Next Article in Journal
Gender and Neurosteroids: Implications for Brain Function, Neuroplasticity and Rehabilitation
Previous Article in Journal
Cardiac Roles of Serotonin (5-HT) and 5-HT-Receptors in Health and Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Ceftazidime-Avibactam (C/A) Resistant, Meropenem Sensitive KPC-Producing Klebsiella pneumoniae in ICU Setting: We Are What We Are Treated with?

1
Department of Medical Sciences, Infectious Diseases, University of Turin, 10124 Turin, Italy
2
School of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
3
Department of Anesthesia, Intensive Care and Emergency—‘Città della Salute e della Scienza’ Hospital, 10126 Turin, Italy
4
Infectious Diseases Unit, Cardinal Massaia Hospital, 14100 Asti, Italy
5
Microbiology and Virology Unit, University Hospital Città della Salute e della Scienza di Torino, 10126 Turin, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(5), 4767; https://doi.org/10.3390/ijms24054767
Submission received: 31 December 2022 / Revised: 20 February 2023 / Accepted: 24 February 2023 / Published: 1 March 2023
(This article belongs to the Section Molecular Microbiology)

Abstract

:
The continuous spread of carbapenem-resistant Klebsiella pneumoniae (CP-Kp) strains presents a severe challenge to the healthcare system due to limited therapeutic options and high mortality. Since its availability, ceftazidime/avibactam (C/A) has become a first-line option against KPC-Kp, but C/A-resistant strains have been reported increasingly, especially with pneumonia or prior suboptimal blood exposure to C/A treatment. A retrospective, observational study was conducted with all patients admitted to the Intensive Care Unit (ICU) dedicated to COVID-19 patients at the City of Health & Sciences in Turin, between 1 May 2021 and 31 January 2022, with the primary endpoint to study strains with resistance to C/A, and secondly to describe the characteristics of this population, with or without previous exposure to C/A. Seventeen patients with colonization or invasive infection due to Klebsiella pneumoniae, C/A resistance, and susceptibility to meropenem (MIC = 2 µg/L) were included; the blaKPC genotype was detected in all isolates revealing D179Y mutation in the blaKPC-2 (blaKPC-33) gene. Cluster analysis showed that 16 out of the 17 C/A-resistant KPC-Kp isolates belonged to a single clone. Thirteen strains (76.5%) were isolated in a 60-day period. Only some patients had a previous infection with non-mutant KPC at other sites (5; 29.4%). Eight patients (47.1%) underwent previous large-spectrum antibiotic treatment, and four patients (23.5%) had prior treatment with C/A. The secondary spread of the D179Y mutation in the blaKPC-2 during the COVID-19 pandemic needs to be addressed constantly by an interdisciplinary interaction between microbiologists, infection control personnel, clinicians, and infectious diseases consultants to properly diagnose and treat patients.

1. Introductions

The continuous spread of carbapenemase-producing Klebsiella pneumoniae (CP-Kp) strains presents a severe challenge to healthcare systems because of the limited therapeutic options, high mortality, and allocated costs worldwide [1,2]. In the early 2000s, CP-Kp was first identified in Europe, and its prevalence has since increased [3]. Current European epidemiology ranges from sporadic imported cases and hospital outbreaks to interregional spread and endemic CP-Kp in healthcare settings [4]. Among carbapenemase producers, Enterobacterales, KPC-producing K. pneumoniae (KPC-Kp) is the most common in Italy [5,6,7,8,9]. Other carbapenemases, namely metallo beta-lactamase (MBL), New Delhi metallo beta-lactamase (NDM), and Verona integron-encoded metallo-β-lactamase (VIM), have also been found in recent years [10,11]. Mobile genetic elements (MGEs), including plasmids, transposable elements, and integrons, are perhaps the most vital factors associated with spreading carbapenemase genes among various bacterial species [12]. The global dissemination of KPC-Kp has been linked to the successful spread of a specific genetic line, designated clonal group 258 (CG258). The KPC-coding gene, blaKPC, is generally found within a Tn4401 transposon, a mobile genetic element originating from the Tn3 transposon family which aids its dissemination [13,14].
Ceftazidime/avibactam (C/A) is a new β-lactam/β-lactamase inhibitor combination that has been available since 2015, and in Italy, since February 2018. Interestingly, avibactam is structurally distinct from other β-lactamase inhibitors used in clinical practice as it lacks a β-lactam core [15]. The mechanism of “reversible” enzymatic inhibition involves the opening of the avibactam ring, which does not include a hydrolysis but consists only in a deacylation [16]. Increasingly over recent years, C/A has become a first-line option against KPC-producing Enterobacterales, especially in critically ill patients with high INCREMENT-CPE scores within endemic settings, and it has been associated with significantly reduced mortality [17]. Unfortunately, episodes of colonization or infection due to C/A-resistant strains have also been reported in the literature with or without prior C/A treatment in primary and secondary resistance features [18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40]. Resistance to C/A is typically caused by the presence of MBLs, as avibactam does not restore their activity. Other mechanisms are the increased expression of mutated blaKPC or other β-lactamase genes, resulting in increased copy number; modification in cell permeability, such as loss of porins; and overexpression of efflux pumps or pyruvate dehydrogenase complex enzyme variants [18,19].
Previous studies showed that the development of resistance often emerged during prolonged C/A treatment [18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40]. In addition, comorbidities, pneumonia, inappropriate antimicrobial plasma concentrations, critical illnesses, and exposure to other colonized patients due to infection control issues may represent other risk factors for resistance [21,22,23,24,25,26]. To note, data regarding optimal dosing regimens for C/A in critically ill patients are scarce, but emerging real-life data suggest that more aggressive pharmacokinetic/pharmacodynamic (PK/PD) targets might improve clinical efficacy and suppress resistance emergence [24,25,26]. As an example, Gaibani et al. [26] described how the development of resistance due to mutations in the blaKPC gene in KPC-Kp strains during C/A treatment was due to initial suboptimal blood exposure to the antimicrobials that lead a selection of hybrid subpopulations.
In this report, we aimed to describe the clinical features, microbiological characteristics, and outcomes of critically ill patients admitted to the intensive care unit (ICU) with a nosocomial spread of meropenem sensitive, C/A resistant, KPC-Kp strains.

2. Results

The study population consisted of 17 patients who developed a colonization or an invasive infection due to K. pneumoniae resistant to C/A (MIC > 256 mg/L). The 17 C/A-resistant K. pneumoniae isolates had the same antimicrobial susceptibility pattern (Table 1), were carbapenem susceptible (meropenem MIC 2 mg/L, imipenem ≤ 1 mg/L, ertapenem 1 mg/L), expressing an ESBL phenotype. All phenotypic carbapenemase detection methods tested negative and blaKPC was detected in all isolates by Xpert® Carba-R. Sequencing of the blaKPC gene revealed D179Y mutation in the blaKPC-2 (blaKPC-33). Cluster analysis using Fourier-transform infrared spectroscopy showed that 16 out of the 17 C/A-resistant KPC-Kp isolates belonged to a single outbreak clone [27]. Most of the isolates overlap in the same period of admission; thirteen patients (76.4%) developed a colonization or infection in the specific time frame between December 2021 to January 2022 (Figure 1).
Half of the patients were male (9; 52.9%) and the median age was 57 years (IQR 47.5–70). Most of the patients were admitted for acute respiratory distress syndrome (ARDS) in COVID-19 (41.1%) or SARS-CoV-2 pneumonia (23.5%) (Table 2). Most cases of mutant KPC-Kp were isolated in rectal swabs (15; 88.2%) or from the respiratory tract (5; 29.4%) (Table 2). Five patients were also previously infected by C/A susceptible KPC-producing K. pneumoniae at other sites (29.4%). The median time from admission to C/A-resistant KPC-producing K. pneumoniae detection was 13 days (IQR 7–34). In addition, co-infection or concomitant superinfection was not uncommon and mostly due to bacteria (i.e., A. baumannii, 17.6%), viruses (i.e., SARS-CoV-2, 64.7%), or fungi (i.e., Aspergillus, 17.6%). Overall, 10 patients (58.8%) died during hospital admission.
Eight patients (47.0%) underwent large-spectrum antibiotic treatment before mutant-KPC-Kp isolation, and only four patients (23.5%) underwent prior treatment with C/A as detailed in Table 1. All patients previously treated with C/A had received combination therapy with fosfomycin. Following the C/A resistant isolate, 7 (41.2%) patients were treated with meropenem/vaborbactam, of which three (42.9%) were for suspected VAP.

3. Discussion

In this study we retrospectively reported on a cohort of seventeen critically ill patients enrolled over nine months during the COVID-19 pandemic, all of whom developed intra-hospital colonization or infection due to mutant-KPC-33 Kp with resistance to C/A, mostly without previous C/A treatment. Although relatively recently introduced, C/A use may select in vivo specific KPC-producing Enterobacterales resistant in patients with prolonged treatment alone or in combination [28,29,30]. Only some of these C/A-resistant strains are reported to harbor new KPC variants exhibiting single amino acid substitutions—one being Asp179Tyr (D179Y), which is particularly common and characterized by a loss of carbapenemase activity and a restoration of carbapenem susceptibility, together with a concomitant reduction of binding to avibactam [28,29]. Different types of variants encoding mutations associated with C/A resistance (e.g., KPC-41, KPC-23, KPC-14, KPC-8, and KPC-50) have been isolated in patients, with or without a history of C/A treatment [28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43]. In Italy, C/A has been available since February 2018, and the emergence of C/A-resistant Enterobacterales strains has been reported since 2020 [40,41,42,43].
Our data confirm that isolates harboring KPCD179Y are undetectable by the main phenotypic carbapenemase detection methods, including immunochromatographic assays [37,38,42]. Since phenotypic detection methods represent the most widespread and cost-saving means of detecting carbapenemases in clinical microbiology laboratories, failure to recognize these mutated-KPC-producing isolates as alert microorganisms could easily facilitate their spread in healthcare facilities. In healthcare settings with a high circulation of KPC mutants, genotypic carbapenemase detection methods should be preferred and recommended together with antimicrobial susceptibility testing for C/A.
According to the phenotypical microbiological characteristics of our cluster of mutant-KPC-Kp, the susceptibility to carbapenems was retained; this was also reported in other case series [28,29]. Interestingly, Shields et al. [29] showed a specific blaKPC, the KPC-3 variant, as a result of plasmid transfer which determines an increase in C/A MICs and a temporary reduction in meropenem MICs ≥ 4-fold, restoring carbapenem-susceptibility in KPC-Kp. The microbiological data and predicted kinetic in humans suggest that infections by C/A-resistant and carbapenem-susceptible KPC-Kp could be treated with carbapenems. However, in a clinical setting, the effectiveness of carbapenems alone on these infections is unclear [30]. In fact, under selective pressure with carbapenems, the MICs of these compounds can increase while the species maintain their resistance to C/A [17,29], probably justifying the use of meropenem/vaborbactam whereby C/A resistance is detected and the genotypic mechanism is not determined. In particular, Shields et al. [29] showed that in vitro sublethal concentration of meropenem in patients with C/A resistant KPC-3-variant with restored susceptibility to meropenem may select for strains with high-level resistance both to C/A and meropenem. To date, no in vivo published data are available on the effectiveness of meropenem treatment in patients with such infections. On the other hand, some authors recently highlighted the in vitro [35,36] and in vivo [19,20] activity of meropenem-vaborbactam against carbapenemase-producing Enterobacterales, including isolates resistant to C/A.
We already reported an increased multidrug-resistant (MDR) pathogens rate during the COVID-19 period [43] and we speculate that the KPC-Kp acquisition was likely healthcare associated through cross-contamination and secondary spread. This calls for greater attention to infection control measures in addition to antimicrobial stewardship suggesting that it is not only a phenomenon of disseminated resistance or inappropriate antibiotic use, since COVID-19 patients were admitted regionally and not within the same hospital. Current infection control strategies appeared effective in containing and limiting the spread of multidrug-resistant pathogens. In clinical practice, however, it is not always simple to translate into practice regional interventions capable of having a tangible impact on the risk of health-care transmission. At the same time, control measures need to be scaled up after interventions to sustain their impact in the long term. As in many other prevention contexts, probably the most correct approach includes bundled strategies. Educational strategies for health personnel might only have an impact if combined with the correct environmental hygiene, monitoring of instruments and equipment, and safe environments and spaces. An easy-to-update data collection system, in addition to weekly meetings between microbiologists and infectious diseases specialists, can guarantee the clinical surveillance of the data and their repeated analysis over time. In addition, we highlight the importance of monitoring C/A susceptibility to prevent selection of resistance and the spread of involved genetic elements. Furthermore, a program of active surveillance for C/A-resistant strains using selective media should be implemented in the case of an outbreak sustained by C/A-resistant strains.

4. Materials and Methods

A retrospective observational study was conducted including all patients admitted to the regional referral ICU due to COVID-19, or COVID-19-associated reasons, at the City of Health & Science in Turin, Italy, between May 2021 and January 2022. A retrospective review of medical charts was performed to collect information about the clinical and microbiological characteristics of patients with C/A-resistant strains.
Isolates collected by various clinical specimens (rectal swabs, urine, blood, respiratory samples, etc.) were identified by MALDI-TOF/MS (Bruker Daltonics GmbH, Bremen, Germany). Rectal swabs were performed weekly as an infection control stewardship protocol at our institution. Antimicrobial susceptibility was determined by a commercially available microdilution assay (Panel NMDR, MicroScan® WalkAway® 96 Plus; Nyon, Beckman Coulter, Switzerland), and C/A minimum inhibitory concentrations (MICs) were confirmed by Etest (bioMérieux, Paris, France). Susceptibility data were interpreted according to current European Committee on Antimicrobial Susceptibility Testing (EUCAST) breakpoints (version 11.0, 2021; https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_11.0_Breakpoint_Tables.pdf, accessed on 31 December 2022). C/A-resistant Enterobacterales isolates were investigated for carbapenemase production using three phenotypical methods (Disc Diffusion Synergy test [KPC, MBL and OXA-48 Confirm Kit, Rosco Diagnostica], modified carbapenem inactivation method [mCIM], lateral flow immunoassay [NG-Test CARBA 5- NG Biotech, France], and a commercial molecular testing (Xpert® Carba-R; Cepheid, Sunnyvale, CA, USA).
C/A resistant isolates were further subjected to cluster analysis using Fourier-transform infrared spectroscopy on IR Biotyper system (Bruker GmbH, Bremen, Germany) which was performed to assess the clonality and sequencing of the blaKPC gene, as previously described [27]. The primary endpoint was to describe the features related to the appearance of strains with resistance to C/A in samples from ICU patients, and secondly to describe the clinical and microbiological characteristics of this population.

4.1. Statistical Analysis

Data were collected in an Excel spreadsheet and analyzed using IBM SPSS Statistics Version 28.0. Continuous variables are reported as mean (standard deviation) or median (interquartile range). Categorical variables are reported as an absolute number (percentage). Demographic and clinical characteristics of the patients were summarized through absolute frequencies and percentages for the qualitative variables, and percentiles (median, first quartile-third quartile) for the quantitative variables.

4.2. Ethics

The study was approved by the Local Ethical Committee (Protocol No. 000819). Data were collected in compliance with Italian laws on privacy protection. Written consent was waived according to Italian regulation, due to the retrospective and observational nature of this study in critically ill patients. Data were pooled and collected anonymously.

5. Conclusions

In conclusion, we report the emergence of C/A-resistant, meropenem-sensitive KPC strains in a high-endemic setting for KPC-Kp, highlighting the importance of surveillance and infection controls to stop the spreading of C/A-resistant strains. Constant interdisciplinary interaction between microbiologists, infection control personnel, clinicians, and infectious disease consultants is essential to ensure new issues are shared promptly and to implement preventive strategies that, following the COVID-19 pandemic, should be more suitable for a large regional perspective rather than single hospitals.

Author Contributions

Conceptualization, S.C. and G.M.; methodology, S.C., I.D.B. and F.G.D.R.; software, G.B.; formal analysis, N.S. and I.D.B.; investigation, N.S., S.C., G.M. and T.L.; data curation, N.S. and G.T.; writing—original draft preparation, N.S. and I.D.B.; writing—review and editing, I.D.B., S.C., G.M., F.G.D.R. and G.B.; supervision, F.G.D.R., G.M., R.C. and L.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by Ethics Committee on 13 March 2020, protocol number 0027840.

Informed Consent Statement

Patient consent was waived due to the retrospective nature of the study on inconscient patients, as approved by Ethics Committee.

Data Availability Statement

Data are available under request to Authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Logan, L.K.; Weinstein, R.A. The Epidemiology of Carbapenem-Resistant Enterobacteriaceae: The Impact and Evolution of a Global Menace. J. Infect. Dis. 2017, 215, S28–S36. [Google Scholar] [CrossRef] [Green Version]
  2. Brink, A.J. Epidemiology of carbapenem-resistant Gram-negative infections globally. Curr. Opin. Infect. Dis. 2019, 32, 609–616. [Google Scholar] [CrossRef] [PubMed]
  3. Brolund, A.; Lagerqvist, N.; Byfors, S.; Struelens, M.J.; Monnet, D.L.; Albiger, B.; Kohlenberg, A.; European Antimicrobial Resistance Genes Surveillance Network (EURGen-Net) capacity survey group. Worsening epidemiological situation of carbapenemase-producing Enterobacteriaceae in Europe, assessment by national experts from 37 countries, July 2018. Eurosurveillance 2019, 24, 1900123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. WHO Regional Office for Europe/European Centre for Disease Prevention and Control. Antimicrobial Resistance Surveillance in Europe 2022–2020 Data; WHO Regional Office for Europe: Copenhagen, Denmark, 2022.
  5. Rossi, M.; Chatenoud, L.; Gona, F.; Sala, I.; Nattino, G.; D’Antonio, A.; Castelli, D.; Itri, T.; Morelli, P.; Bigoni, S.; et al. Characteristics and Clinical Implications of Carbapenemase-Producing Klebsiella pneumoniae Colonization and Infection, Italy. Emerg. Infect. Dis. 2021, 27, 1416–1426. [Google Scholar] [CrossRef]
  6. Grundmann, H.; Glasner, C.; Albiger, B.; Aanensen, D.M.; Tomlinson, C.T.; Andrasević, A.T.; Cantón, R.; Carmeli, Y.; Friedrich, A.W.; Giske, C.G.; et al. Occurrence of carbapenemase-producing Klebsiella pneumoniae and Escherichia coli in the European survey of carbapenemase-producing Enterobacteriaceae (EuSCAPE): A prospective, multinational study. Lancet Infect. Dis. 2017, 17, 153–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Tumbarello, M.; Raffaelli, F.; Giannella, M.; Mantengoli, E.; Mularoni, A.; Venditti, M.; De Rosa, F.G.; Sarmati, L.; Bassetti, M.; Brindicci, G.; et al. Ceftazidime-Avibactam Use for Klebsiella pneumoniae Carbapenemase–Producing K. pneumoniae Infections: A Retrospective Observational Multicenter Study. Clin. Infect. Dis. 2021, 73, 1664–1676. [Google Scholar] [CrossRef] [PubMed]
  8. Corcione, S.; De Benedetto, I.; Shbaklo, N.; Ranzani, F.; Pinna, S.M.; Castiglione, A.; Scabini, S.; Bianco, G.; Cavallo, R.; Mirabella, S.; et al. Ten Years of KPC-Kp Bloodstream Infections Experience: Impact of Early Appropriate Empirical Therapy on Mortality. Biomedicines 2022, 10, 3268. [Google Scholar] [CrossRef]
  9. Cano, A.; Gutiérrez-Gutiérrez, B.; Machuca, I.; Gracia-Ahufinger, I.; Pérez-Nadales, E.; Causse, M.; Castón, J.J.; Guzman-Puche, J.; Torre-Giménez, J.; Kindelán, L.; et al. Risks of Infection and Mortality Among Patients Colonized With Klebsiella pneumoniae Carbapenemase–Producing K. pneumoniae: Validation of Scores and Proposal for Management. Clin. Infect. Dis. 2018, 66, 1204–1210. [Google Scholar] [CrossRef] [Green Version]
  10. Falcone, M.; Daikos, G.L.; Tiseo, G.; Bassoulis, D.; Giordano, C.; Galfo, V.; Leonildi, A.; Tagliaferri, E.; Barnini, S.; Sani, S.; et al. Efficacy of Ceftazidime-avibactam Plus Aztreonam in Patients With Bloodstream Infections Caused by Metallo-β-lactamase–Producing Enterobacterales. Clin. Infect. Dis. 2021, 72, 1871–1878. [Google Scholar] [CrossRef]
  11. Boattini, M.; Comini, S.; Bianco, G.; Iannaccone, M.; Casale, R.; Cavallo, R.; Costa, C. Activity of cefiderocol and synergy of novel β-lactam-β-lactamase inhibitor-based combinations against metallo-β-lactamase-producing gram-negative bacilli: Insights from a two-year study (2019–2020). J. Chemother. 2022, 22, 1–7. [Google Scholar] [CrossRef] [PubMed]
  12. Reyes, J.A.; Melano, R.; Cárdenas, P.A.; Trueba, G. Mobile genetic elements associated with carbapenemase genes in South American Enterobacterales. Braz. J. Infect. Dis. 2020, 24, 231–238. [Google Scholar] [CrossRef] [PubMed]
  13. Kitchel, B.; Rasheed, J.K.; Patel, J.B.; Srinivasan, A.; Navon-Venezia, S.; Carmeli, Y.; Brolund, A.; Giske, C.G. Molecular Epidemiology of KPC-Producing Klebsiella pneumoniae Isolates in the United States: Clonal Expansion of Multilocus Sequence Type 258. Antimicrob. Agents Chemother. 2009, 53, 3365–3370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Chen, L.; Mathema, B.; Chavda, K.D.; DeLeo, F.R.; Bonomo, R.A.; Kreiswirth, B.N. Carbapenemase-producing Klebsiella pneumoniae: Molecular and genetic decoding. Trends Microbiol. 2014, 22, 686–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Tooke, C.L.; Hinchliffe, P.; Bragginton, E.C.; Colenso, C.K.; Hirvonen, V.H.A.; Takebayashi, Y.; Spencer, J. β-Lactamases and β-Lactamase Inhibitors in the 21st Century. J. Mol. Biol. 2019, 431, 3472–3500. [Google Scholar] [CrossRef]
  16. Principe, L.; Lupia, T.; Andriani, L.; Campanile, F.; Carcione, D.; Corcione, S.; De Rosa, F.G.; Luzzati, R.; Stroffolini, G.; Steyde, M.; et al. Microbiological, Clinical, and PK/PD Features of the New Anti-Gram-Negative Antibiotics: β-Lactam/β-Lactamase Inhibitors in Combination and Cefiderocol—An All-Inclusive Guide for Clinicians. Pharmaceuticals 2022, 15, 463. [Google Scholar] [CrossRef]
  17. Di Bella, S.; Giacobbe, D.R.; Maraolo, A.E.; Viaggi, V.; Luzzati, R.; Bassetti, M.; Luzzaro, F.; Principe, L. Resistance to ceftazidime/avibactam in infections and colonisations by KPC-producing Enterobacterales: A systematic review of observational clinical studies. J. Glob. Antimicrob. Resist. 2021, 25, 268–281. [Google Scholar] [CrossRef]
  18. Gaibani, P.; Campoli, C.; Lewis, R.E.; Volpe, S.L.; Scaltriti, E.; Giannella, M.; Pongolini, S.; Berlingeri, A.; Cristini, F.; Bartoletti, M.; et al. In vivo evolution of resistant subpopulations of KPC-producing Klebsiella pneumoniae during ceftazidime/avibactam treatment. J. Antimicrob. Chemother. 2018, 73, 1525–1529. [Google Scholar] [CrossRef] [Green Version]
  19. Tiseo, G.; Falcone, M.; Leonildi, A.; Giordano, C.; Barnini, S.; Arcari, G.; Carattoli, A.; Menichetti, F. Meropenem-Vaborbactam as Salvage Therapy for Ceftazidime-Avibactam-, Cefiderocol-Resistant ST-512 Klebsiella pneumoniae–Producing KPC-31, a D179Y Variant of KPC-3. Open Forum Infect. Dis. 2021, 8, ofab141. [Google Scholar] [CrossRef]
  20. Athans, V.; Neuner, E.A.; Hassouna, H.; Richter, S.S.; Keller, G.; Castanheira, M.; Brizendine, K.D.; Mathers, A.J. Meropenem-Vaborbactam as Salvage Therapy for Ceftazidime-Avibactam-Resistant Klebsiella pneumoniae Bacteremia and Abscess in a Liver Transplant Recipient. Antimicrob. Agents Chemother. 2018, 63, e01551-18. [Google Scholar] [CrossRef] [Green Version]
  21. Wang, Q.; Zhang, Y.; Yao, X.; Xian, H.; Liu, Y.; Li, H.; Chen, H.; Wang, X.; Wang, R.; Zhao, C.; et al. Risk factors and clinical outcomes for carbapenem-resistant Enterobacteriaceae nosocomial infections. Eur. J. Clin. Microbiol. Infect. Dis. 2016, 35, 1679–1689. [Google Scholar] [CrossRef]
  22. Nicolas-Chanoine, M.-H.; Vigan, M.; Laouénan, C.; Robert, J.; E-carb Study Group. Risk factors for carbapenem-resistant Enterobacteriaceae infections: A French case-control-control study. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 383–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Adembri, C.; Novelli, A.; Nobili, S. Some Suggestions from PK/PD Principles to Contain Resistance in the Clinical Setting—Focus on ICU Patients and Gram-Negative Strains. Antibiotics 2020, 9, 676. [Google Scholar] [CrossRef] [PubMed]
  24. Hites, M. Minireview on Novel Anti-infectious Treatment Options and Optimized Drug Regimens for Sepsis. Front. Med. 2021, 8, 640740. [Google Scholar] [CrossRef] [PubMed]
  25. Gaibani, P.; Gatti, M.; Rinaldi, M.; Pesce, C.C.; Lazzarotto, T.; Giannella, M.; Lombardo, D.; Amadesi, S.; Viale, P.; Pea, F.; et al. Suboptimal drug exposure leads to selection of different subpopulations of ceftazidime-avibactam-resistant Klebsiella pneumoniae carbapenemase-producing Klebsiella pneumoniae in a critically ill patient. Int. J. Infect. Dis. 2021, 113, 213–217. [Google Scholar] [CrossRef]
  26. Rakovitsky, N.; Frenk, S.; Kon, H.; Schwartz, D.; Temkin, E.; Solter, E.; Paikin, S.; Cohen, R.; Schwaber, M.J.; Carmeli, Y.; et al. Fourier Transform Infrared Spectroscopy Is a New Option for Outbreak Investigation: A Retrospective Analysis of an Extended-Spectrum-Beta-Lactamase-Producing Klebsiella pneumoniae Outbreak in a Neonatal Intensive Care Unit. J. Clin. Microbiol. 2020, 58, e00098-20. [Google Scholar] [CrossRef]
  27. Rapid Risk Assessment: Emergence of Resistance to Ceftazidime-Avibactam in Carbapenem-Resistant Enterobacteriaceae. European Centre for Disease Prevention and Control. 13 June 2018. Available online: https://www.ecdc.europa.eu/en/publications-data/rapid-risk-assessment-emergence-resistance-ceftazidime-avibactam-carbapenem (accessed on 31 December 2022).
  28. Shields, R.K.; Nguyen, M.H.; Press, E.G.; Chen, L.; Kreiswirth, B.N.; Clancy, C.J. In Vitro Selection of Meropenem Resistance among Ceftazidime-Avibactam-Resistant, Meropenem-Susceptible Klebsiella pneumoniae Isolates with Variant KPC-3 Carbapenemases. Antimicrob. Agents Chemother. 2017, 61, e00079-17. [Google Scholar] [CrossRef] [Green Version]
  29. Shields, R.K.; Chen, L.; Cheng, S.; Chavda, K.D.; Press, E.G.; Snyder, A.; Pandey, R.; Doi, Y.; Kreiswirth, B.N.; Nguyen, M.H.; et al. Emergence of Ceftazidime-Avibactam Resistance Due to Plasmid-Borne bla KPC-3 Mutations during Treatment of Carbapenem-Resistant Klebsiella pneumoniae Infections. Antimicrob. Agents Chemother. 2017, 61, e02097-16. [Google Scholar] [CrossRef] [Green Version]
  30. Corcione, S.; Lupia, T.; Maraolo, A.E.; Pinna, S.M.; Gentile, I.; De Rosa, F.G. Carbapenem-sparing strategy: Carbapenemase, treatment, and stewardship. Curr. Opin. Infect. Dis. 2019, 32, 663–673. [Google Scholar] [CrossRef]
  31. Winkler, M.L.; Papp-Wallace, K.M.; Bonomo, R.A. Activity of ceftazidime/avibactam against isogenic strains of Escherichia coli containing KPC and SHV β-lactamases with single amino acid substitutions in the Ω-loop. J. Antimicrob. Chemother. 2015, 70, 2279–2286. [Google Scholar] [CrossRef] [Green Version]
  32. Zhang, P.; Shi, Q.; Hu, H.; Hong, B.; Wu, X.; Du, X.; Akova, M.; Yu, Y. Emergence of ceftazidime/avibactam resistance in carbapenem-resistant Klebsiella pneumoniae in China. Clin. Microbiol. Infect. 2020, 26, 124.e1–124.e4. [Google Scholar] [CrossRef] [Green Version]
  33. Woodford, N.; Tierno, P.M., Jr.; Young, K.; Tysall, L.; Palepou, M.-F.I.; Ward, E.; Painter, R.E.; Suber, D.F.; Shungu, D.; Silver, L.L.; et al. Outbreak of Klebsiella pneumoniae Producing a New Carbapenem-Hydrolyzing Class A β-Lactamase, KPC-3, in a New York Medical Center. Antimicrob. Agents Chemother. 2004, 48, 4793–4799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wilson, W.R.; Kline, E.G.; Jones, C.E.; Morder, K.T.; Mettus, R.T.; Doi, Y.; Nguyen, M.H.; Clancy, C.J.; Shields, R.K. Effects of KPC Variant and Porin Genotype on the In Vitro Activity of Meropenem-Vaborbactam against Carbapenem-Resistant Enterobacteriaceae. Antimicrob. Agents Chemother. 2019, 63, e02048-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Castanheira, M.; Doyle, T.B.; Deshpande, L.M.; Mendes, R.E.; Sader, H.S. Activity of ceftazidime/avibactam, meropenem/vaborbactam and imipenem/relebactam against carbapenemase-negative carbapenem-resistant Enterobacterales isolates from US hospitals. Int. J. Antimicrob. Agents 2021, 58, 106439. [Google Scholar] [CrossRef]
  36. Antonelli, A.; Giani, T.; Di Pilato, V.; Riccobono, E.; Perriello, G.; Mencacci, A.; Rossolini, G.M. KPC-31 expressed in a ceftazidime/avibactam-resistant Klebsiella pneumoniae is associated with relevant detection issues. J. Antimicrob. Chemother. 2019, 74, 2464–2466. [Google Scholar] [CrossRef] [PubMed]
  37. Bianco, G.; Boattini, M.; Iannaccone, M.; Bondi, A.; Ghibaudo, D.; Zanotto, E.; Peradotto, M.; Cavallo, R.; Costa, C. Carbapenemase detection testing in the era of ceftazidime/avibactam-resistant KPC-producing Enterobacterales: A 2-year experience. J. Glob. Antimicrob. Resist. 2021, 24, 411–414. [Google Scholar] [CrossRef]
  38. Mueller, L.; Masseron, A.; Prod’Hom, G.; Galperine, T.; Greub, G.; Poirel, L.; Nordmann, P. Phenotypic, Biochemical, and Genetic Analysis of KPC-41, a KPC-3 Variant Conferring Resistance to Ceftazidime-Avibactam and Exhibiting Reduced Carbapenemase Activity. Antimicrob. Agents Chemother. 2019, 63, e01111-19. [Google Scholar] [CrossRef] [Green Version]
  39. Gaibani, P.; Re, M.C.; Campoli, C.; Viale, P.; Ambretti, S. Bloodstream infection caused by KPC-producing Klebsiella pneumoniae resistant to ceftazidime/avibactam: Epidemiology and genomic characterization. Clin. Microbiol. Infect. 2020, 26, 516.e1–516.e4. [Google Scholar] [CrossRef]
  40. Iannaccone, M.; Boattini, M.; Bianco, G.; Corcione, S.; Cavallo, R.; Costa, C. Ceftazidime-avibactam susceptible to resistant KPC-producing Enterobacterales bloodstream infections: An observational study. J. Chemother. 2020, 32, 160–162. [Google Scholar] [CrossRef]
  41. Bianco, G.; Boattini, M.; Iannaccone, M.; Cavallo, R.; Costa, C. Bloodstream infection by two subpopulations of Klebsiella pneumoniae ST1685 carrying KPC-33 or KPC-14 following ceftazidime/avibactam treatment: Considerations regarding acquired heteroresistance and choice of carbapenemase detection assay. J. Antimicrob. Chemother. 2020, 75, 3075–3076. [Google Scholar] [CrossRef]
  42. Bianco, G.; Boattini, M.; Comini, S.; Leone, A.; Bondi, A.; Zaccaria, T.; Cavallo, R.; Costa, C. Implementation of Chromatic Super CAZ/AVI® medium for active surveillance of ceftazidime-avibactam resistance: Preventing the loop from becoming a spiral. Eur. J. Clin. Microbiol. Infect. Dis. 2022, 41, 1165–1171. [Google Scholar] [CrossRef]
  43. Shbaklo, N.; Corcione, S.; Vicentini, C.; Giordano, S.; Fiorentino, D.; Bianco, G.; Cattel, F.; Cavallo, R.; Zotti, C.M.; De Rosa, F.G. An Observational Study of MDR Hospital-Acquired Infections and Antibiotic Use during COVID-19 Pandemic: A Call for Antimicrobial Stewardship Programs. Antibiotics 2022, 11, 695. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Distribution and overlap of mutant KPC-Kp isolates.
Figure 1. Distribution and overlap of mutant KPC-Kp isolates.
Ijms 24 04767 g001
Table 1. Susceptibility profile of mutant KPC-Kp.
Table 1. Susceptibility profile of mutant KPC-Kp.
AntibioticSensitivityMIC (µg/L)MIC Breakpoint
S≤R>
AmpicillinR>888
Amoxicillin/clavulanic acid >32
PiperacillinR>1688
Pip/TazobactamR>1688
TicarcillinR>16816
Cefazolin >160.0014
Cefuroxime OV R>80.0018
Ceftazidime R>3214
Cefotaxime R1612
CefepimeR>814
Cefixime R>111
Ertapenem R>10.50.5
Imipenem S≤124
Meropenem S228
Aztreonam I214
Ciprofloxacin R>10.250.5
Levofloxacin R>10.51
Amikacin R>1688
Gentamicin S≤222
Tobramicin R>422
Colistin R>422
Fosfomicin IVR>643232
Ceftazidime/avibactam R>888
Ceftolozane/tazobactam R>422
Trimetoprim-sulfametoxazoleR>4/7624
Carbapenemase +
Carbapenemase KPC +
Table 2. Clinical characteristics of patients with mutant KPC-Kp.
Table 2. Clinical characteristics of patients with mutant KPC-Kp.
Sex/AgePrinciple DiagnosisComorbiditiesCOVID-19Time from ICU AdmissionKPC Mutant SpecimenKPC Sensitive IsolationPrevious InfectionsA. baumannii Intestinal ColonizationPrevious Broad-Spectrum ATBPrevious Antifungal TherapyCZA Treatment before IsolationIn-Hospital Mortality
F/72ARDS COVID-19AH, NIDDM, Obesity, malignancyYes3RSYes nonoyesnonono
M/52HAP in COVID-19 Yes64Blood, UCYes VAP E. coli, MRSA, A. baumannii, KPC + BSI MRSE, KPC, E. faecium.yesyesyesyesyes
M/56Difficult weaning Smoker, COPD No51BALYes Serratia marcescens + Pseudomonas in BAL, CMV, BSI MRSE. yesyesyesyesYes
M/35Pulmonary embolism in trauma (vv-ECMO)ObesityNo72BAL Yes BSI A. baumannii, SSTI VREyesyesnoyesno
F/61Difficult weaning in H. influenzae SCAP No14RSYes BSI H. influenzae, MRSE, S. marcescens, P. aeruginosa.noyesyesnoyes
M/49ARDS COVID-19ObesityYes15RSYes VAP H. influenzaenoyesnonono
F/72ARDS COVID-19AH, autoimmune diseaseYes17RSYes VAP H. influenzae, CAPAnoyesYesnoyes
M/46ARDS COVID-19Previous smoker, haematological diseaseYes10RSNoVAP A. baumannii, KPC, MSSAyesnononoyes
M/71Difficult weaning AH, COPD, smokerNo87RSYesVAP A. baumannii, KPC, K. oxytoca, P. aeruginosa; C. difficile, BSI MRSE + Candidayesyesyesnoyes
F/35ARDS COVID-19, pulomonary embolism in ECMO VVAH, NIDDM, obesityYes8RSYesVAP MRSE + K. aerogenesyesyesnonono
F/34ARDS COVID-19obesityYes5RSYesVAP E. coli + S. marcescens, CAPAnoyesyesnono
F/55ARDS COVID-19AH, haematological diseaseYes13RS, bloodYesUTI E. faecalis + E. coli, BSI S. capitis, UTI E. colinoyesnonono
F/69ARDS COVID-19AHYes13RSNoVAP P. aeruginosa, CAPAnoyesyesyesyes
F/58ARDS COVID-19AH, obesity, NIDDM, respiratory and autoimmune disease Yes12RSYesVAP MRSA + Proteusnoyesyesnono
M/57ARDS COVID-19autoimmune diseaseYes15RSYesVAP P. aeruginosa, BSI C. albicans, C. auris colonization.noyesyesnoyes
M/69ARDS COVID-19autoimmune disease, respiratory disease, CKDYes5RSNoVAP P. aeruginosanoyesnonoyes
M/71ARDS COVID-19Previous smoker, NIDDM, CRF, Obesity, Malignancy, CKDYes6RSYes noyesyesnoyes
Abbreviations: ARDS: acute respiratory distress syndrome, AH: arterial hypertension, BAL: broncoalveolar lavage, VAP: ventilator associated pneumonia, BSI: blood-stream infection, CAPA: COVID-19 Associated Pulmonary Aspergillosis, CKD: chronic kidney disease, COPD: Chronic obstructive pulmonary disease, CRF: chronic renal failure, HAP: hospital-acquired pneumoniae, KPC: Klebsiella pneumoniae carbapenemase, MRSE: Methicillin-Resistant Staphylococcus epidermidis, MSSA: methicillin-sensitive Staphylococcus aureus, NIDDM: non-insulin-dependent diabetes mellitus, RS: rectal swab, SCAP: Severe Community-Acquired Pneumonia, SSTI: Skin and soft tissue infections (SSTI).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Corcione, S.; De Benedetto, I.; Shbaklo, N.; Torsello, G.; Lupia, T.; Bianco, G.; Cavallo, R.; Brazzi, L.; Montrucchio, G.; De Rosa, F.G. Ceftazidime-Avibactam (C/A) Resistant, Meropenem Sensitive KPC-Producing Klebsiella pneumoniae in ICU Setting: We Are What We Are Treated with? Int. J. Mol. Sci. 2023, 24, 4767. https://doi.org/10.3390/ijms24054767

AMA Style

Corcione S, De Benedetto I, Shbaklo N, Torsello G, Lupia T, Bianco G, Cavallo R, Brazzi L, Montrucchio G, De Rosa FG. Ceftazidime-Avibactam (C/A) Resistant, Meropenem Sensitive KPC-Producing Klebsiella pneumoniae in ICU Setting: We Are What We Are Treated with? International Journal of Molecular Sciences. 2023; 24(5):4767. https://doi.org/10.3390/ijms24054767

Chicago/Turabian Style

Corcione, Silvia, Ilaria De Benedetto, Nour Shbaklo, Giulia Torsello, Tommaso Lupia, Gabriele Bianco, Rossana Cavallo, Luca Brazzi, Giorgia Montrucchio, and Francesco Giuseppe De Rosa. 2023. "Ceftazidime-Avibactam (C/A) Resistant, Meropenem Sensitive KPC-Producing Klebsiella pneumoniae in ICU Setting: We Are What We Are Treated with?" International Journal of Molecular Sciences 24, no. 5: 4767. https://doi.org/10.3390/ijms24054767

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop