR GNB), in particular extended‐spectrum β‐lactamase‐producing (ESBL‐E) and carbapenem‐resistant Enterobacterales (CRE), pose a major threat in solid organ transplantation (SOT). Outcome prediction and therapy are challenging due to the scarcity of randomized clinical trials (RCTs) or well‐designed observational studies focused on this population.

Methods

Narrative review with a focus on the contributions provided by the ongoing multinational INCREMENT‐SOT consortium (ClinicalTrials identifier NCT02852902) in the fields of epidemiology and clinical management.

Results

The Spanish Society of Transplantation (SET), the Group for Study of Infection in Transplantation of the Spanish Society of Infectious Diseases and Clinical Microbiology (GESITRA‐SEIMC), and the Spanish Network for Research in Infectious Diseases (REIPI) recently published their recommendations for the management of MDR GNB infections in SOT recipients. We revisit the SET/GESITRA‐SEIMC/REIPI document taking into consideration new evidence that emerged on the molecular epidemiology, prognostic stratification, and treatment of post‐transplant ESBL‐E and CRE infections. Results derived from the INCREMENT‐SOT consortium may support the therapeutic approach to post‐transplant bloodstream infection (BSI). The initiatives devoted to sparing the use of carbapenems in low‐risk ESBL‐E BSI or to repurposing existing non‐β‐lactam antibiotics for CRE in both non‐transplant and transplant patients are reviewed, as well as the eventual positioning in the specific SOT setting of recently approved antibiotics.

Conclusion

Due to the clinical complexity and relative rarity of ESBL‐E and CRE infections in SOT recipients, multinational cooperative efforts such as the INCREMENT‐SOT Project should be encouraged. In addition, RCTs focused on post‐transplant serious infection remain urgently needed.

Keywords: antibiotics, carbapenem‐resistant Enterobacterales, extended‐spectrum β‐lactamase‐producing Enterobacterales, INCREMENT‐SOT Project, solid organ transplantation, treatment

1. INTRODUCTION

Multidrug‐resistant gram‐negative bacilli (MDR GNB), including extended‐spectrum β‐lactamase‐producing (ESBL‐E) and carbapenem‐resistant Enterobacterales (CRE), are prevalent infectious agents following solid organ transplantation (SOT). 1 , 2 , 3 Infections by MDR GNB are associated with increased risk for recurrent infection, allograft dysfunction, and mortality as compared to infections due to non‐resistant pathogens. 3 , 4 In addition, these infections are frequently treated with old or novel antimicrobials for which scarce data on safety and efficacy are available. 3 Such circumstances highlight the need to improve the clinical management of ESBL‐E and CRE infections among SOT recipients.

In 2018, the Spanish Society of Transplantation (SET), the Group for Study of Infection in Transplantation of the Spanish Society of Infectious Diseases and Clinical Microbiology (GESITRA‐SEIMC), and the Spanish Network for Research in Infectious Diseases (REIPI) published their recommendations on the management of MDR GNB infections in SOT recipients. 3 This consensus document covers a wide range of relevant aspects of clinical management, from microbiological diagnosis to therapeutic challenges. 3 Since the release of the SET/GESITRA‐SEIMC/REIPI document, additional works have reviewed management principles of MDR GNB in the setting of SOT, 5 , 6 including the recently published guidelines from the Infectious Diseases Community of Practice of the American Society of Transplantation. 5 In addition, the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) has also issued its guidelines, although not restricted to transplant patients. 7

The INCREMENT‐SOT Project is a large international retrospective cohort that includes nearly 800 consecutive SOT recipients diagnosed with bloodstream infection (BSI) due to ESBL‐E and CRE between 2004 and 2016 (ClinicalTrials identifier NCT02852902). 8 , 9 , 10 , 11 The INCREMENT‐SOT consortium is currently integrated by 38 tertiary centers in 16 countries (Figure 1), and this cohort study has been recently extended to cover the period 2016–2021. In addition, 14 centers in seven countries from the consortium (seven hospitals in Spain, two in Italy, one in Belgium, one in the UK, one in Turkey, one in Malta, and one in the USA) have participated in the generation of an international microbial collection of 174 ESBL‐E and CRE isolates, representing 20% of the post‐transplant BSI episodes recorded in the clinical database. The study is mainly aimed at providing high‐level observational data on the molecular epidemiology and therapeutic management of ESBL‐E and CRE BSI in the SOT setting. Other aspects, such as pre‐transplant screening, prophylaxis, or decolonization strategies, are outside the intended scope of the INCREMENT‐SOT Project.

FIGURE 1.

FIGURE 1

The INCREMENT‐solid organ transplantation (SOT) Project is a multinational consortium including 38 SOT centers. This Project generated a large international retrospective cohort of 788 consecutive recipients with bloodstream infection (BSI) due to extended‐spectrum β‐lactamase‐producing (ESBL‐E) and carbapenem‐resistant Enterobacterales (CRE) for the period 2004 to 2016 (ClinicalTrials identifier NCT02852902)

In the present work, we review the main contributions made by the INCREMENT‐SOT consortium and summarize additional recent evidence concerning epidemiological trends, risk factors, use of predictive scores to guide clinical management, and new therapeutic alternatives among SOT recipients with ESBL‐E and CRE infection. To this end, we conducted a set of computer‐based PubMed/MEDLINE (National Library of Medicine, Bethesda, MD) searches using the MeSH terms “SOT” (and specific transplant types) AND “extended‐spectrum β‐lactamase” or “carbapenemase‐producing” (or “carbapenem‐resistant”) AND “Enterobacteriaceae” (or “Enterobacterales”) AND “infection” or “bacteremia” (and specific syndromes) to identify literature pertaining to the topic published until January 2022. In addition, separate searches were performed for each of the antibiotic agents reviewed. Only clinical studies covering molecular epidemiology and therapeutic aspects were considered. The bibliographies of the selected articles were scrutinized for additional relevant references.

2. ESBL‐E INFECTIONS AFTER SOT: EPIDEMIOLOGY, MOLECULAR CHARACTERIZATION, AND CLINICAL FEATURES

The rates of ESBL‐E infection in SOT recipients show geographical variations and have increased in recent years. Approximately 40% of infectious episodes caused by Enterobacterales after SOT are due to ESBL‐E, primarily Escherichia coli and Klebsiella pneumoniae. 12 , 13 , 14 , 15 , 16 A systematic review involving 1089 recipients reported that as much as 18% of them were colonized with ESBL‐E (95% confidence interval [CI]: 5%–36%). The prevalence of ESBL‐E colonization for pre‐transplant candidates was estimated at 7% (95% CI: 5%–9%). Stratifying by transplant type, the ESBL‐E colonization rate was 17% for liver transplant (LT) recipients and 24% for kidney transplant (KT) recipients. According to the continent, no significant differences in the prevalence were identified between studies performed in Europe and North America (15% and 31.4%, respectively). 4

Molecular characterization of the INCREMENT‐SOT strain collection corresponding to 174 recipients with BSI classified 75% of isolates as ESBL‐E and 25% as CRE (10% of which harbored both ESBL and carbapenemase genes). E. coli (53.0%) and K. pneumoniae (40.9%) were the most frequent ESBL‐E, whereas K. pneumoniae (83.7%) accounted for most CRE isolates. bla CTX‐M β‐lactamase genes were the most frequent among ESBL producers, while bla KPC and bla OXA‐48 were the more prevalent carbapenemase genes. 17 Our data are consistent with those obtained from a prospective cohort study conducted between 2014 and 2018 in seven Spanish centers (ENTHERE study), which reported 78.0% of ESBL‐E and 21% of CRE among 531 isolates from LT and KT recipients. 16 In this study, E. coli producing cefotaximase‐Munich (CTX‐M)‐1 group and K. pneumoniae harboring bla OXA‐48 (either alone or in combination with bla CTX‐M‐1) were the most prevalent Enterobacterales causing colonization. 16 A further study analyzing the etiology of post‐transplant BSI in a Spanish center detected an increasing rate of ESBL‐producing strains, mainly K. pneumoniae, from 7% in the period 2007–2008 to 34% in 2015–2016. 2 A nationwide study carried out in Switzerland reported that the proportion of ESBL‐E infections in SOT recipients rose in recent years to 11.4%, although the proportion of ESBL produces among E. coli isolates remained stable over time (in contrast to the increasing trend observed for non‐E. coli strains). 18

Risk factors for developing post‐transplant ESBL‐E infection include previous antibiotic exposure, pre‐transplant colonization, perioperative prophylaxis, prolonged tracheal intubation, long‐term hospitalization, urinary tract obstruction and instrumentation, kidney‐pancreas transplantation, post‐transplant renal replacement therapy (RRT), and recurrent urinary tract infection (UTI), as reviewed elsewhere. 3 , 19 A recent multicenter case‐control study in the USA comprising 988 ESBL‐E BSI episodes identified various risk factors of relevance, including the isolation of ESBL‐E in a prior culture, a corticosteroid‐containing immunosuppression regimen, acute rejection treated with corticosteroids, and previous exposure to third‐generation cephalosporins, echinocandins and trimethoprim‐sulfamethoxazole. 20

The incidence of ESBL‐E infection among LT recipients has been reported to reach 5.5%–13%. 13 , 21 , 22 The median time from transplantation to infection was reported to be 15 days (range: 3–105 days) in a unicentric retrospective cohort in France that included 710 patients between 2001 and 2010. 22 Since the urinary tract is one of the most frequent sources of infection, KT recipients are exposed to a higher incidence. According to a meta‐analysis, the proportion of KT recipients that developed a UTI due to ESBL‐E was 2% in North America, 5% in Europe, 17% in South America, and 33% in Asia. These patients face an almost three‐times increased risk of recurrence compared to those infected with non‐ESBL‐producing strains. 23 In a 3‐year, monocentric retrospective study in France on 659 KT recipients, the median time from transplantation to the occurrence of UTI due to ESBL‐E was 48 days (range: 17–253). 24

The most common sources of post‐transplant BSI by ESBL‐E are intravascular catheters and complicated UTI (cUTI). 25 In a large single‐center retrospective study in Brazil, BSI was more frequent among LT recipients (7% of 238 patients) than KT recipients (3% of 759 patients; p‐value = .005). 26 In this study, the 30‐day mortality rate among 54 recipients that developed ESBL‐E BSI was 26%. However, the proportion of cases of bacteremia secondary to cUTI was significantly higher among KT than LT recipients (60% vs. 11%, p‐value = .008), and the 30‐day mortality rate in these patients was 6.7% only. 26 In the INCREMENT‐SOT cohort, we reported a 30‐day all‐cause mortality rate of 2.9% in the subcohort of 306 KT recipients with non‐severe monomicrobial ESBL‐E BSI secondary to UTI between 2004 and 2016. 11 Consistent with this more favorable outcome, in a second analysis comparing the efficacy of meropenem versus ertapenem in KT recipients with non‐severe bacteremic urinary tract infections due to ESBL‐E, only one meropenem‐treated patient died within the first 30 days from BSI onset. 9

Improved outcome prediction in a complex clinical setting such as MDR GNB infection after SOT is critical in order to inform the decision‐making process. Wang et al. recently developed a clinical prediction tool for post‐transplant ESBL‐E BSI on the basis of a multicenter cohort with 897 patients. They generated a predictive model consisting of 10 variables, which fell into four clinical categories: prior colonization or infection with Enterobacterales, recent antimicrobial exposure, the severity of underlying illness, and type of immunosuppression. The authors recommend that any patient scoring 2 or more points should be deemed at high risk of ESBL‐E BSI and considered for empiric carbapenem therapy while awaiting the results of susceptibility testing. 27

The multinational INCREMENT project preceded INCREMENT‐SOT and was focused on infections due to ESBL‐E and CRE in the general population (ClinicalTrials.gov Identifier NCT01764490). As part of this study, an easy‐to‐collect predictive scoring model for 30‐day all‐cause mortality in ESBL‐E BSI was developed, which has been proven to accurately stratify patients with this condition. 28 Key risk factors included in this score were: age >50 years (3 points), infection due to Klebsiella spp. rather than other species (2 points), a source of infection other than urinary tract infections (3 points), and ultimately or rapidly fatal disease according to the McCabe classification (i.e., death is expected to occur in <5 years as a consequence of the underlying condition, 4 points), a condition of acute severity as measured by a Pitt score >3 (3 points), presentation of BSI with severe sepsis or septic shock (4 points), and inappropriateness of early targeted therapy (2 points). Mortality rates for INCREMENT‐ESBL scores <11 and >11 points were near 5% and 40%, respectively. 28 The predictive accuracy of this score for risk stratification in SOT recipients remains to be investigated.

3. CRE INFECTIONS IN THE SOT POPULATION

The emergence of CRE constitutes a major threat for SOT recipients since infections caused by these difficult‐to‐treat pathogens lead to significant morbidity and mortality. 3 Mortality rates reported for CRE infections following SOT vary from 20% to 80% and are significantly higher in the case of BSI as compared to non‐bacteremic syndromes. 29 , 30 , 31 , 32 , 33 Post‐transplant CRE infections tend to occur within the first two months after the procedure, and the infection site frequently correlates with the transplant type. 3 , 34 Thus, LT recipients commonly develop the surgical site (SSI) and intraabdominal infection (IAI), which are often associated with BSI. On the other hand, UTI and SSI are more common among KT recipients, with a high relapse rate. Finally, lung transplant (LuT) recipients are typically at risk of developing pneumonia. 3 , 5

The molecular epidemiology of CRE in the SOT setting has been characterized in several recent studies. As part of the INCREMENT‐SOT project, we performed a comparative genomic analysis of 35 carbapenemase‐producing K. pneumoniae (CP‐KP) strains from BSI episodes in nine centers (five in Spain, one in Italy, one in Belgium, one in Malta, and one in the USA). bla KPC was detected in 68.6% of isolates, in association with sequence type 512 (ST512) (40%, Italy and Spain), ST258 (20%, Italy and USA), ST307 (6%, Italy), and ST101 (3%, Italy). In addition, bla OXA‐48 was detected in 28.6% of CP‐KP isolates, in association with ST15 (11%, Spain and Belgium), ST11 (9%, Spain), ST392 (3%, Spain), ST405 (3%, Spain), and ST496 (3%, Malta). Finally, bla VIM was found in a single K. pneumoniae isolate of ST1 (3%, Spain). 17 Clancy et al. described a series of 17 recipients with CRE BSI from a single US center, with 16 of them infected with the sequence‐type 258 (ST258) harboring the bla KPC‐2 gene and the remaining patient with a KPC‐3‐producing ST37 strain. 35 In a prospective cohort study in another US center, Macesic et al. performed a comparative genomic analysis of 95 CRE isolates (57 K. pneumoniae, 20 E. coli, 15 Enterobacter cloacae complex, two Citrobacter freundii, and one K. oxytoca) from LT recipients with either colonization (80 isolates) or infection (15 isolates). They identified 26 CRE clades, with K. pneumoniae ST258 being the most common (39%), followed by E. cloacae complex ST252 (8%), K. pneumoniae ST17 (7%), and K. pneumoniae ST307 (5%). The bla KPC gene was detected in 66% of isolates. 36 An Italian study performing genotypic characterization of 81 Klebsiella pneumoniae carbapenemase (KPC)‐producing K. pneumoniae isolates from LT or LuT donors and recipients showed that 83% of them belonged to clonal group 258, in particular to ST258 (20%) and ST512 (60%). The remaining isolates (17%) fell within seven other STs, the most common being ST101. 37 Another recent study that describes the molecular epidemiology of CP‐KP in 80 recipients from Brazil identified 88.8% and 11.2% of isolates harboring bla KPC−2 and bla NDM‐1 respectively. The most common STs were ST11/KPC‐2 (63%), ST437/KPC‐2 (10%) and ST258/KPC‐2 (4%) within clonal complex 11/258 (CC11/258), ST16/KPC‐2 (10%) within CC17, and ST15/NDM‐1 (9%). 38

The incidence of CRE infection in SOT populations varies considerably across centers and transplant types. Reported rates range from 1% to 16% for LT recipients, 1%–11% for KT recipients, and 1%–8% for LuT recipients. 5 , 39 Higher rates of CRE colonization and infection have been reported in studies focused on LT recipients and conducted in centers with established endemicity. 37 , 39 , 40 , 41 A multinational retrospective cohort (CRECOOLT study) included 840 LT recipients from 15 centers in Italy, Brazil, the USA, Spain, and Israel that had been found to be colonized by CRE either before or at transplantation (24.2%) or within the first 180 days after the procedure (75.8%). The overall infection rate was 29.7%, which occurred at a median of 19 post‐transplant days (range: 9–42). 42 A prospective monocentric cohort study on 386 LT recipients in Brazil also revealed high incidence rates of CRE colonization (17.6% and 30.8% before or following transplantation), although the proportion of patients that developed infection was lower (15.7% after a median of 11 days). 41 In a prospective cohort of 553 LT recipients from an Italian center, 7% were pre‐transplant carriers and 19% acquired colonization after the procedure, with a significant increase over the study period (2010–2017). 39 The overall rate of CRE infection was 10.3%, within a median of 31 days (range: 11–115) from transplantation. This figure rose to 34.7% among patients with previous CRE colonization. 39 Finally, in a large cohort of KT recipients colonized with CRE, the median time from transplantation to the first CRE‐positive culture was 42 days, and nearly 38% of patients developed an infection, mostly UTI. 43

Risk factors for CRE infection in LT recipients include CRE carriage acquired before or after transplantation, Model for End‐Stage Liver Disease (MELD) score, multi‐organ transplantation, need for reintervention or RRT, acute kidney injury, prolonged mechanical ventilation, and graft rejection. 39 , 41 , 42 , 44 Freire et al. found the following risk factors in a single‐center Brazilian cohort of 331 KT recipients with pre‐transplant CRE colonization: recipient age at CRE acquisition >50 years, median lymphocyte count ≤700 cells/mcL, carbapenem use, and colonization by a polymyxin‐resistant strain. 43 Other risk factors reported in different studies include multi‐organ transplantation, ureteral stent, length of hospital stay, deceased donor allograft, pre‐transplant CR‐KP infection or colonization, diabetes mellitus, and receipt of antimicrobials other than trimethoprim‐sulfamethoxazole. 31 , 32

Optimization of the management of CRE infection relies on reducing the risk of acquisition, improving early detection, and choosing aggressive antibiotic therapy. 45 In patients with CRE colonization and clinical suspicion of infection, a careful assessment of the underlying conditions and the extent of colonization may serve to guide empirical therapy. As part of the afore‐mentioned CRECOOLT study, Giannella et al. developed a risk prediction model for CRE infection after LT based on the following risk factors: CRE colonization within 60 days before or after transplantation, multisite post‐transplant colonization, prolonged mechanical ventilation, acute kidney injury, and surgical reintervention. The discrimination accuracy of the model in the derivation and bootstrapped validation datasets were acceptable (area under the curve [AUC] of 74.6 and 73.9, respectively). 42 As part of the INCREMENT‐SOT project, we developed the INCREMENT‐SOT‐CPE score to predict 30‐day all‐cause mortality in SOT recipients with BSI due to CRE. 8 The score is based on a previously derived and validated tool for the general population 46 , 47 , 48 enriched with additional transplant‐specific variables (i.e., adequate therapy, source control, lymphopenia, and cytomegalovirus [CMV] disease) (Table 1). The score was able to classify patients into three strata according to the mortality risk: 0–7 (low risk), 8–11 (high risk), and 12–17 (very high risk) (Figure 2). Of note, the INCREMENT‐SOT‐CPE score still remains to be externally validated.

TABLE 1.

The INCREMENT‐SOT‐CPE score for predicting all‐cause 30‐day mortality in solid organ transplantation (SOT) recipients with carbapenem‐resistant Enterobacterales and bloodstream infection (CRE BSI) (adapted from Pérez‐Nadales et al. 80 )

Variable Regression beta coefficients (95% CI) Score
INCREMENT‐CPE score ≥8 a 2.62 (1.79–3.56) 8
CMV disease in the previous 30 days 2.38 (0.58–4.34) 7
Lymphopenia ≤600 cells/mcL 1.24 (0.55–1.97) 4
No source control 0.98 (0.17–1.83) 3
Inappropriate empirical therapy 0.64 (−0.26–1.45) 2
Interaction INCREMENT‐CPE score ≥8 * CMV disease in the previous 30 days b −2.39 (−4.90–−0.10) −7
Maximum score 17

Abbreviations: CMV, cytomegalovirus; CPE, carbapenemase‐producing Enterobacterales; CRE, carbapenem‐resistant Enterobacterales.

a

The INCREMENT‐CPE mortality score was developed to predict all‐cause mortality in carbapenemase‐producing Enterobacterales and includes the following variables: severe sepsis or shock at presentation (5 points), Pitt bacteremia score ≥6 (4 points), Charlson comorbidity index ≥2 (3 points), and source of bloodstream infection other than urinary or biliary tract (3 points).

b

The negative interaction coefficient means that the effect of the combined action of 2 predictors is less than the sum of the individual effects. Consequently, in our model, the maximum score in a patient with all risk factors would be 17.

FIGURE 2.

FIGURE 2

Thirty‐day all‐cause mortality for solid organ transplantation (SOT) recipients with carbapenem‐resistant Enterobacterales and bloodstream infection (CRE BSI) according to increasing categories in the INCREMENT‐SOT‐CPE score and type of active therapy administered (adapted from Pérez‐Nadales et al. 80 )

4. THERAPEUTIC OPTIONS FOR ESBL‐E AND CRE INFECTIONS IN THE SOT POPULATION

As commented above, the evidence‐based SET/GESITRA‐SEIMC/REIPI document was published in 2018. 3 This multidisciplinary initiative was justified by the rapid spread across transplant centers of cephalosporin‐ and carbapenem‐resistant strains and by the particular challenges that transplant and infectious diseases physicians face in treating MDR GNB infections. Beyond their increased susceptibility to infection related to long‐term immunosuppression, the SOT population shows distinct clinical characteristics in comparison to non‐transplant patients, such as the repeated exposure to the healthcare system and invasive procedures, the frequent presence of renal or liver impairment associated with nephrotoxic therapies (i.e., calcineurin inhibitors [CNIs]) or underlying conditions, and the risk of drug‐to‐drug interactions. The limited therapeutic options for MDR GNB often require the use of second‐line agents with unacceptable toxicity, such as colistin. On the other hand, the evidence available to guide the management of these difficult‐to‐treat pathogens derives from randomized clinical trials (RCTs) and observational studies that rarely provide separate outcomes for SOT recipients, thus compromising the extrapolation of results.

In this section, we summarize some of the SET/GESITRA‐SEIMC/REIPI recommendations in the light of the new evidence that emerged since their publication, with a focus on the contributions made by the INCREMENT‐SOT consortium for the treatment of ESBL‐E and CRE BSI. The initiatives taken to design carbapenem‐sparing regimens by repositioning existing drugs—either β‐lactams or belonging to other classes—are reviewed. Finally, we summarize the main features and approved indications of the novel antibiotics, as well as the existing experience and their potential positioning in the treatment of post‐transplant infection (Table 2).

TABLE 2.

Recently approved antibiotic agents with potential activity against extended‐spectrum β‐lactamase‐producing (ESBL‐E) and carbapenem‐resistant Enterobacterales (CRE)

Agent Approved indications Dose (normal renal function) Experience in SOT recipients Considerations
Cefiderocol

cUTI, HAP/VAP (FDA)

Infections due to aerobic GNB with limited treatment options (EMA)

2 g IV every 8 h Case reports

The only agent with activity against MBL‐producing CRE

FDA warning due to increased all‐cause mortality in phase III RCT compared to best available therapy

Potential for resistance selection

Ceftazidime‐avibactam

cUTI, cIAI, HAP/VAP (FDA, EMA)

Infections due to aerobic GNB with limited treatment options (EMA)

2/0.5 g IV every 8 h Case reports, case series, comparative retrospective studies

Activity against OXA‐48‐producing CRE

Potential activity against MBL‐producing CRE in combination with aztreonam

Meropenem‐vaborbactam

cUTI (FDA)

cUTI, cIAI, HAP/VAP (EMA)

Infections due to aerobic GNB with limited treatment options (EMA)

2/2 g IV every 8 h Case reports

Lower potential for resistance selection than CAZ‐AVI

No additional activity beyond meropenem for MDR Pseudomonas

No activity against OXA‐48‐type and MBL‐producing CRE

Imipenem‐cilastatin‐relebactam

cUTI, cIAI, HAP/VAP (FDA)

HAP/VAP, infections due to aerobic GNB with limited treatment options (EMA)

500/500/125 mg IV every 6 h None No activity against OXA‐48‐type, GES‐type, and MBL‐producing CRE
Plazomicin cUTI (FDA) 15 mg/Kg IV every 24 h None

Potential role of monotherapy

Increased risk of nephrotoxicity with calcineurin inhibitors

Not available in Europe

Eravacycline cIAI (FDA, EMA) 1 mg/Kg IV every 24 h Small case series

More potent activity, better tissue penetration, and lower potential for resistance selection than tigecycline

Interaction with strong CYP3A4 inducers

Unfavorable PK/PD profile for BSI

Should be avoided in cUTI

Abbreviations: BSI, bloodstream infection; CAZ‐AVI, ceftazidime‐avibactam; cIAI, complicated intraabdominal infection; CRE, carbapenem‐resistant Enterobacterales; cUTI, complicated urinary tract infection; EMA, European Medicines Agency; FDA, Food and Drug Administration; HAP/VAP, hospital‐acquired pneumonia/ventilator‐associated pneumonia; MBL, metallo‐β‐lactamase; PK/PD, pharmacokinetic/pharmacodynamic; RCT, randomized clinical trial; SOT, solid organ transplantation.

4.1. Old drugs for emerging problems

4.1.1. Repositioning β‐lactams

A notable amount of research has been devoted over the last years to defining the role of non‐carbapenem‐based approaches for ESBL‐E BSI from low‐risk (urinary and biliary) sources, with the aim to spare the use of carbapenems and the subsequent ecological pressure. 49 , 50 Existing β‐lactam/β‐lactamase inhibitor combinations (BLBLI) appear as a good alternative since a variable proportion of ESBL‐E isolates across geographical areas (from 20% to over 60%) remain susceptible in vitro to amoxicillin‐clavulanate or piperacillin‐tazobactam. 51 , 52 , 53 , 54 , 55 , 56 Gutiérrez‐Gutiérrez et al. analyzed more than 600 patients with monomicrobial BSI due to ESBL‐E (mainly CTX‐M‐type‐producing E. coli) within the INCREMENT project and found a comparable 14‐day cure/improvement rate and 30‐day all‐cause mortality in those receiving active BLBLI or carbapenem monotherapy, either in the empirical or targeted therapy groups. The non‐inferiority of BLBLIs was consistent across different subgroup analyses (e.g., type of ESBL‐E, source of infection, or clinical severity) and after propensity score (PS) adjustment. 57 These findings were supported by previous, smaller observational studies. 58 , 59 Conflictive results, however, had been also reported, with poorer outcomes for BLBLI (particularly with piperacillin‐tazobactam for K. pneumoniae). 60 , 61 , 62 Concerns exist on the potential impact of the inoculum effect (i.e., a significant increase in minimal inhibitory concentrations [MICs] with increasing bacterial density), the source of infection, or the coexistence of CTX‐M‐type enzymes with other β‐lactamases (oxacillinase [OXA]‐1/30). 63 In an attempt to clarify this key question, an open‐label non‐inferiority RCT compared piperacillin‐tazobactam versus meropenem as the definitive therapy for BSI due to ceftriaxone‐resistant E. coli or K. pneumoniae (MERINO trial). Since the primary outcome of 30‐day all‐cause mortality was more common in the piperacillin‐tazobactam than in the meropenem arm (12.3% vs. 3.7%, respectively), the non‐inferiority assumption could not be demonstrated. 64 This difference was attenuated, however, when non‐susceptible strains centrally tested by broth microdilution and whole‐genome sequencing were excluded, 65 or by considering only patients with a urinary tract source or low comorbidity burden. 50

On the basis of moderate‐quality evidence, the SET/GESITRA‐SEIMC/REIPI document states that carbapenems should be recommended as the empirical or targeted treatment of SOT recipients with moderate‐to‐severe ESBL‐E infections, although BLBLI therapy seems reasonable for nonbacteremic episodes, particularly UTI. 3 Ongoing immunosuppression and the frequent instrumentation of the urinary tract may limit the extrapolation to the SOT population of results showing the non‐inferiority of BLBLI therapy. 66 With such uncertainties in mind, we took advantage of the INCREMENT‐SOT Project to compare the effectiveness of active BLBLI‐based monotherapy initiated within the first 72 h from the onset of monomicrobial ESBL‐E BSI secondary to UTI with that of carbapenems in the setting of KT. Sensitivity analyses were conducted at 24‐hour and 7‐day windows. Overall, 306 ESBL‐E BSI episodes were included, with E. coli (62.1%) and Klebsiella spp. (35.0%) accounting for the majority of isolates. Consistent with the low‐risk nature of the source of infection, therapeutic failure (i.e., lack of cure or clinical improvement and/or death from any cause) was rare (8.2% on day 7 and 13.4% on day 30), as was all‐cause mortality (2.9%). Most patients received carbapenem‐based monotherapy for the first 72 h (68.6%), whereas BLBLI was used in 10.8% of cases only (mostly piperacillin‐tazobactam at 4/0.5 g every 8 h). Hospital‐acquired BSI and the Pitt bacteremia score were independent predictors of therapeutic failure by day 7. We observed no significant differences in the rates of therapeutic failure between KT recipients treated with carbapenem or BLBLI monotherapy, either at day 7 (9.0% vs. 3.0%, respectively; risk difference: ‐6.01%; 95% CI: ‐0.16–0.04; odds ratio [OR]: 3.18; p‐value = .267) or day 30 (13.8% vs. 9.1%; risk difference: ‐4.72%; 95% CI: ‐0.17–0.08; OR: 1.60; p‐value = .459). Due to baseline imbalances between groups, we constructed a PS for the use of carbapenem monotherapy (that included the geographical area, pre‐transplant heart failure, chronic pulmonary disease, presence of a rapidly or ultimately fatal disease, and early receipt of active therapy). Again, neither the risk of therapeutic failure at day 7 (PS‐adjusted OR: 4.36; 95% CI: 0.51–37.38; p‐value = .179) or day 30 (PS‐adjusted OR: 2.59; 95% CI: 0.66–10.21; p‐value = .175) were significantly influenced by the type of antibiotic regimen. 11 +++ Although caution should be exercised in the interpretation of these results due to the low numbers in the BLBLI group and the risk of underpowered sample size, our study would suggest that—provided in vitro susceptibility—BLBLIs may be a reasonable alternative to carbapenems for ESBL‐E BSI from a urinary source in KT recipients, supporting SET/GESITRA‐SEIMC/REIPI recommendations. On the other hand, this conclusion is in line with the results reported for hematological neutropenic patients, including hematopoietic stem cell transplant recipients. 67 The debate on to what extent carbapenems may be safely spared in favor of BLBLIs is far from being settled, 68 , 69 pending on the results of ongoing RCTs. 70

Another relevant question is whether ertapenem and meropenem are equally effective for ESBL‐E BSI from a urinary source. The increasing prevalence of ESBL producers among uropathogens in KT recipients has contributed to the widespread use of carbapenems. 71 In contrast to group 2 carbapenems (imipenem, meropenem, or doripenem), ertapenem displays no significant activity against non‐fermenting GNB and would contribute to decrease—or at least not add to—the risk of selective pressure on Pseudomonas aeruginosa and Acinetobacter baumannii. 72 , 73 , 74 Moreover, ertapenem has more convenient dosing for outpatient parenteral antibiotic therapy (OPAT). Nevertheless, doubts have been raised on the probability of attaining appropriate pharmacokinetic/pharmacodynamic (PK/PD) parameters with the conventional ertapenem dosing in patients with septic shock (due to their increased volume of distribution) or for ESBL‐E isolates different than E. coli. 75 We performed a subanalysis of the INCREMENT‐SOT cohort restricted to KT recipients with bacteremic UTI that received targeted monotherapy with ertapenem or meropenem (100 and 101 patients, respectively). The clinical cure rate by day 14 was comparable between patients treated with ertapenem (45.0%) or meropenem (50.5%), whereas the median hospital stay was shorter in the former group (10.5 vs. 14 days; p‐value = .008). In the PS‐adjusted multivariable analysis, targeted therapy with ertapenem was not associated with a lower odds of achieving clinical cure as compared with meropenem (OR: 1.29; 95% CI: 0.51–3.22). This result was confirmed by the inverse probability of treatment weighting and PS‐matching. The desirability of outcome ranking analysis suggested that ertapenem may have some advantages in this setting, such as once‐daily administration, lower ecological impact, and the possibility of OPAT. 9

4.1.2. The role of combination therapy

The potential benefit of combination therapy for MDR GNB remains controversial in the non‐transplant population. Available studies are observational in nature and often limited by low sample sizes and heterogeneity in the definition of “combination therapy” (i.e., number of agents, in vitro activity, duration). 7 , 76 For instance, a large multicenter study comprising 661 patients with BSI and non‐bacteremic infection due to KPC‐producing K. pneumoniae reported that combination therapy with at least two drugs displaying in vitro activity was associated with lower mortality, mainly in patients with septic shock. Meropenem‐containing combinations were effective only if the MIC value of the isolate was ≤8 mg/L. 77 Some 78 but not all meta‐analyses 79 have reported differences in different outcomes—clinical success, microbiological cure, or mortality—between patients treated with monotherapy or combination therapy for CRE. The recent ESCMID guidelines conditionally recommend combination therapy for certain scenarios only, such as severe infections due to metallo‐β‐lactamases (MBL)‐producing CRE or isolates with resistance to new antibiotics. 7 Existing experience in the SOT setting was much scarce, highlighting the contributions of the INCREMENT‐SOT Consortium. Recipients with CRE BSI treated with a single active agent overall showed increased 30‐day mortality as compared to those receiving combination therapy. This difference, however, was only evident within the highest strata of the INCREMENT‐SOT CPE score (adjusted hazard ratio [aHR] for 12–17 points: 2.82; 95% CI: 1.13–7.06; aHR for 8–11 points: 9.93; 95% CI: 2.08–47.40). In contrast, no apparent benefit derived from the use of combination therapy was observed among patients in the low‐risk stratum (≤7 points) (Figure 2). 80 These results mirror those obtained from the INCREMENT cohort for non‐transplant patients, 81 and lead us to propose an algorithm for clinical management (Figure 3). It should be noted that considerable heterogeneity existed in the specific combinations administered, with most of them including an aminoglycoside, colistin, or tigecycline. In addition, the potential impact of “immortal time bias” on the protective effect observed for combination therapy among recipients with higher scores cannot be completely ruled out, although patients dying within the first 48 h after the blood cultures were obtained had been excluded from the analysis. 80 Finally, these studies were conducted before the introduction of ceftazidime‐avibactam (CAZ‐AVI) or other new BLBLIs reviewed below.

FIGURE 3.

FIGURE 3

Proposed algorithm for the choice of definitive therapy for post‐transplant CRE BSI (adapted from Pérez‐Nadales et al. 80 ). BSI, bloodstream infection; CMV, cytomegalovirus; CRE, carbapenem‐resistant Enterobacterales; SOT, solid organ transplantation

4.1.3. Non β‐lactam alternatives

In addition to BLBLIs, the reprofiling of existing antibiotics has been proposed as an alternative basis for the design of carbapenem‐sparing regimens for ESBL‐E and, in some cases, CRE. These “revisited” drugs include aminoglycosides, tigecycline, fosfomycin, and colistin among others. 50 All of them are positioned as second‐line regimens by the SET/GESITRA‐SEIMC/REIPI document due to their adverse effects and the increased mortality rate when compared to β‐lactams. 3

Aminoglycoside monotherapy—with preference given to amikacin—may constitute a valid choice for selected cases with limited therapeutic options, such as non‐bacteremic UTI due to ESBL‐E. 82 , 83 In the INCREMENT cohort the use of aminoglycosides as the only active agent for a median of 4 days was not associated with increased mortality in ESBL‐E BSI compared to carbapenems, although the number of patients analyzed was low. 84 The GESITRA/SEIMC consensus statement on the management of post‐transplant UTI published in 2015 supported monotherapy with gentamicin or amikacin for cystitis due to CRE. 85 The administration of aminoglycosides for more than 2–3 days among SOT recipients, however, is limited by the development of nephrotoxicity, particularly in the presence of concomitant CNI exposure. 86

Tigecycline is not affected by β‐lactamases and retains in vitro activity against most ESBL‐E isolates—with the notable exception of the Morganellaceae family—and a variable proportion (from 40% to more than 90%) of CRE isolates, including MBLs producers. 87 , 88 , 89 , 90 , 91 , 92 There have been some favorable experiences with tigecycline in the treatment of MDR GNB, 93 , 94 and a meta‐analysis pooling data from 21 studies (none of them randomized) suggested efficacy in CRE similar to other antibiotics, particularly as combination therapy or at high doses. 95 In an Italian study comprising 125 patients with KPC‐producing K. pneumoniae BSI, combination therapy with at least two active agents was associated with a lower mortality rate. The best outcomes were obtained with the triple‐drug regimen comprising tigecycline, colistin, and meropenem. 96 Nevertheless, and likely due to its bacteriostatic action, tigecycline has been consistently shown to be associated with higher mortality when used off‐label to treat serious infections, 97 , 98 which resulted in a warning by the Food and Drug Administration (FDA). The low urine concentration achieved with tigecycline prevents its use for UTIs. As for SOT recipients, experience with tigecycline for CRE is mainly limited to single case reports 99 or small series. 100 , 101 , 102 The SET/GESITRA‐SEIMC/REIPI recommendations state that tigecycline may be considered as the combination drug associated with a carbapenem (provided that the MIC value ≤8 mg/L) for non‐urinary CRE infections, whereas monotherapy should be reserved to non‐severe infections. 3 After the publication of this document, a single‐center retrospective study found that SOT recipients with polymicrobial IAI treated with tigecycline were less likely to achieve favorable clinical outcomes and experienced more adverse events (AEs) than comparator broad‐spectrum agents. 103 On the other hand, KT recipients on CNIs may be at an increased risk of developing tigecycline‐induced acute pancreatitis. 104 , 105 , 106

Despite its five decades of existence, fosfomycin has recently regained attention due to its unique mode of action—the irreversible inhibition of the first cytoplasmic step of peptidoglycan synthesis—that results in a potent bactericidal activity with minimal cross‐resistance with other classes of antibiotics. 107 , 108 A non‐negligible proportion of ESBL‐E and CRE isolates remain susceptible, 109 although decreasing rates are being gradually reported. 110 , 111 , 112 Oral formulations (calcium salt and trometamol) have a favorable safety profile, whereas the intravenous fosfomycin disodium is associated with a high sodium intake that may limit its use in patients with heart failure or undergoing hemodialysis. 113 A multicenter retrospective study performed in Spain included 143 episodes of uncomplicated UTI (cystitis) in KT recipients treated with oral fosfomycin, with rates of clinical and microbiological cure of 83.9% and 70.2%, respectively. Half of the isolates had an MDR phenotype (ESBL‐E in 14.0%, CRE in 3.5%), although the odds of microbiological cure were not decreased in these episodes (71.4%). 114 Similar results have been also reported for asymptomatic bacteriuria within the first post‐transplant months. 115 Concordant with these observational experiences, the administration of a single 4‐g dose of intravenous fosfomycin disodium before the placement or removal of a urinary catheter or a double‐J ureteral stent resulted in a lower incidence of symptomatic UTI or asymptomatic bacteriuria during the first weeks after KT as compared with placebo. 116 Evidence supporting the use of intravenous fosfomycin for serious GNB infection is still emerging. 117 A recent open‐label RCT compared intravenous fosfomycin versus a β‐lactam (ceftriaxone or meropenem) for patients with BSI from urinary source due to MDR (mainly ESBL‐producing) E. coli. While clinical or microbiological failure was lower with fosfomycin than with the comparator (14.3% vs. 19.7%, respectively), the primary outcome of non‐inferiority was not met due to an increased rate of AE‐related discontinuations in the former group, mostly due to the occurrence of heart failure (which happened only in patients over 80 years and those with preexisting heart disease or renal failure). 118 Although fosfomycin showed high efficacy, this trial exemplifies that patients to be treated with intravenous fosfomycin at doses of 4–6 g every 6–8 h should be properly selected. Moreover, this formulation is not available in some countries. The SET/GESITRA‐SEIMC/REIPI recommendations limit the use of fosfomycin trometamol for non‐severe infections with an adequate site penetration (in particular UTI), whereas intravenous fosfomycin should only be considered for CRE infections as part of a combination regimen which includes at least one more active agent, preferably as three‐drug regimens. 3

4.2. New β‐lactam actors in the MDR landscape

4.2.1. Cefiderocol

Cefiderocol is a third‐generation cephalosporin structurally related to ceftazidime and cefepime, which is conjugated with a catechol moiety on the C‐3 side chain. Similar to other β‐lactams, cefiderocol transits the outer cell membrane of gram‐negative bacteria by passive diffusion through porins. In addition, it actively enters the periplasmic compartment—where high concentrations are achieved—by exploiting the bacterial siderophore‐iron complex pathway as a “Trojan horse”. 119 This mode of action overcomes common mechanisms of β‐lactam resistance such as loss of porin expression or up‐regulation of efflux pumps and results in stability against hydrolysis by various types of carbapenemases, including most serine enzymes (e.g., KPC, OXA type) and MBLs. 120 Cefiderocol also retains antibacterial activity against ESBL‐ and AmpC‐producing strains. 121 Its clinical development program comprised three non‐inferiority phase II/III RCTs. 122 , 123 , 124 One of them compared cefiderocol with the best available therapy (BAT)—as chosen by the investigator and including a maximum of three drugs—in patients with hospital‐acquired pneumonia (HAP), BSI, or cUTI due to carbapenem‐resistant GNB. Pseudomonas aeruginosa and A. baumannii were the most common pathogens in the microbiological modified intention‐to‐treat (mITT) population. The rate of clinical cure was similar between both groups across the qualifying infectious syndromes. Nevertheless, numerically more deaths occurred in patients receiving cefiderocol (33.7% vs. 18.4% in the BAT arm), mainly in the subgroup with Acinetobacter infection. 122 This unexpected finding led the FDA to issue a warning, although the underlying explanation is unclear and might be related to the heterogeneity in patient characteristics, small sample size, and imbalances between study groups. The remaining two trials demonstrated the non‐inferiority of cefiderocol to high‐dose, extended‐infusion meropenem in terms of all‐cause mortality in patients with HAP, 123 or to imipenem‐cilastatin in terms of clinical and microbiological outcomes in cUTI. 124 The safety profile observed is concordant with that expected for cephalosporins, and dose adjustment is required in moderate to severe renal impairment. 119 Cefiderocol was FDA approved in 2019 for the treatment of cUTI (including pyelonephritis) and in 2020 for HAP and ventilator‐associated pneumonia (VAP). The European Medicines Agency (EMA) granted a generic approval for infections due to aerobic GNBs with limited therapeutic options. A common feature of the novel agents for the treatment of ESBL‐E and CRE reviewed herein is the scarcity of data available for the specific population of SOT recipients. This limitation also applies for cefiderocol, with only a few cases reported. 125 , 126 , 127 , 128 A LT recipient that developed liver abscesses and BSI due to KPC‐producing K. pneumoniae resistant to CAZ‐AVI was successfully treated with cefiderocol for >10 days. 126 AKT recipient was treated with cefiderocol in combination with CAZ‐AVI a polymyxin B for a complicated IAI (cIAI) due to two genetically different K. pneumoniae strains carrying bla NDM‐1, bla OXA‐232, and bla CTX‐M‐15 genes, with bacterial clearance. 125 It has been also reported the case of an LT recipient with cUTI due to an MDR C. freundii strain harboring bla KPC‐1 and bla NDM‐3 that was extensively resistant to CAZ‐AVI (MIC >256 mg/L), in which clinical cure was achieved after a 10‐day course of cefiderocol (MIC = 1 mg/L). 127 The rapid development of high‐level resistance in E. cloacae within the first weeks of cefiderocol therapy has been recently reported in an LT recipient. Whole‐genome sequencing identified functional alterations in the cirA gene encoding for the catecholate siderophore receptor used by the cefiderocol‐ferric complex to enter the periplasmic compartment. 128 Pending the generation of clinical experience, cefiderocol appears as a promising option for MDR GNB infection in SOT recipients.

4.2.2. New BLBLI combinations

Three new BLBLIs have been recently added to the therapeutic armamentarium against MDR GNB: CAZ‐AVI, meropenem‐vaborbactam (MER‐VAB), and imipenem‐cilastatin‐relebactam (IMI‐REL). 129 , 130 , 131 All of them retain activity against ESBL‐E and AmpC‐producing Enterobacterales, although some important differences must be noted. Avibactam is the first member of the diazabicyclooctanone non‐β‐lactam class and exhibits the broadest spectrum of β‐lactamase inhibition, including Ambler class A (e.g., KPC‐ and Guiana extended‐spectrum [GES]‐type), class C (Amp‐C cephalosporinases), and some class D enzymes (notably OXA‐48‐like). 132 The FDA approved CAZ‐AVI in 2015 for the treatment of cUTI and cIAI, and in 2018 for patients with HAP/VAP. Soon after its release, descriptions emerged of the development of CAZ‐AVI resistance in KPC‐producing strains, typically driven by the D179Y substitution in the bla KPC‐3 gene that affects the stability of the Ω‐loop. 133 , 134 Vaborbactam is a new cyclic boronate β‐lactamase inhibitor with potent activity against KPC‐2 and KPC‐3 enzymes, as well as ESBLs (e.g., sulfhydryl variable, temonera, CTX‐M) 135 and class C cephalosporinases. 130 Vaborbactam is also able to overcome CAZ‐AVI resistance due to D179Y mutations at the KPC binding site. 136 Of note, carbapenem‐hydrolyzing class D enzymes are not affected by vaborbactam, 137 a notable limitation in European countries such as Spain where OXA‐48‐like enzymes are the most common type of carbapenemase. 138 , 139 Resistance to MER‐VAB among KPC‐producing isolates is mediated by a combination of KPC production and mutations in the OmpK35 and OmpK36 porins, although the odds of resistance developing seem to be lower than with CAZ‐AVI. 140 , 141 Finally, relebactam is also a diazabicyclooctane β‐lactamase inhibitor, like avibactam, that bears a piperidine ring in the R1 side chain. 131 , 142 Similar to MER‐VAB, IMI‐REL is highly effective against CRE isolates carrying bla KPC‐2 and bla KPC‐3 genes and ESBL‐E and AmpC‐producing Enterobacterales. Nevertheless, the inhibitory activity against most OXA‐type and GES‐type carbapenemase‐producing strains is poor. 143 , 144 It should be highlighted that none of these novel BLBLI combinations retain significant activity in the presence of MBLs. 129

Experience with new BLBLIs among SOT recipients has been mainly restricted to case reports 145 , 146 , 147 , 148 and small series with no comparator group. 149 , 150 The SET/GESITRA‐SEIMC/REIPI document did not make any mention of MER‐VAB or IMI‐REL and simply suggested—based on low‐quality evidence (CIII)—that CAZ‐AVI may be considered for the treatment of CRE infections if the strain shows in vitro susceptibility. 3 In view of this research gap, we have conducted a subanalysis within the INCREMENT‐SOT cohort in which the effectiveness of CAZ‐AVI was compared with BAT (mainly including colistin, tigecycline, or fosfomycin) in 210 recipients diagnosed with monomicrobial CRE BSI from 2016 to 2021. In the primarily targeted therapy cohort, 85 and 81 patients were analyzed within the CAZ‐AVI and BAT groups, respectively. There were significant differences favoring CAZ‐AVI over BAT in the 14‐day (81.2% vs. 58.0%, p = .001) and 30‐day (81.2% vs. 60.5%, p = .004) clinical success rate as well as in 30‐day mortality (12.9% vs. 27.2%, P = 0.036) that persisted after multivariate adjustment. Interestingly, we were able to validate in this cohort the good performance of the INCREMENT‐SOT CPE Score to predict the risk of death (adjusted OR per one‐point increment: 1.17; 95% CI: 1.08–1.28, p < 0.001) (data not yet published).

4.3. Non‐β‐lactam classes or the promising newcomers

4.3.1. Plazomicin

Plazomicin is a novel semisynthetic parenteral aminoglycoside that inhibits bacterial protein synthesis. 151 Thanks to various structural modifications it retains activity in the presence of most aminoglycoside modifying enzymes, as well as ESBLs and carbapenemases (including MBLs). 151 , 152 , 153 A phase III RCT showed the non‐inferiority of plazomicin to meropenem for cUTI. The rate of microbiological eradication at the test‐of‐cure visit in patients with ESBL‐E infection was numerically higher in the plazomicin group (82.4% vs. 75.0%, respectively). 154 A second small open‐label trial recruited patients with CRE BSI or HAP/VAP that were randomized to receive plazomicin or colistin in combination with meropenem or tigecycline. The composite outcome of death from any cause at 28 days or disease‐related complications in the microbiological mITT population was numerically lower with plazomicin than colistin. Such a benefit was observed only in the subgroup of patients with BSI, although the numbers were too low to draw reliable conclusions since the trial was prematurely terminated due to slow enrollment. 155 Plazomicin received FDA approval in 2018 for adult patients with cUTI, whereas the application for marketing authorization in Europe was withdrawn by the manufacturer in June 2020. Unfortunately, clinical experience in SOT recipients is still lacking. Of note, this population was excluded from the pivotal trial for cUTI as per study protocol. 154 Similar to other aminoglycosides, the risk of nephrotoxicity remains a concern with plazomicin, in particular among patients with impaired baseline renal function and higher cumulative drug exposure. 154 This safety profile would further limit its use in the KT setting. It is likely that therapeutic drug monitoring will be needed to optimize exposure and clinical outcomes. 156 Other AEs observed include ototoxicity, gastrointestinal disturbances, and hypotension. 151

4.3.2. Eravacycline

Eravacycline is a novel, fully synthetic fluorocycline that reversibly binds to the 30S ribosomal subunit, blocking the elongation phase of bacterial protein synthesis. In line with other members of the tetracycline class, eravacycline shows a broad spectrum of in vitro activity that comprises methicillin‐resistant Staphylococcus aureus, vancomycin‐resistant enterococci, anaerobes, A. baumannii and some ESBL‐E and CRE. 157 Susceptibility rates among CRE isolates range from 96.2% to 98.0% for E. coli and from 66.6% to 84.0% for K. pneumoniae. 158 , 159 , 160 , 161 Interestingly, eravacycline shows reasonable activity against New Delhi MBL (NDM)‐producing Enterobacterales, with a reported susceptibility rate of 66.2% according to the CLSI interpretive criteria, 162 and has been shown to be 2‐ to 4‐fold more active than tigecycline against CRE. 163 In addition, it remains active against the main acquired tetracycline‐specific resistance mechanisms (efflux pumps and ribosomal protection proteins). 163 On the basis of two non‐inferiority phase III RCTs, 164 , 165 intravenous eravacycline was approved by the FDA and EMA in 2018 for the treatment of cIAI. Eravacycline requires dose adjustment in severe liver function impairment (Child‐Pugh class C) and with the concomitant use of strong CYP3A4 inducers. In addition to its improved activity, eravacycline offers advantages over tigecycline, such as higher serum and tissue concentrations, the existence of both oral and intravenous formulations, and better gastrointestinal tolerability. 157 Again, the available experience in SOT is very limited, as this condition was considered an exclusion criterion in pivotal trials. 164 , 165 A retrospective study with 66 patients from three US centers that received eravacycline for a mean of 13.1 days—mainly as monotherapy for off‐label indications such as pneumonia or skin and soft tissue infection—included 7 SOT recipients (10.6%). The overall rate of clinical improvement was 95.5%, although outcomes for the SOT group were not separately provided. 166 The high volume of distribution of eravacycline (58.3–320.0 L) confers a theoretically unfavorable PK/PD profile for treating BSI, although the pooled clinical and microbiologic outcomes in patients with bacteremic cIAI recruited in the pivotal trials were similar to those observed in the carbapenem arms. 167

5. CONCLUSIONS AND FUTURE RESEARCH AVENUES

As outlined in the present review, MDR GNB constitutes a rapidly evolving threat to modern medicine that adopts particular epidemiological, clinical, and therapeutic features in the SOT population. Notable advances have been made in the management of ESBL‐E and CRE infections after SOT since the publication, 4 years ago, of the SET/GESITRA‐SEIMC/REIPI recommendations. 3 The findings generated by the INCREMENT‐SOT consortium have helped to refine the prognostic stratification in SOT recipients with CRE BSI—highlighting the impact of previous CMV disease and lymphopenia—as well as the role of antibiotic combination therapy in this setting. 80 Other relevant contributions include the comparable effectiveness of active BLBLI‐based monotherapy versus carbapenems for ESBL‐E BSI in KT recipients, 11 +++ and the potential advantages of ertapenem compared to group 2 carbapenems to minimize selective antibiotic pressure in such patients 9 . Beyond the reprofiling of existing antibiotics (such as aminoglycosides, tigecycline, or fosfomycin) to spare the use of carbapenems against ESBL‐E or to provide alternative agents in CRE infection, recent years have witnessed the approval of novel β‐lactam and non‐β‐lactam agents. In view of their activity in the presence of most serine carbapenemases and ESBLs and their favorable PK/PD and safety profiles, newer BLBLIs combinations and cefiderocol are particularly promising. The clinical experience reported with these agents in SOT recipients (summarized in Table 3), however, is so far limited. In this line, the INCREMENT‐SOT Project is generating real‐life data for CAZ‐AVI in the treatment of post‐transplant CRE BSI. Despite these achievements, many challenges persist. In particular, the development of effective agents that retain activity against MBL‐producing Enterobacterales represents an unmet need. It must be highlighted that the literature supporting the clinical benefit of combination therapy for patients with more severe infections—higher INCREMENT‐SOT‐CPE scores—was produced before the novel therapeutic options became available (particularly new BLBLIs). Therefore, further studies should be conducted to eventually confirm such findings. A better understanding is required of how immunosuppression should be managed in recipients with serious ESBL‐E and CRE infections (e.g., BSI, cIAI, or HAP/VAP) and the long‐term consequences on graft function. In addition, robust evidence derived from RCTs to guide the treatment of ESBL‐E and CRE infections after SOT is still essentially lacking. The INCREMENT‐SOT Cohort exemplifies the usefulness of multinational initiatives specifically aimed at providing high‐quality observational data on the optimal therapeutic approach to MDR GNB infections in the SOT population.

TABLE 3.

Summary of the reported experience with novel antibiotic agents for the treatment of solid organ transplantation (SOT) recipients with extended‐spectrum β‐lactamase‐producing (ESBL‐E) and carbapenem‐resistant Enterobacterales (CRE) infections

Agent, reference Type of SOT Type of infection Isolate, molecular resistance mechanism (if available) Outcomes
Cefiderocol 125 KT recipient BSI and cIAI following allograft nephrectomy Two different carbapenem‐resistant Klebsiella pneumoniae strains (ST14 and ST2497) carrying bla NDM‐1, bla OXA‐232, bla CTX‐M‐15, armA, and tet(D)

Clinical and microbiological response

Non‐related death (ischemic colitis and multiorgan failure)

Cefiderocol 126 LT recipient BSI and cIAI (liver abscesses) Carbapenem‐resistant K. pneumoniae Clinical and microbiological response
Cefiderocol 127 LT recipient cUTI Citrobacter freundii complex carrying bla NDM‐1 and bla KPC‐3

Clinical and microbiological response

Non‐related death (angiosarcoma)

Cefiderocol 128 LT recipient BSI and cIAI (multiple biloma and hepatic abscesses) Carbapenem‐resistant Enterobacter cloacae (ST96) carrying bla NDM‐5 and bla OXA‐48 Initial microbiological response, the emergence of resistant isolate (mutations in cirA gene) leading to relapse of infection and death
Meropenem‐vaborbactam 145 LT recipient BSI and cIAI (liver abscess due to hepatic artery thrombosis) Carbapenem‐ and CAZ‐AVI‐resistant K. pneumoniae carrying mutant bla KPC‐2 with D179Y mutation within the KPC Ω‐loop Clinical and microbiological response
Ceftazidime‐avibactam 146 KT recipient Recurrent UTI Carbapenem‐resistant K. pneumoniae Clinical and microbiological response
Ceftazidime‐avibactam 147 KT recipient BSI, cUTI, and pneumonia (probable donor‐derived infection) Carbapenem‐resistant K. pneumoniae carrying bla KPC‐2 Initial clinical and microbiological response, relapse of BSI after 11 days with an appropriate response after a second course
Ceftazidime‐avibactam 148 KT recipient Vertebral osteomyelitis Carbapenem‐resistant K. pneumoniae carrying bla KPC Clinical response
Ceftazidime‐avibactam 149 LuT recipients (n = 10) Pneumonia and/or tracheobronchitis (n = 9), BSI and cIAI (n = 1) Carbapenem‐resistant K. pneumoniae carrying bla KPC‐2

Clinical and microbiological response in 9/10 (90%)

Treatment failure in 1/10 (10%)

Relapse of lower respiratory tract infection in 5/10 (50%)

30‐ and 90‐day survival of 100% and 90%

Ceftazidime‐avibactam 150 LuT recipients (n = 4) Pneumonia (n = 10) MDR K. pneumoniae Clinical and microbiological response in 10/10 (100%)
Eravacycline 166 SOT recipients (n = 7) within a larger cohort (n = 66) NA for SOT recipients MDR GNB or GPC (NA for SOT recipients)

NA for SOT recipients

Overall cohort:

Clinical response in 63/66 (95.5%)

Treatment‐emergent adverse events in 3/66 (4.5%)

Abbreviations: BSI, bloodstream infection; CAZ‐AVI, ceftazidime‐avibactam; cIAI, complicated intraabdominal infection; cUTI, complicated urinary tract infection; GNB, gram‐negative bacilli; GPC, gram‐positive cocci; KT, kidney transplantation; LT, liver transplantation; LuT, lung transplantation; MDR, multidrug‐resistant; NA, not available; SOT, solid organ transplantation.

CONFLICT OF INTEREST

The authors declare no conflict of interest.

AUTHOR CONTRIBUTIONS

Elena Pérez‐Nadales, Mario Fernández‐Ruiz, and Julian Torre‐Cisneros conceived the review study. Elena Pérez‐Nadales and Mario Fernández‐Ruiz drafted the manuscript. Belén Gutiérrez‐Gutiérrez, Álvaro Pascual, Jesús Rodríguez‐Baño, Luis Martínez‐Martínez, José María Aguado, and Julian Torre‐Cisneros critically reviewed the manuscript.

Pérez‐Nadales E, Fernández‐Ruiz M, Gutiérrez‐Gutiérrez B, et al. Extended‐spectrum β‐lactamase‐producing and carbapenem‐resistant Enterobacterales bloodstream infection after solid organ transplantation: Recent trends in epidemiology and therapeutic approaches. Transpl Infect Dis. 2022;24:e13881. 10.1111/tid.13881

Elena Pérez‐Nadales and Mario Fernández‐Ruiz contributed equally to this work.

REFERENCES

  • 1. Camargo LFA, Marra AR, Pignatari ACC, et al. Nosocomial bloodstream infections in a nationwide study: comparison between solid organ transplant patients and the general population. Transpl Infect Dis. 2015;17(2):308–313. 10.1111/TID.12356 [DOI] [PubMed] [Google Scholar]
  • 2. Oriol I, Sabé N, Simonetti AF, et al. Changing trends in the aetiology, treatment and outcomes of bloodstream infection occurring in the first year after solid organ transplantation: a single‐centre prospective cohort study. Transpl Int. 2017;30(9):903–913. 10.1111/TRI.12984 [DOI] [PubMed] [Google Scholar]
  • 3. Aguado JM, Silva JT, Fernández‐Ruiz M, et al. Management of multidrug resistant gram‐negative bacilli infections in solid organ transplant recipients: SET/GESITRA‐SEIMC/REIPI recommendations. Transplant Rev. 2018;32(1):36–57. 10.1016/j.trre.2017.07.001 [DOI] [PubMed] [Google Scholar]
  • 4. Alevizakos M, Kallias A, Flokas ME, Mylonakis E. Colonization with extended‐spectrum beta‐lactamase‐producing Enterobacteriaceae in solid organ transplantation: A meta‐analysis and review. Transpl Infect Dis. 2017;19(4). 10.1111/TID.12718 [DOI] [PubMed] [Google Scholar]
  • 5. Pouch SM, Satlin MJ. Carbapenem‐resistant Enterobacteriaceae in special populations: solid organ transplant recipients, stem cell transplant recipients, and patients with hematologic malignancies. Virulence. 2017;8(4):391–402. 10.1080/21505594.2016.1213472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Smibert O, Satlin MJ, Nellore A, Peleg AY. Carbapenem‐resistant Enterobacteriaceae in solid organ transplantation: management principles. Curr Infect Dis Rep. 2019;21(7):26. 10.1007/S11908-019-0679-4 [DOI] [PubMed] [Google Scholar]
  • 7. Paul M, Carrara E, Retamar P, et al. European Society of clinical microbiology and infectious diseases (ESCMID) guidelines for the treatment of infections caused by Multidrug‐resistant gram‐negative bacilli (endorsed by ESICM ‐European Society of intensive care Medicine). Clin Microbiol Infect. 2022;28(4):521–547. [DOI] [PubMed] [Google Scholar]
  • 8. Pérez‐Nadales E, Gutiérrez‐Gutiérrez B, Natera AM, et al. Predictors of mortality in solid organ transplant recipients with bloodstream infections due to carbapenemase‐producing Enterobacterales: the impact of cytomegalovirus disease and lymphopenia. Am J Transplant. 2020;20(6):1629–1641. 10.1111/ajt.15769 [DOI] [PubMed] [Google Scholar]
  • 9. Gutiérrez‐Gutiérrez B, Pérez‐Nadales E, Pérez‐Galera S, et al. Propensity score and desirability of outcome ranking analysis of ertapenem for treatment of nonsevere bacteremic urinary tract infections due to extended‐spectrum‐beta‐lactamase‐producing Enterobacterales in kidney transplant recipients. Antimicrob Agents Chemother. 2021;65(11):e0110221. 10.1128/AAC.01102-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Gutiérrez‐Gutiérrez B, Pérez‐Nadales E, Rodríguez‐Baño J, Torre‐Cisneros J. Reply to ‘CMV merits further evolutionary and biological view’. Am J Transplant. 2020;20(5):1467–1468. 10.1111/AJT.15867 [DOI] [PubMed] [Google Scholar]
  • 11. Pierrotti LC, Pérez‐Nadales E, Fernández‐Ruiz M, et al. Efficacy of β‐lactam/β‐lactamase inhibitors to treat extended‐spectrum beta‐lactamase‐producing Enterobacterales bacteremia secondary to urinary tract infection in kidney transplant recipients (INCREMENT‐SOT Project). Transpl Infect Dis. 2021;23(3):e13520. 10.1111/tid.13520k [DOI] [PubMed] [Google Scholar]
  • 12. Vidal E, Torre‐Cisneros J, Blanes M, et al. Bacterial urinary tract infection after solid organ transplantation in the RESITRA cohort. Transpl Infect Dis. 2012;14(6):595–603. 10.1111/J.1399-3062.2012.00744.X [DOI] [PubMed] [Google Scholar]
  • 13. Santoro‐Lopes G, De Gouvêa EF. Multidrug‐resistant bacterial infections after liver transplantation: an ever‐growing challenge. World J Gastroenterol. 2014;20(20):6201–6210. 10.3748/WJG.V20.I20.6201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Pouch SM, Patel G. Multidrug‐resistant gram‐negative bacterial infections in solid organ transplant recipients‐Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant. 2019;33(9):e13594. 10.1111/CTR.13594 [DOI] [PubMed] [Google Scholar]
  • 15. Linares L, Cervera C, Cofán F, et al. Risk factors for infection with extended‐spectrum and AmpC beta‐lactamase‐producing gram‐negative rods in renal transplantation. Am J Transplant. 2008;8(5):1000–1005. 10.1111/J.1600-6143.2008.02197.X [DOI] [PubMed] [Google Scholar]
  • 16. Fernández‐Martínez M, González‐Rico C, Gozalo‐Margüello M, et al. Molecular characterization of multidrug resistant Enterobacterales strains isolated from liver and kidney transplant recipients in Spain. Sci Rep. 2021;11(1):1–9. 10.1038/s41598-021-90382-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Perez‐Nadales E, Gracia‐Ahufinger I, Lopez‐Cerero L, et al. Characterisation of an international microbial collection of ESBL‐ and carbapenemase‐producing Enterobacteriaceae isolated from bloodstream infections in solid organ transplant patients: the INCREMENT‐SOT Project (ClinicalTrials.gov ID NTC02852902). In: 29th ECCMID, 13‐16 April 2019, Amsterdam, the Netherlands. https://neumoexpertosdotorg.files.wordpress.com/2019/04/final_programme_web.pdf
  • 18. Kohler P, Wolfensberger A, Stampf S, et al. Temporal trends, risk factors and outcomes of infections due to extended‐spectrum β‐lactamase producing Enterobacterales in Swiss solid organ transplant recipients between 2012 and 2018. Antimicrob Resist Infect Control. 2021;10(1):50. 10.1186/S13756-021-00918-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Silva JT, Fernández‐Ruiz M, Aguado JM. Multidrug‐resistant gram‐negative infection in solid organ transplant recipients: implications for outcome and treatment. Curr Opin Infect Dis. 2018;31(6):499–505. 10.1097/QCO.0000000000000488 [DOI] [PubMed] [Google Scholar]
  • 20. Anesi JA, Blumberg EA, Han JH, et al. Impact of donor multidrug‐resistant organisms on solid organ transplant recipient outcomes. Transpl Infect Dis. 2021;24(1):e13783. 10.1111/TID.13783 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Linares L, Cervera C, Hoyo I, et al. Klebsiella pneumoniae infection in solid organ transplant recipients: epidemiology and antibiotic resistance. Transplant Proc. 2010;42(8):2941–2943. 10.1016/J.TRANSPROCEED.2010.07.080 [DOI] [PubMed] [Google Scholar]
  • 22. Bert F, Larroque B, Paugam‐Burtz C, et al. Pretransplant fecal carriage of extended‐spectrum β‐lactamase–producing Enterobacteriaceae and infection after liver transplant, France. Emerg Infect Dis. 2012;18(6):908. 10.3201/EID1806.110139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Alevizakos M, Nasioudis D, Mylonakis E. Urinary tract infections caused by ESBL‐producing Enterobacteriaceae in renal transplant recipients: a systematic review and meta‐analysis. Transpl Infect Dis. 2017;19(6). 10.1111/TID.12759 [DOI] [PubMed] [Google Scholar]
  • 24. Pilmis B, Scemla A, Join‐Lambert O, et al. ESBL‐producing Enterobacteriaceae‐related urinary tract infections in kidney transplant recipients: incidence and risk factors for recurrence. Infect Dis. 2015;47(10):714–718. 10.3109/23744235.2015.1051107 [DOI] [PubMed] [Google Scholar]
  • 25. Linares L, García‐Goez JF, Cervera C, et al. Early bacteremia after solid organ transplantation. Transplant Proc. 2009;41(6):2262–2264. 10.1016/J.TRANSPROCEED.2009.06.079 [DOI] [PubMed] [Google Scholar]
  • 26. Aguiar EB, Maciel LC, Halpern M, et al. Outcome of bacteremia caused by extended‐spectrum β‐lactamase‐producing Enterobacteriaceae after solid organ transplantation. Transplant Proc. 2014;46(6):1753–1756. 10.1016/J.TRANSPROCEED.2014.05.003 [DOI] [PubMed] [Google Scholar]
  • 27. Wang R, Han JH, Lautenbach E, et al. Clinical prediction tool for extended‐spectrum beta‐lactamase‐producing Enterobacterales as the etiology of a bloodstream infection in solid organ transplant recipients. Transpl Infect Dis. 2021;23(4):e13599. 10.1111/TID.13599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Palacios‐Baena ZR, Gutierrez‐Gutierrez B, De Cueto M, et al. Development and validation of the INCREMENT‐ESBL predictive score for mortality in patients with bloodstream infections due to extended‐spectrum‐β‐lactamase‐producing Enterobacteriaceae. J Antimicrob Chemother. 2017;72(3):906–913. 10.1093/JAC/DKW513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Kalpoe JS, Sonnenberg E, Factor SH, et al. Mortality associated with carbapenem‐resistant Klebsiella pneumoniae infections in liver transplant recipients. Liver Transpl. 2012;18(4):468–474. 10.1002/LT.23374 [DOI] [PubMed] [Google Scholar]
  • 30. Pereira MR, Scully BF, Pouch SM, et al. Risk factors and outcomes of carbapenem‐resistant Klebsiella pneumoniae infections in liver transplant recipients. Liver Transpl. 2015;21(12):1511–1519. 10.1002/LT.24207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Pouch SM, Kubin CJ, Satlin MJ, et al. Epidemiology and outcomes of carbapenem‐resistant Klebsiella pneumoniae bacteriuria in kidney transplant recipients. Transpl Infect Dis. 2015;17(6):800. 10.1111/TID.12450 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Freire MP, Abdala E, Moura ML, et al. Risk factors and outcome of infections with Klebsiella pneumoniae carbapenemase‐producing Klebsiella pneumoniae in kidney transplant recipients. Infection. 2015;43(3):315–323. 10.1007/S15010-015-0743-4 [DOI] [PubMed] [Google Scholar]
  • 33. Simkins J, Muggia V, Cohen HW, Minamoto GY. Carbapenem‐resistant Klebsiella pneumoniae infections in kidney transplant recipients: a case‐control study. Transpl Infect Dis. 2014;16(5):775–782. 10.1111/TID.12276 [DOI] [PubMed] [Google Scholar]
  • 34. Lanini S, Costa AN, Puro V, et al. Incidence of carbapenem‐resistant gram negatives in Italian transplant recipients: a nationwide surveillance study. PLoS One. 2015;10(4):e0123706. 10.1371/JOURNAL.PONE.0123706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Clancy CJ, Chen L, Shields RK, et al. Epidemiology and molecular characterization of bacteremia due to carbapenem‐resistant Klebsiella pneumoniae in transplant recipients. Am J Transplant. 2013;13(10):2619–2633. 10.1111/AJT.12424 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Macesic N, Gomez‐Simmonds A, Sullivan SB, et al. Genomic surveillance reveals diversity of multidrug‐resistant organism colonization and infection: a prospective cohort study in liver transplant recipients. Clin Infect Dis. 2018;67(6):905–912. 10.1093/CID/CIY199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Errico G, Gagliotti C, Monaco M, et al. Colonization and infection due to carbapenemase‐producing Enterobacteriaceae in liver and lung transplant recipients and donor‐derived transmission: a prospective cohort study conducted in Italy. Clin Microbiol Infect. 2019;25(2):203–209. 10.1016/J.CMI.2018.05.003 [DOI] [PubMed] [Google Scholar]
  • 38. Raro OHF, da Silva RMC, Filho EMR, et al. Carbapenemase‐producing Klebsiella pneumoniae From Transplanted Patients in Brazil: phylogeny, resistome, virulome and mobile genetic elements harboring blaKPC–2 or blaNDM–1. Front Microbiol. 2020;11:1563. 10.3389/FMICB.2020.01563/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Giannella M, Bartoletti M, Campoli C, et al. The impact of carbapenemase‐producing Enterobacteriaceae colonization on infection risk after liver transplantation: a prospective observational cohort study. Clin Microbiol Infect. 2019;25(12):1525–1531. 10.1016/J.CMI.2019.04.014 [DOI] [PubMed] [Google Scholar]
  • 40. Massa E, Michailidou E, Agapakis D, et al. Colonization and infection with extensively drug resistant gram‐negative bacteria in liver transplant recipients. Transplant Proc. 2019;51(2):454–456. 10.1016/J.TRANSPROCEED.2019.01.076 [DOI] [PubMed] [Google Scholar]
  • 41. Freire MP, Oshiro ICVS, Pierrotti LC, et al. Carbapenem‐resistant Enterobacteriaceae acquired before liver transplantation: impact on recipient outcomes. Transplantation. 2017;101(4):811–820. 10.1097/TP.0000000000001620 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Giannella M, Freire M, Rinaldi M, et al. Development of a risk prediction model for Carbapenem‐resistant Enterobacteriaceae infection after liver transplantation: a multinational cohort study. Clin Infect Dis. 2021;73(4):E955–E966. 10.1093/CID/CIAB109 [DOI] [PubMed] [Google Scholar]
  • 43. Freire MP, Carvalho LB, Reusing JO, et al. Carbapenem‐resistant Enterobacteriaceae among kidney transplant recipients ‐ insights on the risk of acquisition and CRE infection. Infect Dis (London, England). 2021;53(6):430–439. 10.1080/23744235.2021.1887511 [DOI] [PubMed] [Google Scholar]
  • 44. Giannella M, Bartoletti M, Morelli MC, et al. Risk factors for infection with carbapenem‐resistant Klebsiella pneumoniae after liver transplantation: the importance of pre‐ and posttransplant colonization. Am J Transplant. 2015;15(6):1708–1715. 10.1111/AJT.13136 [DOI] [PubMed] [Google Scholar]
  • 45. Wong D, Van Duin D. Carbapenemase‐producing organisms in solid organ transplantation. Curr Opin Organ Transplant. 2019;24(4):490–496. 10.1097/MOT.0000000000000664 [DOI] [PubMed] [Google Scholar]
  • 46. Gutiérrez‐Gutiérrez B, Salamanca E, de Cueto M, et al. Effect of appropriate combination therapy on mortality of patients with bloodstream infections due to carbapenemase‐producing Enterobacteriaceae (INCREMENT): a retrospective cohort study. Lancet Infect Dis. 2017;17(7):726–734. 10.1016/S1473-3099(17)30228-1 [DOI] [PubMed] [Google Scholar]
  • 47. Gutiérrez‐Gutiérrez B, Salamanca E, de Cueto M, et al. A predictive model of mortality in patients with bloodstream infections due to Carbapenemase‐producing Enterobacteriaceae . Mayo Clin Proc. 2016;91(10):1362–1371. 10.1016/J.MAYOCP.2016.06.024 [DOI] [PubMed] [Google Scholar]
  • 48. Machuca I, Gutiérrez‐Gutiérrez B, Rivera‐Espinar F, et al. External validation of the INCREMENT‐CPE mortality score in a carbapenem‐resistant Klebsiella pneumoniae bacteraemia cohort: the prognostic significance of colistin resistance. Int J Antimicrob Agents. 2019;54(4):442–448. 10.1016/j.ijantimicag.2019.07.017 [DOI] [PubMed] [Google Scholar]
  • 49. Harris PNA, Tambyah PA, Paterson DL. β‐lactam and β‐lactamase inhibitor combinations in the treatment of extended‐spectrum β‐lactamase producing Enterobacteriaceae: time for a reappraisal in the era of few antibiotic options? Lancet Infect Dis. 2015;15(4):475–485. 10.1016/S1473-3099(14)70950-8 [DOI] [PubMed] [Google Scholar]
  • 50. Gutiérrez‐Gutiérrez B, Rodríguez‐Baño J. Current options for the treatment of infections due to extended‐spectrum beta‐lactamase‐producing Enterobacteriaceae in different groups of patients. Clin Microbiol Infect. 2019;25(8):932–942. 10.1016/J.CMI.2019.03.030 [DOI] [PubMed] [Google Scholar]
  • 51. Hayami H, Takahashi S, Ishikawa K, et al. Second nationwide surveillance of bacterial pathogens in patients with acute uncomplicated cystitis conducted by Japanese Surveillance Committee from 2015 to 2016: antimicrobial susceptibility of Escherichia coli, Klebsiella pneumoniae, and Staphylococcus saprophyticus. J Infect Chemother. 2019;25(6):413–422. 10.1016/J.JIAC.2019.02.021 [DOI] [PubMed] [Google Scholar]
  • 52. Toy T, Pak GD, Duc TP, et al. Multicountry distribution and characterization of extended‐spectrum β‐Lactamase‐associated gram‐negative bacteria from bloodstream infections in Sub‐Saharan Africa. Clin Infect Dis. 2019;69(Suppl 6):S449–S458. 10.1093/CID/CIZ450 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Dowzicky MJ, Chmelařová E. Antimicrobial susceptibility of gram‐negative and gram‐positive bacteria collected from Eastern Europe: Results from the tigecycline evaluation and surveillance trial (T.E.S.T.), 2011–2016. J Glob Antimicrob Resist. 2019;17:44–52. 10.1016/J.JGAR.2018.11.007 [DOI] [PubMed] [Google Scholar]
  • 54. Chang YT, Coombs G, Ling T, et al. Epidemiology and trends in the antibiotic susceptibilities of gram‐negative bacilli isolated from patients with intra‐abdominal infections in the Asia‐Pacific region, 2010–2013. Int J Antimicrob Agents. 2017;49(6):734–739. 10.1016/J.IJANTIMICAG.2017.01.030 [DOI] [PubMed] [Google Scholar]
  • 55. Logan LK, Braykov NP, Weinstein RA, Laxminarayan R. Extended‐spectrum β‐Lactamase‐producing and third‐generation cephalosporin‐resistant Enterobacteriaceae in children: trends in the United States, 1999–2011. J Pediatric Infect Dis Soc. 2014;3(4):320–328. 10.1093/JPIDS/PIU010 [DOI] [PubMed] [Google Scholar]
  • 56. Ponce‐de‐Leon A, Rodríguez‐Noriega E, Morfín‐Otero R, et al. Antimicrobial susceptibility of gram‐negative bacilli isolated from intra‐abdominal and urinary‐tract infections in Mexico from 2009 to 2015: results from the study for monitoring antimicrobial resistance trends (SMART). PLoS One. 2018;13(6). 10.1371/JOURNAL.PONE.0198621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Gutiérrez‐Gutiérrez B, Pérez‐Galera S, Salamanca E, et al. A multinational, preregistered cohort study of β‐Lactam/β‐Lactamase inhibitor combinations for treatment of bloodstream infections due to extended‐spectrum‐β‐Lactamase‐producing Enterobacteriaceae. Antimicrob Agents Chemother. 2016;60(7):4159–4169. 10.1128/AAC.00365-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Rodríguez‐Baño J, Navarro MD, Retamar P, Picón E, Pascual Á. β‐Lactam/β‐lactam inhibitor combinations for the treatment of bacteremia due to extended‐spectrum β‐lactamase‐producing Escherichia coli: a post hoc analysis of prospective cohorts. Clin Infect Dis. 2012;54(2):167–174. 10.1093/CID/CIR790 [DOI] [PubMed] [Google Scholar]
  • 59. Ng TM, Khong WX, Harris PNA, et al. Empiric piperacillin‐tazobactam versus Carbapenems in the treatment of bacteraemia due to extended‐spectrum Beta‐Lactamase‐producing enterobacteriaceae. PLoS One. 2016;11(4):e0153696. 10.1371/JOURNAL.PONE.0153696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Tamma PD, Han JH, Rock C, et al. Carbapenem therapy is associated with improved survival compared with piperacillin‐tazobactam for patients with extended‐spectrum β‐lactamase bacteremia. Clin Infect Dis. 2015;60(9):1319–1325. 10.1093/CID/CIV003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ofer‐Friedman H, Shefler C, Sharma S, et al. Carbapenems versus piperacillin‐tazobactam for bloodstream infections of nonurinary source caused by extended‐spectrum Beta‐Lactamase‐producing enterobacteriaceae. Infect Control Hosp Epidemiol. 2015;36(8):981–985. 10.1017/ICE.2015.101 [DOI] [PubMed] [Google Scholar]
  • 62. Tsai HY, Chen YH, Tang HJ, et al. Carbapenems and piperacillin/tazobactam for the treatment of bacteremia caused by extended‐spectrum β‐lactamase‐producing Proteus mirabilis. Diagn Microbiol Infect Dis. 2014;80(3):222–226. 10.1016/J.DIAGMICROBIO.2014.07.006 [DOI] [PubMed] [Google Scholar]
  • 63. Perez F, Bonomo RA. Editorial commentary: Bloodstream infection caused by extended‐spectrum β‐lactamase‐producing gram‐negative bacteria: how to define the best treatment regimen? Clin Infect Dis. 2015;60(9):1326–1329. 10.1093/CID/CIV007 [DOI] [PubMed] [Google Scholar]
  • 64. Harris PNA, Tambyah PA, Lye DC, et al. Effect of piperacillin‐tazobactam vs meropenem on 30‐day mortality for patients with E. coli or Klebsiella pneumoniae bloodstream infection and ceftriaxone resistance: a randomized clinical trial. JAMA. 2018;320(10):984–994. 10.1001/JAMA.2018.12163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Henderson A, Paterson DL, Chatfield MD, et al. Association between minimum inhibitory concentration, beta‐Lactamase genes and mortality for patients treated with piperacillin/tazobactam or meropenem from the MERINO Study. Clin Infect Dis. 2021;73(11):e3842–e3850. 10.1093/CID/CIAA1479 [DOI] [PubMed] [Google Scholar]
  • 66. Vardakas KZ, Tansarli GS, Rafailidis PI, Falagas ME. Carbapenems versus alternative antibiotics for the treatment of bacteraemia due to Enterobacteriaceae producing extended‐spectrum β‐Lactamases: a systematic review and meta‐analysis. J Antimicrob Chemother. 2012;67(12):2793–2803. 10.1093/JAC/DKS301 [DOI] [PubMed] [Google Scholar]
  • 67. Gudiol C, Royo‐Cebrecos C, Abdala E, et al. Efficacy of β‐Lactam/β‐Lactamase inhibitor combinations for the treatment of bloodstream infection due to extended‐spectrum‐β‐Lactamase‐producing enterobacteriaceae in hematological patients with neutropenia. Antimicrob Agents Chemother. 2017;61(8):e00164‐e00167. 10.1128/AAC.00164-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Paterson DL, Isler B, Harris PNA. PRO: carbapenems should be used for ALL infections caused by ceftriaxone‐resistant Enterobacterales. JAC Antimicrob Resist. 2021;3(1):dlab013. 10.1093/JACAMR/DLAB013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Rodríguez‐Baño J, Gutiérrez‐Gutiérrez B, Pascual A. CON: carbapenems are NOT necessary for all infections caused by ceftriaxone‐resistant Enterobacterales. JAC Antimicrob Resist. 2021;3(1):dlaa112. 10.1093/JACAMR/DLAA112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Bitterman R, Koppel F, Mussini C, et al. Piperacillin‐tazobactam versus meropenem for treatment of bloodstream infections caused by third‐generation cephalosporin‐resistant Enterobacteriaceae: a study protocol for a non‐inferiority open‐label randomised controlled trial (PeterPen). BMJ Open. 2021;11(2):e040210. 10.1136/BMJOPEN-2020-040210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Origüen J, Fernández‐Ruiz M, López‐Medrano F, et al. Progressive increase of resistance in Enterobacteriaceae urinary isolates from kidney transplant recipients over the past decade: narrowing of the therapeutic options. Transpl Infect Dis. 2016;18(4):575–584. 10.1111/TID.12547 [DOI] [PubMed] [Google Scholar]
  • 72. Nicolau DP, Carmeli Y, Crank CW, et al. Carbapenem stewardship: does ertapenem affect Pseudomonas susceptibility to other carbapenems? A review of the evidence. Int J Antimicrob Agents. 2012;39(1):11–15. 10.1016/J.IJANTIMICAG.2011.08.018 [DOI] [PubMed] [Google Scholar]
  • 73. Graber CJ, Hutchings C, Dong F, Lee W, Chung JK, Tran T. Changes in antibiotic usage and susceptibility in nosocomial Enterobacteriaceae and Pseudomonas isolates following the introduction of ertapenem to hospital formulary. Epidemiol Infect. 2012;140(1):115–125. 10.1017/S0950268811000082 [DOI] [PubMed] [Google Scholar]
  • 74. Goldstein EJC, Citron DM, Peraino V, Elgourt T, Meibohm AR, Lu S. Introduction of ertapenem into a hospital formulary: effect on antimicrobial usage and improved in vitro susceptibility of Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2009;53(12):5122–5126. 10.1128/AAC.00064-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Gutiérrez‐Gutiérrez B, Bonomo RA, Carmeli Y, et al. Ertapenem for the treatment of bloodstream infections due to ESBL‐producing Enterobacteriaceae: a multinational pre‐registered cohort study. J Antimicrob Chemother. 2016;71(6):1672–1680. 10.1093/JAC/DKV502 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Savoldi A, Carrara E, Piddock LJV, et al. The role of combination therapy in the treatment of severe infections caused by carbapenem resistant gram‐negatives: a systematic review of clinical studies. BMC Infect Dis. 2021;21(1):5254. 10.1186/S12879-021-06253-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Tumbarello M, Viale P, Bassetti M, De Rosa FG, Spanu T, Viscoli C. Infections caused by KPC‐producing Klebsiella pneumoniae: differences in therapy and mortality in a multicentre study–authors’ response. J Antimicrob Chemother. 2015;70(10):2922. 10.1093/JAC/DKV200 [DOI] [PubMed] [Google Scholar]
  • 78. Hou SY, Wu D, Feng XH. Polymyxin monotherapy versus polymyxin‐based combination therapy against carbapenem‐resistant Klebsiella pneumoniae: a systematic review and meta‐analysis. J Glob Antimicrob Resist. 2020;23:197–202. 10.1016/J.JGAR.2020.08.024 [DOI] [PubMed] [Google Scholar]
  • 79. Li D, Fei F, Yu H, et al. Ceftazidime‐avibactam therapy versus ceftazidime‐avibactam‐based combination therapy in patients with carbapenem‐resistant gram‐negative pathogens: a meta‐analysis. Front Pharmacol. 2021;12:2283. 10.3389/FPHAR.2021.707499/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Pérez‐Nadales E, Gutiérrez‐Gutiérrez B, Natera AM, et al. Predictors of mortality in solid organ transplant recipients with bloodstream infections due to carbapenemase‐producing Enterobacterales: the impact of cytomegalovirus disease and lymphopenia. Am J Transplant. 2019;20(6):1629–1641. 10.1111/AJT.15769 [DOI] [PubMed] [Google Scholar]
  • 81. Gutiérrez‐Gutiérrez B, Salamanca E, de Cueto M, et al. Effect of appropriate combination therapy on mortality of patients with bloodstream infections due to carbapenemase‐producing Enterobacteriaceae (INCREMENT): a retrospective cohort study. Lancet Infect Dis. 2017;17(7):726–734. 10.1016/S1473-3099(17)30228-1 [DOI] [PubMed] [Google Scholar]
  • 82. Ipekci T, Seyman D, Berk H, Celik O. Clinical and bacteriological efficacy of amikacin in the treatment of lower urinary tract infection caused by extended‐spectrum beta‐lactamase‐producing Escherichia coli or Klebsiella pneumoniae . J Infect Chemother. 2014;20(12):762–767. 10.1016/J.JIAC.2014.08.007 [DOI] [PubMed] [Google Scholar]
  • 83. Cho SY, Choi SM, Park SH, Lee DG, Choi JH, Yoo JH. Amikacin therapy for urinary tract infections caused by extended‐spectrum β‐lactamase‐producing Escherichia coli . Korean J Intern Med. 2016;31(1):156–161. 10.3904/KJIM.2016.31.1.156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Palacios‐Baena ZR, Gutiérrez‐Gutiérrez B, Calbo E, et al. Empiric therapy with carbapenem‐sparing regimens for bloodstream infections due to extended‐spectrum β‐Lactamase‐producing Enterobacteriaceae: results from the INCREMENT cohort. Clin Infect Dis. 2017;65(10):1615–1623. 10.1093/CID/CIX606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Vidal E, Cervera C, Cordero E, et al. Executive summary. Management of urinary tract infection in solid organ transplant recipients: Consensus statement of the Group for the Study of Infection in Transplant Recipients (GESITRA) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) and the Spanish Network for Research in Infectious Diseases (REIPI). Enferm Infecc Microbiol Clin. 2015;33(10):680–687. 10.1016/J.EIMC.2015.03.020 [DOI] [PubMed] [Google Scholar]
  • 86. Severe nephrotoxicity caused by the combined use of gentamicin and cyclosporine in renal allograft recipients. https://pubmed.ncbi.nlm.nih.gov/3526659/. Accessed January 21, 2022. [PubMed]
  • 87. Magiorakos AP, Srinivasan A, Carey RB, et al. Multidrug‐resistant, extensively drug‐resistant and pandrug‐resistant bacteria: an international expert proposal for interim standard definitions for acquired resistance. Clin Microbiol Infect. 2012;18(3):268–281. 10.1111/J.1469-0691.2011.03570.X [DOI] [PubMed] [Google Scholar]
  • 88. Pfaller MA, Huband MD, Streit JM, Flamm RK, Sader HS. Surveillance of tigecycline activity tested against clinical isolates from a global (North America, Europe, Latin America and Asia‐Pacific) collection (2016). Int J Antimicrob Agents. 2018;51(6):848–853. 10.1016/J.IJANTIMICAG.2018.01.006 [DOI] [PubMed] [Google Scholar]
  • 89. Hernández‐García M, Pérez‐Viso B, Carmen Turrientes M, et al. Characterization of carbapenemase‐producing Enterobacteriaceae from colonized patients in a university hospital in Madrid, Spain, during the R‐GNOSIS project depicts increased clonal diversity over time with maintenance of high‐risk clones. J Antimicrob Chemother. 2018;73(11):3039–3043. 10.1093/JAC/DKY284 [DOI] [PubMed] [Google Scholar]
  • 90. Sader HS, Flamm RK, Jones RN. Tigecycline activity tested against antimicrobial resistant surveillance subsets of clinical bacteria collected worldwide. Diagn Microbiol Infect Dis. 2013;76(2):217–221. 10.1016/J.DIAGMICROBIO.2013.02.009 [DOI] [PubMed] [Google Scholar]
  • 91. Wang Q, Wang X, Wang J, et al. Phenotypic and genotypic characterization of carbapenem‐resistant enterobacteriaceae: data from a longitudinal large‐scale CRE study in China (2012–2016). Clin Infect Dis. 2018;67(suppl_2):S196–S205. 10.1093/CID/CIY660 [DOI] [PubMed] [Google Scholar]
  • 92. Vasoo S, Cunningham SA, Cole NC, et al. In vitro activities of ceftazidime‐avibactam, aztreonam‐avibactam, and a panel of older and contemporary antimicrobial agents against carbapenemase‐producing gram‐negative Bacilli. Antimicrob Agents Chemother. 2015;59(12):7842–7846. 10.1128/AAC.02019-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Heizmann WR, Löschmann PA, Eckmann C, von Eiff C, Bodmann KF, Petrik C. Clinical efficacy of tigecycline used as monotherapy or in combination regimens for complicated infections with documented involvement of multiresistant bacteria. Infection. 2015;43(1):37–43. 10.1007/S15010-014-0691-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Poulakou G, Kontopidou F V., Paramythiotou E, et al. Tigecycline in the treatment of infections from multi‐drug resistant gram‐negative pathogens. J Infect. 2009;58(4):273–284. 10.1016/J.JINF.2009.02.009 [DOI] [PubMed] [Google Scholar]
  • 95. Ni W, Han Y, Liu J, et al. Tigecycline treatment for carbapenem‐resistant enterobacteriaceae infections: a systematic review and meta‐analysis. Medicine. 2016;95(11):e3126. 10.1097/MD.0000000000003126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Tumbarello M, Sanguinetti M, Montuori E, et al. Predictors of mortality in patients with bloodstream infections caused by extended‐spectrum‐beta‐lactamase‐producing Enterobacteriaceae: importance of inadequate initial antimicrobial treatment. Antimicrob Agents Chemother. 2007;51(6):1987–1994. 10.1128/AAC.01509-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Cai Y, Wang R, Liang B, Bai N, Liu Y. Systematic review and meta‐analysis of the effectiveness and safety of tigecycline for treatment of infectious disease. Antimicrob Agents Chemother. 2011;55(3):1162–1172. 10.1128/AAC.01402-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Yahav D, Lador A, Paul M, Leibovici L. Efficacy and safety of tigecycline: a systematic review and meta‐analysis. J Antimicrob Chemother. 2011;66(9):1963–1971. 10.1093/JAC/DKR242 [DOI] [PubMed] [Google Scholar]
  • 99. Yang M, Gao H, Wang X, Qian S. Tigecycline treatment in a liver transplant infant with carbapenem‐resistant Escherichia coli infection: case report. Medicine. 2019;98(39):e17339. 10.1097/MD.0000000000017339 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Bergamasco MD, Barroso Barbosa M, de Oliveira Garcia D, et al. Infection with Klebsiella pneumoniae carbapenemase (KPC)‐producing Klebsiella pneumoniae in solid organ transplantation. Transpl Infect Dis. 2012;14(2):198–205. 10.1111/J.1399-3062.2011.00688.X [DOI] [PubMed] [Google Scholar]
  • 101. Chen F, Shen C, Pang X, et al. Effectiveness of tigecycline in the treatment of infections caused by carbapenem‐resistant gram‐negative bacteria in pediatric liver transplant recipients: a retrospective study. Transpl Infect Dis. 2020;22(1):e13199. 10.1111/TID.13199 [DOI] [PubMed] [Google Scholar]
  • 102. Mouloudi E, Massa E, Piperidou M, et al. Tigecycline for treatment of carbapenem‐resistant Klebsiella pneumoniae infections after liver transplantation in the intensive care unit: a 3‐year study. Transplant Proc. 2014;46(9):3219–3221. 10.1016/J.TRANSPROCEED.2014.09.160 [DOI] [PubMed] [Google Scholar]
  • 103. Liebenstein T, Schulz LT, Viesselmann C, et al. Effectiveness and safety of tigecycline compared with other broad‐spectrum antimicrobials in abdominal solid organ transplant recipients with polymicrobial intraabdominal infections. Pharmacotherapy. 2017;37(2):151–158. 10.1002/PHAR.1883 [DOI] [PubMed] [Google Scholar]
  • 104. Lin J, Wang R, Chen J. Tigecycline‐induced acute pancreatitis in a renal transplant patient: a case report and literature review. BMC Infect Dis. 2018;18(1):201. 10.1186/S12879-018-3103-Z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Yazirli B, Kara E, Inkaya AC, et al. A case report of tigecycline induced acute pancreatitis in a renal transplant patient and review of the literature: Should we avoid tigecycline in patients on calcineurin inhibitors? Transpl Infect Dis. 2021;23(4):e13593. 10.1111/TID.13593 [DOI] [PubMed] [Google Scholar]
  • 106. Chow KM, Pang WF, Chan GCK, Leung CB, Szeto CC, Li PKT. Beware of drug interaction between tigecycline and tacrolimus. Nephrology. 2020;25(1):99–100. 10.1111/NEP.13594 [DOI] [PubMed] [Google Scholar]
  • 107. Falagas ME, Vouloumanou EK, Samonis G, Vardakasa KZ. Fosfomycin. Clin Microbiol Rev. 2016;29(2):321–347. 10.1128/CMR.00068-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Maraki S, Samonis G, Rafailidis PI, Vouloumanou EK, Mavromanolakis E, Falagas ME. Susceptibility of urinary tract bacteria to fosfomycin. Antimicrob Agents Chemother. 2009;53(10):4508–4510. 10.1128/AAC.00721-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Bassetti M, Graziano E, Berruti M, Giacobbe DR. The role of fosfomycin for multidrug‐resistant gram‐negative infections. Curr Opin Infect Dis. 2019;32(6):617–625. 10.1097/QCO.0000000000000597 [DOI] [PubMed] [Google Scholar]
  • 110. Zarakolu P, Eser ÖK, Otlu B, et al. In‐vitro activity of fosfomycin against Escherichia coli and Klebsiella pneumoniae bloodstream isolates and frequency of OXA‐48, NDM, KPC, VIM, IMP types of carbapenemases in the carbapenem‐resistant groups. J Chemother. Published online September 8, 2021. 10.1080/1120009X.2021.1963618 [DOI] [PubMed] [Google Scholar]
  • 111. Wang H, Min C, Li J, et al. Characterization of fosfomycin resistance and molecular epidemiology among carbapenem‐resistant Klebsiella pneumoniae strains from two tertiary hospitals in China. BMC Microbiol. 2021;21(1):1221. 10.1186/S12866-021-02165-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Mueller L, Cimen C, Poirel L, Descombes MC, Nordmann P. Prevalence of fosfomycin resistance among ESBL‐producing Escherichia coli isolates in the community, Switzerland. Eur J Clin Microbiol Infect Dis. 2019;38(5):945–949. 10.1007/S10096-019-03531-0 [DOI] [PubMed] [Google Scholar]
  • 113. Michalopoulos AS, Livaditis IG, Gougoutas V. The revival of fosfomycin. Int J Infect Dis. 2011;15(11):e732–e739. 10.1016/J.IJID.2011.07.007 [DOI] [PubMed] [Google Scholar]
  • 114. López‐Medrano F, Silva JT, Fernández‐Ruiz M, et al. Oral fosfomycin for the treatment of lower urinary tract infections among kidney transplant recipients – results of a Spanish multicenter cohort. Am J Transplant. 2020;20(2):451–462. 10.1111/AJT.15614 [DOI] [PubMed] [Google Scholar]
  • 115. Ruiz‐Ruigómez M, Fernández‐Ruiz M, Silva JT, et al. Efficacy and safety of oral fosfomycin for asymptomatic bacteriuria in kidney transplant recipients: Results from a Spanish multicenter cohort. Antimicrob Agents Chemother. 2021;65(5):e02267‐20. 10.1128/AAC.02267-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Rosado‐Canto R, Parra‐Avila I, Tejeda‐Maldonado J, et al. Perioperative fosfomycin disodium prophylaxis against urinary tract infection in renal transplant recipients: a randomized clinical trial. Nephrol Dial Transplant. 2020;35(11):1996–2003. 10.1093/NDT/GFZ261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kaye KS, Rice LB, Dane AL, et al. Fosfomycin for injection (ZTI‐01) versus piperacillin‐tazobactam for the treatment of complicated urinary tract infection including acute pyelonephritis: ZEUS, a phase 2/3 randomized trial. Clin Infect Dis. 2019;69(12):2045–2056. 10.1093/CID/CIZ181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Sojo‐Dorado J, López‐Hernández I, Rosso‐Fernandez C, et al. Effectiveness of fosfomycin for the treatment of multidrug‐resistant Escherichia coli bacteremic urinary tract infections: a randomized clinical trial. JAMA Netw open. 2022;5(1):e2137277. 10.1001/JAMANETWORKOPEN.2021.37277 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Ong'uti S, Czech M, Robilotti E, Holubar M. Cefiderocol: a new cephalosporin stratagem against multidrug resistant gram‐negative bacteria. Clin Infect Dis. 2022;74(7):1303–1312. 10.1093/CID/CIAB757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Yao J, Wang J, Chen M, Cai Y. Cefiderocol: an overview of its in‐vitro and in‐vivo activity and underlying resistant mechanisms. Front Med. 2021;8:741940. 10.3389/FMED.2021.741940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Ito A, Nishikawa T, Ota M, et al. Stability and low induction propensity of cefiderocol against chromosomal AmpC β‐lactamases of Pseudomonas aeruginosa and Enterobacter cloacae . J Antimicrob Chemother. 2018;73(11):3049–3052. 10.1093/JAC/DKY317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Bassetti M, Echols R, Matsunaga Y, 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(2):226–240. 10.1016/S1473-3099(20)30796-9 [DOI] [PubMed] [Google Scholar]
  • 123. Wunderink RG, Matsunaga Y, Ariyasu M, 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(2):213–225. 10.1016/S1473-3099(20)30731-3 [DOI] [PubMed] [Google Scholar]
  • 124. Portsmouth S, van Veenhuyzen D, Echols R, et al. Cefiderocol versus imipenem‐cilastatin for the treatment of complicated urinary tract infections caused by gram‐negative uropathogens: a phase 2, randomised, double‐blind, non‐inferiority trial. Lancet Infect Dis. 2018;18(12):1319–1328. 10.1016/S1473-3099(18)30554-1 [DOI] [PubMed] [Google Scholar]
  • 125. Contreras DA, Fitzwater SP, Nanayakkara DD, et al. Coinfections of two strains of NDM‐1‐ and OXA‐232‐coproducing Klebsiella pneumoniae in a kidney transplant patient. Antimicrob Agents Chemother. 2020;64(4):e00948‐19. 10.1128/AAC.00948-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Bavaro DF, Belati A, Diella L, et al. Cefiderocol‐based combination therapy for ‘Difficult‐to‐Treat’ gram‐negative severe infections: real‐life case series and future perspectives. Antibiotics. 2021;10(6):652. 10.3390/ANTIBIOTICS10060652 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Molina KC, Bajrovic V, Bonomo RA. Carbapenemase‐producing Enterobacterales in solid organ transplantation: tip of the iceberg? Transpl Infect Dis. 2022;24(2):e13780. 10.1111/TID.13780 [DOI] [PubMed] [Google Scholar]
  • 128. Klein S, Boutin S, Kocer K, et al. Rapid development of cefiderocol resistance in carbapenem‐resistant Enterobacter cloacae during therapy is associated with heterogeneous mutations in the catecholate siderophore receptor cira. Clin Infect Dis. 2022;74(5):905–908. 10.1093/CID/CIAB511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Yahav D, Giske CG, Gramatniece A, Abodakpi H, Tam VH, Leibovici L. New β‐Lactam‐β‐Lactamase inhibitor combinations. Clin Microbiol Rev. 2020;34(1):1–61. 10.1128/CMR.00115-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Lee YR, Baker NT. Meropenem‐vaborbactam: a carbapenem and beta‐lactamase inhibitor with activity against carbapenem‐resistant Enterobacteriaceae. Eur J Clin Microbiol Infect Dis. 2018;37(8):1411–1419. 10.1007/S10096-018-3260-4 [DOI] [PubMed] [Google Scholar]
  • 131. Heo YA. Imipenem/cilastatin/relebactam: a review in gram‐negative bacterial infections. Drugs. 2021;81(3):377–388. 10.1007/S40265-021-01471-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Kiratisin P, Kazmierczak K, Stone GG. In vitro activity of ceftazidime/avibactam and comparators against carbapenemase‐producing Enterobacterales and Pseudomonas aeruginosa isolates collected globally between 2016 and 2018. J Glob Antimicrob Resist. 2021;27:132–141. 10.1016/J.JGAR.2021.08.010 [DOI] [PubMed] [Google Scholar]
  • 133. Shields RK, Potoski BA, Haidar G, et al. Clinical outcomes, drug toxicity, and emergence of ceftazidime‐avibactam resistance among patients treated for carbapenem‐resistant enterobacteriaceae infections. Clin Infect Dis. 2016;63(12):1615–1618. 10.1093/cid/ciw636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Gaibani P, Campoli C, Lewis RE, et al. In vivo evolution of resistant subpopulations of KPC‐producing Klebsiella pneumoniae during ceftazidime/avibactam treatment. J Antimicrob Chemother. 2018;73(6):1525–1529. 10.1093/JAC/DKY082 [DOI] [PubMed] [Google Scholar]
  • 135. Temkin E, Torre‐Cisneros J, Beovic B, et al. Ceftazidime‐avibactam as salvage therapy for infections caused by carbapenem‐resistant organisms. Antimicrob Agents Chemother. 2017;61(2):e01964‐16. 10.1128/AAC.01964-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Noval M, Banoub M, Claeys KC, Heil E. The battle is on: new Beta‐Lactams for the treatment of multidrug‐resistant gram‐negative organisms. Curr Infect Dis Rep. 2020;22(1):1. 10.1007/S11908-020-0710-9 [DOI] [PubMed] [Google Scholar]
  • 137. Castanheira M, Doyle TB, Kantro V, Mendes RE, Shortridge D. Meropenem‐vaborbactam activity against carbapenem‐resistant enterobacterales isolates collected in U.S. hospitals during 2016 to 2018. Antimicrob Agents Chemother. 2020;64(2):e01951‐19. 10.1128/AAC.01951-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. García‐Castillo M, García‐Fernández S, Gómez‐Gil R, et al. Activity of ceftazidime‐avibactam against carbapenemase‐producing Enterobacteriaceae from urine specimens obtained during the infection‐carbapenem resistance evaluation surveillance trial (iCREST) in Spain. Int J Antimicrob Agents. 2018;51(3):511–515. 10.1016/J.IJANTIMICAG.2018.01.011 [DOI] [PubMed] [Google Scholar]
  • 139. Rivera‐izquierdo M, Láinez‐ramos‐bossini AJ, Rivera‐izquierdo C, et al. OXA‐48 carbapenemase‐producing enterobacterales in Spanish hospitals: an updated comprehensive review on a rising antimicrobial resistance. Antibiotics. 2021;10(1):1–16. 10.3390/ANTIBIOTICS10010089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Pogue JM, Bonomo RA, Kaye KS. Ceftazidime/avibactam, meropenem/vaborbactam, or both? clinical and formulary considerations. Clin Infect Dis. 2019;68(3):519–524. 10.1093/CID/CIY576 [DOI] [PubMed] [Google Scholar]
  • 141. Ackley R, Roshdy D, Meredith J, et al. Meropenem‐vaborbactam versus ceftazidime‐avibactam for treatment of carbapenem‐resistant enterobacteriaceae infections. Antimicrob Agents Chemother. 2020;64(5):e02313–e02319. 10.1128/AAC.02313-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Papp‐Wallace KM, Barnes MD, Alsop J, et al. Relebactam is a potent inhibitor of the KPC‐2 β‐Lactamase and restores imipenem susceptibility in KPC‐producing Enterobacteriaceae. Antimicrob Agents Chemother. 2018;62(6):e00174‐18. 10.1128/AAC.00174-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Lob SH, Karlowsky JA, Young K, et al. In vitro activity of imipenem‐relebactam against resistant phenotypes of Enterobacteriaceae and Pseudomonas aeruginosa isolated from intraabdominal and urinary tract infection samples ‐ SMART Surveillance Europe 2015‐2017. J Med Microbiol. 2020;69(2):207–217. 10.1099/JMM.0.001142 [DOI] [PubMed] [Google Scholar]
  • 144. Karlowsky JA, Lob SH, Kazmierczak KM, Young K, Motyl MR, Sahm DF. In vitro activity of imipenem/relebactam against Enterobacteriaceae and Pseudomonas aeruginosa isolated from intraabdominal and urinary tract infection samples: SMART surveillance United States 2015‐2017. J Glob Antimicrob Resist. 2020;21:223–228. 10.1016/J.JGAR.2019.10.028 [DOI] [PubMed] [Google Scholar]
  • 145. Athans V, Neuner EA, Hassouna H, et al. Meropenem‐vaborbactam as salvage therapy for ceftazidime‐avibactam‐resistant Klebsiella pneumoniae bacteremia and abscess in a liver transplant recipient. Antimicrob Agents Chemother. 2018;63(1):e01551‐18. 10.1128/AAC.01551-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Caravaca‐Fontán F, Jiménez‐Álvaro S, Marcén‐Letosa R, Fernández‐Rodríguez A, Rodríguez‐Navarro CQ. Ceftazidime‐avibactam in urinary tract infections due to carbapenemase‐producing Klebsiella in kidney transplantation. Nefrologia. 2015;35(4):412–413. 10.1016/J.NEFRO.2015.01.001 [DOI] [PubMed] [Google Scholar]
  • 147. Wang Z, Ma K, Chen Z, et al. Successful treatment of early post‐transplant bloodstream and pulmonary infection caused by carbapenem‐resistant Klebsiella pneumoniae with a combination of ceftazidime‐avibactam and carbapenem: a case report. Transplant Proc. 2020;52(9):2742–2746. 10.1016/J.TRANSPROCEED.2020.08.006 [DOI] [PubMed] [Google Scholar]
  • 148. Cani E, Moussavi F, Ocheretyaner E, Sharma R, Brown C, Eilertson B. Carbapenem‐resistant Klebsiella pneumoniae vertebral osteomyelitis in a renal transplant recipient treated with ceftazidime‐avibactam. Transpl Infect Dis. 2018;20(2):e12837. 10.1111/TID.12837 [DOI] [PubMed] [Google Scholar]
  • 149. Chen W, Sun L, Guo L, et al. Clinical outcomes of ceftazidime‐avibactam in lung transplant recipients with infections caused by extensively drug‐resistant gram‐negative bacilli. Ann Transl Med. 2020;8(3):39–39. 10.21037/ATM.2019.10.40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Amore D, Pecoraro Y, Carillo C, et al. Use of ceftazidime‐avibactam and ceftolozane‐tazobactam after lung transplantation. Transplant Proc. 2020;52(5):1605–1607. 10.1016/J.TRANSPROCEED.2020.02.051 [DOI] [PubMed] [Google Scholar]
  • 151. Bilinskaya A, Linder KE, Kuti JL. Plazomicin: an intravenous aminoglycoside antibacterial for the treatment of complicated urinary tract infections. Expert Rev Anti Infect Ther. 2020;18(8):705–720. 10.1080/14787210.2020.1759419 [DOI] [PubMed] [Google Scholar]
  • 152. Johnston BD, Thuras P, Porter SB, et al. Activity of plazomicin against carbapenem‐intermediate or ‐resistant Escherichia coli isolates from the United States and international sites in relation to clonal background, resistance genes, co‐resistance, and region. J Antimicrob Chemother. 2021;76(8):2061–2070. 10.1093/JAC/DKAB150 [DOI] [PubMed] [Google Scholar]
  • 153. Serio AW, Keepers T, Krause KM. Plazomicin is active against metallo‐β‐lactamase‐producing Enterobacteriaceae. Open forum Infect Dis. 2019;6(4):ofz123. 10.1093/OFID/OFZ123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Wagenlehner FME, Cloutier DJ, Komirenko AS, et al. Re: once‐daily plazomicin for complicated urinary tract infections. J Urol. 2019;202(4):641–642. 10.1056/NEJMoa1801467 [DOI] [PubMed] [Google Scholar]
  • 155. McKinnell JA, Dwyer JP, Talbot GH, et al. Plazomicin for infections caused by carbapenem‐resistant Enterobacteriaceae. N Engl J Med. 2019;380(8):791–793. 10.1056/NEJMC1807634 [DOI] [PubMed] [Google Scholar]
  • 156. Trang M, Seroogy JD, Van Wart SA, et al. Population pharmacokinetic analyses for plazomicin using pooled data from phase 1, 2, and 3 clinical studies. Antimicrob Agents Chemother. 2019;63(4):e02329‐18. 10.1128/AAC.02329-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Zhanel GG, Cheung D, Adam H, et al. Review of eravacycline, a novel fluorocycline antibacterial agent. Drugs. 2016;76(5):567–588. 10.1007/S40265-016-0545-8 [DOI] [PubMed] [Google Scholar]
  • 158. Johnston BD, Thuras P, Porter SB, et al. Activity of cefiderocol, ceftazidime‐avibactam, and eravacycline against carbapenem‐resistant Escherichia coli isolates from the United States and international sites in relation to clonal background, resistance genes, coresistance, and region. Antimicrob Agents Chemother. 2020;64(10):e00797‐20. 10.1128/AAC.00797-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Morrissey I, Olesky M, Hawser S, et al. In vitro activity of eravacycline against gram‐negative Bacilli isolated in clinical laboratories worldwide from 2013 to 2017. Antimicrob Agents Chemother. 2020;64(3):e01699‐19. 10.1128/AAC.01699-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Lee YL, Ko WC, Sen Lee W, et al. In‐vitro activity of cefiderocol, cefepime/zidebactam, cefepime/enmetazobactam, omadacycline, eravacycline and other comparative agents against carbapenem‐nonsusceptible Enterobacterales: results from the Surveillance of Multicenter Antimicrobial Resistance in Taiwan (SMART) in 2017‐2020. Int J Antimicrob Agents. 2021;58(3):106377. 10.1016/J.IJANTIMICAG.2021.106377 [DOI] [PubMed] [Google Scholar]
  • 161. Chatzidimitriou M, Chatzivasileiou P, Sakellariou G, et al. Ceftazidime/avibactam and eravacycline susceptibility of carbapenem‐resistant Klebsiella pneumoniae in two Greek tertiary teaching hospitals. Acta Microbiol Immunol Hung. 2021;68(2):65–72. 10.1556/030.2021.01364 [DOI] [PubMed] [Google Scholar]
  • 162. Lutgring JD, Balbuena R, Reese N, et al. Antibiotic Susceptibility of NDM‐producing Enterobacterales collected in the United States in 2017 and 2018. Antimicrob Agents Chemother. 2020;64(9). 10.1128/AAC.00499-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Livermore DM, Mushtaq BS, Warner AM, Woodforda AN. In vitro activity of eravacycline against carbapenem‐resistant Enterobacteriaceae and Acinetobacter baumannii . Antimicrob Agents Chemother. 2016;60(6):3840–3844. 10.1128/AAC.00436-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Solomkin J, Evans D, Slepavicius A, et al. Assessing the efficacy and safety of eravacycline vs ertapenem in complicated intra‐abdominal infections in the investigating gram‐negative infections treated with eravacycline (IGNITE 1) trial: a randomized clinical trial. JAMA Surg. 2017;152(3):224–232. 10.1001/JAMASURG.2016.4237 [DOI] [PubMed] [Google Scholar]
  • 165. Solomkin JS, Gardovskis J, Lawrence K, et al. IGNITE4: results of a phase 3, randomized, multicenter, prospective trial of eravacycline vs meropenem in the treatment of complicated intraabdominal infections. Clin Infect Dis. 2019;69(6):921–929. 10.1093/CID/CIY1029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Hobbs ALV, Gelfand MS, Cleveland KO, Saddler K, Sierra‐Hoffman MA. A retrospective multicenter evaluation of eravacycline utilization in community and academic hospitals. J Glob Antimicrob Resist. Published online November 14, 2021. 10.1016/J.JGAR.2021.10.020 [DOI] [PubMed] [Google Scholar]
  • 167. Di Felice VG, Efimova E, Izmailyan S, Napolitano LM, Chopra T. Efficacy and Tolerability of Eravacycline in Bacteremic Patients with Complicated Intra‐Abdominal Infection: A Pooled Analysis from the IGNITE1 and IGNITE4 Studies. Surg Infect. 2021;22(5):556–561. 10.1089/SUR.2020.241 [DOI] [PubMed] [Google Scholar]

Articles from Transplant Infectious Disease are provided here courtesy of Wiley

RESOURCES