Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1962 2022-10-28 14:34:05 |
2 format correct + 1 word(s) 1963 2022-10-31 05:18:12 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Giamarellou, H.;  Karaiskos, I. Pandrug-Resistant Acinetobacter baumannii. Encyclopedia. Available online: https://encyclopedia.pub/entry/31837 (accessed on 27 July 2024).
Giamarellou H,  Karaiskos I. Pandrug-Resistant Acinetobacter baumannii. Encyclopedia. Available at: https://encyclopedia.pub/entry/31837. Accessed July 27, 2024.
Giamarellou, Helen, Ilias Karaiskos. "Pandrug-Resistant Acinetobacter baumannii" Encyclopedia, https://encyclopedia.pub/entry/31837 (accessed July 27, 2024).
Giamarellou, H., & Karaiskos, I. (2022, October 28). Pandrug-Resistant Acinetobacter baumannii. In Encyclopedia. https://encyclopedia.pub/entry/31837
Giamarellou, Helen and Ilias Karaiskos. "Pandrug-Resistant Acinetobacter baumannii." Encyclopedia. Web. 28 October, 2022.
Pandrug-Resistant Acinetobacter baumannii
Edit

Carbapenem resistance in Gram-negative bacteria has come into sight as a serious global threat. Carbapenem-resistant Gram-negative pathogens and their main representatives Klebsiella pneumoniaeAcinetobacter baumannii, and Pseudomonas aeruginosa are ranked in the highest priority category for new treatments.

cefiderocol eravacycline A. baumannii

1. Epidemiological Issues

Acinetobacter is an important cause of hospital-acquired infections, occurring mainly in ICU patients and among residents of long-term care facilities [1]. The most common infections encountered in the clinical setting are BSI, including catheter-relating bloodstream infections (CRBSI) and HAP, including VAP [2]. The most worrisome phenomenon of the last couple of years is the rise of PDR strains characterized as non-susceptible to all conventional antimicrobial agents [3]. In a systemic review of the current epidemiology and prognosis of PDR Gram—negative bacteria—a total of 526 PDR isolates were reported with 172 of them being PDR A. baumannii. The majority of PDR strains were isolated from ICU units, with a potential to cause hospital outbreaks, dissemination between hospitals and long-term facilities, as well as international transmission to other countries. PDR infections were associated with excess mortality, mounting up to 71%, and were independently high regardless of the infection source [4]. Notably, in a cohort study of 91 patients infected (n = 62) or colonized (n = 29) with PDR carbapenemase producing A. baumannii (CRAB), a three-fold increased hazard of mortality was observed in favor of patients with an infection caused by PDR CRAB [5]. Likewise, the comparison of patients with CRAB infections to patients with infections caused by carbapenem-susceptible A. baumannii was linked to increased mortality, prolongation of hospital stay, increased rate of ICU utilization, and hospital charges [6].

2. Therapeutic Options

2.1. Antibiotics with Activity In Vitro against Carbapenemase Producing A. baumannii

The optimal therapeutic strategy for the management of carbapenemase producing A. baumannii (CRAB) infections exhibiting extensive drug-resistant phenotypes is very limited [7]. There is no “standard of care” treatment regimen for the therapy of CRAB. Sulbactam, meropenem, tigecycline, as well as polymyxins, the last-resort antibiotics in recent decades, have been used in critically ill patients for the treatment of CRAB infections [8]. Sulbactam, an irreversible β-lactamase inhibitor, has demonstrated activity against A. baumannii strains; unfortunately, it is administrated in combination with ampicillin (3 gr of ampicillin-sulbactam is comprised of 2 gr of ampicillin and 1 gr of sulbactam) [9]. For the treatment of CRAB infections, a dose of 9 gr ampicillin-sulbactam every 8 h with extended infusion of 4 h (total dose of 27 gr ampicillin-sulbactam in a patient with normal renal function) is suggested [8][10]. Polymyxins and mainly colistin is the most common antibiotic utilized in clinical practice for infections caused by CRAB [11][12][13]. In a systematic review and meta-analysis of polymyxins-based vs. non-polymyxins-based therapies in infections caused by CRAB, polymyxins-based therapies in terms of clinical efficacy had an advantage over non-polymyxins-based therapies (OR, 1.99; 95% CI, 1.31 to 3.03; p =0.001) [14]. The dosage of polymyxins is illustrated in detail in the International Consensus Guidelines for the Optimal Use of the Polymyxins [15]. Tigecycline, although it demonstrates being in vitro susceptible to A. baumannii [16], has been linked with higher mortality and lower microbiological eradication in two meta-analyses [17][18]. Improved clinical rates and lower mortality rates have been demonstrated when administrating a high dose of tigecycline (loading dose of 200 mg followed by 100 mg every 12 h) [19]. Thus, a high dose of tigecycline is recommended for the treatment of CRAB infections. Meropenem as a high-dose extended infusion of 3 gr every 8 h with a 3-h infusion has been utilized in combination therapy for the treatment of CRAB infections [8]. Lastly, in response to the medical need for new treatment options, cefiderocol and eravacycline, two new antimicrobial agents with in vitro susceptibility, have been recently approved [20][21]. The major problem is that the distribution of newly approved antimicrobial agents is suboptimal, with eravacycline being unavailable in Europe [22] and cefiderocol being used in compassionate access [23] or been recently launched in a minority of European markets (i.e., United Kingdom, Germany, and Italy) [24].
A respectable spectrum of antimicrobial combinations has been evaluated in vitro and in animal models, predominately based on polymyxins, rifampicin, fosfomycin, sulbactam, and carbapenems with promising results [25]. On the other hand, a variety of clinical studies evaluating in vitro synergy have failed to demonstrate superiority [26][27][28][29]. Indicatively, clinical studies comparing colistin monotherapy to colistin–rifampicin [26], colistin–fosfomycin [27], and colistin–meropenem combinations [28][29] depicted similar mortality rates with no significantly statistical difference in clinical cure. In a multicenter study from Italy, two hundred and ten ICU patients with infections due to XDR A. baumannii received either colistin methanesulphate (CMS) as monotherapy at a dose of 2 MU every 8 h intravenously, or CMS plus rifampicin 600 mg every 12 h intravenously. The thirty-day mortality in the combination and in the monotherapy arm was 43.3% and 42.9%, respectively, with no difference observed in terms of infection-related death and length of hospitalization [26]. In another study, ninety-four patients infected with CRAB (mostly HAP or VAP) were randomized to receive a combination of intravenous CMS at a dosage of 5 mg of colistin base activity/kg of body weight daily plus intravenous fosfomycin sodium at a dosage of 4 g every 12 h (47 patients in the combination group) or intravenous CMS (47 patients in the monotherapy group). Favorable clinical outcomes, mortality at the end of study treatment, and mortality at 28 days were not significantly different between groups [27]. The major drawback of both studies was the suboptimal dose of CMS (without a loading dose) utilized [26][27]. It is of great significance to analyze the two clinical trials evaluating the role of colistin monotherapy vs. colistin in combination with meropenem, due to large number of participants and the application of updated dose schemes [28][29]. The effectiveness of colistin monotherapy (9 million unit loading dose, followed by 4.5 million units every 12 h) to colistin–meropenem combination (2 gr prolonged infusion every 8 h) therapy for the treatment of severe infections caused by CRAB was evaluated in a randomized trial (with blinded outcome assessment). The majority of the patients had HAP, VAP, or bacteremia. Clinical failure rates for patients who received monotherapy versus combination therapy were 83% (125/151) vs. 81% (130/161) (p = 0.64), whereas mortality at 28 days was 46% (70/151) vs. 52% (84/161) (p = 0.4) for patients with A. baumannii infections [28]. In the second trial, 214 patients were enrolled in the colistin monotherapy arm and 211 in the meropenem-colistin combination arm. A. baumannii was the most common bacteria isolated (77%) and the most prevalent infections were nosocomial pneumonia and BSI. There were no differences between monotherapy and combination therapy in respect to 30-day mortality (43% vs. 37%, p = 0.21) and clinical failure rates (45% vs. 38%, p = 0.18) [29]. The results of both clinical trials strongly encourage the avoidance of colistin–carbapenem combination therapy for carbapenem-resistant A baumannii infections, regardless of the infection course.

2.2. Salvage Treatment

A combination therapy with at least two agents, with in vitro activity whenever applicable, has been proposed by the IDSA guidelines for the treatment of moderate to severe CRAB infections [8]. The major issue, not referred to in the guidelines, is the treatment of PDR CRAB infections. Therapeutic options in these cases are based on in vitro and animal studies [25][30]. Two case series study with triple combination therapy have been reported for the treatment of PDR CRAB and are gradually implemented in clinical practice as salvage treatments due to the lack of other therapeutic choices [31][32], as shown in Table 2. The first study from Greece evaluated the triple combination therapy of intravenous high dose ampicillin-sulbactam (dose of 9 gr every 8 h), high dose of tigecycline (200 mg loading dose followed by 100 mg every 12 h), and intravenous CMS (9 million units loading dose, followed by 4.5 million units every 12 h) in 10 ICU patients with a VAP infection caused by A. baumannii with a PDR phenotype. The Charlson comorbidity index was ≥3 and the median APACHE score was of 23 ± 3. A successful clinical outcome was observed in 90% (9/10), whereas microbiological eradication was identified in 70% (7/10 patients). The 28-day mortality was of 10%, whereas nephrotoxicity was observed in one patient [31]. In another study, 20 patients with a median APACE score of 19.5 (range, 10–28) with infections caused by colistin-resistant A. baumannii were evaluated. The most common infections were VAP and bacteremia in 65% (13/20) and 10% (2/20), respectively. Three patients were characterized as colonization and were not treated, whereas the remaining 17 patients were treated in the majority with various CMS-based combination regimens. The most prevalent combination was a combination of carbapenem, ampicillin-sulbactam and CMS prescribed in seven patients. Mortality was depicted as lower in a statistical matter between triple combination and patients receiving other antimicrobial agents for the treatment of colistin-resistant A. baumannii (0% vs. 60%, p = 0.03) [33].

2.3. New Antimicrobials

Cefiderocol

In the SIDERO-CR-2014-2016 surveillance in vitro study, European clinical isolates comprising MDR non-fermenter A. baumannii was tested against cefiderocol and 94.9% had a cefiderocol MIC ≤ 2 mg/L [34]. CREDIBLE-CR was a randomized, open-label, multicenter trial of cefiderocol (n = 101) and the best available treatment (BAT) (n = 49) for the treatment of severe infections (cUTI, nosocomial pneumonia, BSI, or sepsis) caused by carbapenem-resistant Gram-negative pathogens. In 118 patients in the carbapenem-resistant microbiological intent to treat (ITT) population, the most common baseline pathogen was A. baumannii in 46% (54/118). Cefiderocol was administrated as monotherapy in 83% (66/80) and combination therapy (mostly colistin-based regimens) was given in 71% (27/38) in the BAT arm. The clinical cure rates in the cefiderocol (22/49) and comparator (13/25) regarding A. baumannii were similar (45% vs. 52%). An increase in all-cause mortality was observed in patients treated with cefiderocol as compared to BAT. However, the greatest mortality imbalance disfavoring cefiderocol was noted in the nosocomial pneumonia subgroup, followed by BSI. The difference in 49-day mortality stratified for pathogen was the highest for Acinetobacter spp. (50% (21/ 42) vs. 18% (3/17) in cefiderocol and BAT-treated patients, respectively [35]. Deaths due to treatment failure in the cefiderocol group occurred more often in the patients infected with Acinetobacter spp. Of the 16 deaths due to treatment failure, 13 involved Acinetobacter spp. [34][35]. In conclusion, treatment failure was linked with infection caused by Acinetobacter spp., pulmonary infection at baseline, and by increases in cefiderocol MIC while on therapy [34][35]. An additional phase 3 trial, named APEKS-NP, evaluated hospital-acquired, ventilator-associated, or health-care-associated Gram-negative pneumonia and found cefiderocol was non-inferior to high-dose meropenem in patients. Fourteen-day all-cause mortality, clinical cure, and microbiologic eradication were similar between treatment groups for participants infected with A. baumannii; however, this group only comprised 16% of the study population, of which 66% of isolates were carbapenemase-resistant [36]. Cefiderocol has also been administrated as compassionate use in a limited number of case series with infections caused by XDR and PDR A. baumannii pathogens, resulting in a clinical success of 80% (20/25) [23][37]. Overall, the necessity of further studies to elucidate the true role of cefiderocol against A. baumannii infections in real life patients is needed.

Eravacycline

Eravacycline is a synthetic fluorocycline antibacterial agent that is structurally similar to tigecycline with two modifications at the D-ring of its tetracycline core [21]. In vitro activity of eravacycline against A. baumannii isolates (n = 2097) worldwide (from 2013 to 2017) revealed an MIC90s of 1 mg/L, demonstrating improved potency up to 4-fold greater than that of tigecycline [38]. Eravacycline has successfully completed clinical trial phase 3 for the treatment of cIAI; however, A. baumannii infections only comprised 3% of the total isolated pathogens [39]. Clinical studies with infections caused by CRAB reporting efficacy of eravacycline are lacking and are limited to one study. In a retrospective report of 93 adults hospitalized for pneumonia with DTR A. baumannii, 27 patients received eravacycline and were compared to those receiving the best available therapy. Eravacycline-based combination therapy had similar outcomes to the best available combination therapy. However, when taking under consideration patients with secondary bacteremia and coinfection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), eravacycline was associated with higher 30-day mortality (33% vs. 15%; p = 0.048), lower microbiologic cure (17% vs. 59%; p = 0.004), and longer durations of mechanical ventilation (10.5 vs. 6.5 days; p = 0.016), highlighting the avoidance of use in bacteremic patients [40]. However, eravacycline could be a suitable candidate for the treatment of cIAI caused by XDR, and even PDR pathogens. Therefore, further clinical studies addressing the efficacy of eravacycline in difficult-to-treat infections is required.

New β-Lactamase Inhibitor

Durlobactam, previously known as ETX2514, is a novel diazabicyclooctane class of β-lactamase inhibitor specifically designed to inhibit class D β-lactamases, in addition to class A and C enzymes. Durlobactam is combined with sulbactam, and targets infections caused by A. baumannii [41]. It has completed clinical trials in combination with sulbactam for the treatment of hospitalized adults with complicated urinary tract infection (cUTI) (Phase 2, clinicaltrials.gov identifier: NCT03445195) [42] and for the treatment of HAP and VAP caused by A. baumannii vs. colistin plus imipenem and the results are pending (Phase 3, clinicaltrials.gov identifier: NCT03894046).

References

  1. Nguyen, M.; Joshi, S.G. Carbapenem resistance in Acinetobacter baumannii, and their importance in hospital-acquired infections: A scientific review. Appl. Microbiol. 2021, 131, 2715–2738.
  2. Peleg, A.Y.; Seifert, H.; Paterson, D.L. Acinetobacter baumannii: Emergence of a successful pathogen. Clin. Microbiol. Rev. 2008, 21, 538–582.
  3. Kofteridis, D.P.; Andrianaki, A.M.; Maraki, S.; Mathioudaki, A.; Plataki, M.; Alexopoulou, C.; Ioannou, P.; Samonis, G.; Valachis, A. Treatment pattern, prognostic factors, and outcome in patients with infection due to pan-drug-resistant gram-negative bacteria. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 965–970.
  4. Karakonstantis, S.; Kritsotakis, E.I.; Gikas, A. Pandrug-resistant Gram-negative bacteria: A systematic review of current epidemiology, prognosis and treatment options. J. Antimicrob. Chemother. 2020, 75, 271–282.
  5. Karakonstantis, S.; Gikas, A.; Astrinaki, E.; Kritsotakis, E.I. Excess mortality due to pandrug-resistant Acinetobacter baumannii infections in hospitalized patients. J. Hosp. Infect. 2020, 106, 447–453.
  6. Pogue, J.M.; Zhou, Y.; Kanakamedala, H.; Cai, B. Burden of illness in carbapenem-resistant Acinetobacter baumannii infections in US hospitals between 2014 and 2019. BMC Infect. Dis. 2022, 22, 36.
  7. Karaiskos, I.; Lagou, S.; Pontikis, K.; Rapti, V.; Poulakou, G. The “Old” and the “New” antibiotics for MDR Gram-negative pathogens: For whom, when, and how. Front. Public Health 2019, 7, 151.
  8. Tamma, P.D.; Aitken, S.L.; Bonomo, R.A.; Mathers, A.J.; van Duin, D.; Clancy, C.J. Infectious Diseases Society of America guidance on the treatment of AmpC β-lactamase-producing Enterobacterales, carbapenem-resistant Acinetobacter baumannii, and Stenotrophomonas maltophilia infections. Clin. Infect. Dis. 2022, 74, ciab1013.
  9. Abdul-Mutakabbir, J.C.; Griffith, N.C.; Shields, R.K.; Tverdek, F.P.; Escobar, Z.K. Contemporary perspective on the treatment of Acinetobacter baumannii infections: Insights from the society of infectious diseases pharmacists. Infect. Dis. Ther. 2021, 10, 2177–2202.
  10. Betrosian, A.P.; Frantzeskaki, F.; Xanthaki, A.; Georgiadis, G. High-dose ampicillin-sulbactam as an alternative treatment of late-onset VAP from multidrug-resistant Acinetobacter baumannii. Scand. J. Infect. Dis. 2007, 39, 38–43.
  11. Liu, J.; Shu, Y.; Zhu, F.; Feng, B.; Zhang, Z.; Liu, L.; Wang, G. Comparative efficacy and safety of combination therapy with high-dose sulbactam or colistin with additional antibacterial agents for multiple drug-resistant and extensively drug-resistant Acinetobacter baumannii infections: A systematic review and network meta-analysis. J. Glob. Antimicrob. Resist. 2021, 24, 136–147.
  12. Giacobbe, D.R.; Karaiskos, I.; Bassetti, M. How do we optimize the prescribing of intravenous polymyxins to increase their longevity and efficacy in critically ill patients? Expert Opin. Pharmacother. 2022, 23, 5–8.
  13. Karaiskos, I.; Souli, M.; Galani, I.; Giamarellou, H. Colistin: Still a lifesaver for the 21st century? Expert Opin. Drug Metab. Toxicol. 2017, 13, 59–71.
  14. Lyu, C.; Zhang, Y.; Liu, X.; Wu, J.; Zhang, J. Clinical efficacy and safety of polymyxins based versus non-polymyxins based therapies in the infections caused by carbapenem-resistant Acinetobacter baumannii: A systematic review and meta-analysis. BMC Infect. Dis. 2020, 20, 296.
  15. Tsuji, B.T.; Pogue, J.M.; Zavascki, A.P.; Paul, M.; Daikos, G.L.; Forrest, A.; Giacobbe, D.R.; Viscoli, C.; Giamarellou, H.; Ilias Karaiskos, I.; et al. International Consensus Guidelines for the Optimal Use of the Polymyxins: Endorsed by the American College of Clinical Pharmacy (ACCP), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), Infectious Diseases Society of America (IDSA), International Society for Anti-infective Pharmacology (ISAP), Society of Critical Care Medicine (SCCM), and Society of Infectious Diseases Pharmacists (SIDP). Pharmacotherapy 2019, 39, 10–39.
  16. Pournaras, S.; Koumaki, V.; Gennimata, V.; Kouskouni, E.; Tsakris, A. In Vitro Activity of Tigecycline Against Acinetobacter baumannii: Global Epidemiology and Resistance Mechanisms. Adv. Exp. Med. Biol. 2016, 97, 1–14.
  17. Yahav, D.; Lador, A.; Paul, M.; Leibovici, L. Efficacy and safety of tigecycline: A systematic review and meta-analysis. J. Antimicrob. Chemother. 2011, 66, 1963–1971.
  18. Mei, H.; Yang, T.; Wang, J.; Wang, R.; Cai, Y. Efficacy and safety of tigecycline in treatment of pneumonia caused by MDR Acinetobacter baumannii: A systematic review and meta-analysis. J. Antimicrob. Chemother. 2019, 74, 3423–3431.
  19. Zha, L.; Pan, L.; Guo, J.; French, N.; Villanueva, E.V.; Tefsen, B. Effectiveness and safety of high dose tigecycline for the treatment of severe infections: A systematic review and meta-analysis. Adv. Ther. 2020, 37, 1049–1064.
  20. Giacobbe, D.R.; Ciacco, E.; Girmenia, C.; Pea, F.; Rossolini, G.M.; Sotgiu, G.; Tascini, C.; Tumbarello, M.; Viale, P.; Bassetti, M.; et al. Evaluating cefiderocol in the treatment of multidrug-resistant Gram-negative bacilli: A review of the emerging data. Infect. Drug Resist. 2020, 13, 4697–4711.
  21. Lee, Y.R.; Burton, C.E. Eravacycline, a newly approved fluorocycline. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 1787–1794.
  22. Rex, J.H.; Outterson, K. New Antibiotics Are Not Being Registered or Sold in Europe in a Timely Manner. 2020. Available online: https://amr.solutions/2020/09/07/new-antibiotics-are-not-being-registered-or-sold-in-europe-in-a-timely-manner (accessed on 15 July 2022).
  23. Oliva, A.; Ceccarelli, G.; De Angelis, M.; Sacco, F.; Miele, M.C.; Mastroianni, C.M.; Venditti, M. Cefiderocol for compassionate use in the treatment of complicated infections caused by extensively and pan-resistant Acinetobacter baumannii. J. Glob. Antimicrob. Resist. 2020, 23, 292–296.
  24. Application for Inclusion of FETCROJA/FETROJA (Cefiderocol) on the WHO Model List of Essential Medicines. 2020. Available online: https://cdn.who.int/media/docs/default-source/essential-medicines/2021-eml-expert-committee/applications-for-addition-of-new-medicines/a.7_cefiderocol.pdf?sfvrsn=db365d99_4 (accessed on 15 July 2022).
  25. Karakonstantis, S.; Ioannou, P.; Samonis, G.; Kofteridis, D.P. Systematic review of antimicrobial combination options for pandrug-resistant Acinetobacter baumannii. Antibiotics 2021, 10, 1344.
  26. Durante-Mangoni, E.; Signoriello, G.; Andini, R.; Mattei, A.; De Cristoforo, M.; Murino, P.; Bassetti, M.; Malacarne, P.; Petrosillo, N.; Galdieri, N.; et al. Colistin and rifampicin compared with colistin alone for the treatment of serious infections due to extensively drug-resistant Acinetobacter baumannii: A multicenter, randomized clinical trial. Clin. Infect. Dis. 2013, 57, 349–358.
  27. Sirijatuphat, R.; Thamlikitkul, V. Preliminary study of colistin versus colistin plus fosfomycin for treatment of carbapenem-resistant Acinetobacter baumannii infections. Antimicrob. Agents Chemother. 2014, 58, 5598–5601.
  28. Paul, M.; Daikos, G.L.; Durante-Mangoni, E.; Yahav, D.; Carmeli, Y.; Benattar, Y.D.; Skiada, A.; Andini, R.; Eliakim-Raz, N.; Nutman, A.; et al. Colistin alone versus colistin plus meropenem for treatment of severe infections caused by carbapenem-resistant Gram-negative bacteria: An open-label, randomised controlled trial. Lancet Infect. Dis. 2018, 18, 391–400.
  29. Kaye, K.; Marchaim, D.; Thamlikitkul, V.; Carmeli, Y.; Chiu, C.H.; Daikos, G.; Dhar, S.; Durante-Mangoni, E.; Gikas, A.; Kotanidou, A.; et al. Results from the OVERCOME Trial: Colistin monotherapy versus combination therapy for the treatment of pneumonia or bloodstream infection due to extensively drug resistant Gram-negative bacilli. In Proceedings of the 31st European Congress of Clinical Microbiology & Infectious Diseases (ECCMID), Vienna, Austria, 9–12 July 2021.
  30. Lenhard, J.R.; Smith, N.M.; Bulman, Z.P.; Tao, X.; Thamlikitkul, V.; Shin, B.S.; Nation, R.L.; Li, J.; Bulitta, J.B.; Tsuji, B.T. High-dose ampicillin-sulbactam combinations combat polymyxin-resistant Acinetobacter baumannii in a hollow-fiber infection model. Antimicrob. Agents Chemother. 2017, 61, e01268-16.
  31. Assimakopoulos, S.F.; Karamouzos, V.; Lefkaditi, A.; Sklavou, C.; Kolonitsiou, F.; Christofidou, M.; Fligou, F.; Gogos, C.; Marangos, M. Triple combination therapy with high-dose ampicillin/sulbactam, high-dose tigecycline and colistin in the treatment of ventilator-associated pneumonia caused by pan-drug resistant Acinetobacter baumannii: A case series study. Infez. Med. 2019, 27, 11–16.
  32. Qureshi, Z.A.; Hittle, L.E.; O’Hara, J.A.; Rivera, J.I.; Syed, A.; Shields, R.K.; Pasculle, A.W.; Ernst, R.K.; Doi, Y. Colistin-resistant Acinetobacter baumannii: Beyond carbapenem resistance. Clin. Infect. Dis. 2015, 60, 1295–1303.
  33. Longshaw, C.; Manissero, D.; Tsuji, M.; Echols, R.; Yamano, Y. In vitro activity of the siderophore cephalosporin, cefiderocol, against molecularly characterized, carbapenem-non-susceptible Gram-negative bacteria from Europe. JAC Antimicrob. Resist. 2020, 2, dlaa060.
  34. Bassetti, M.; Echols, R.; Matsunaga, Y.; Ariyasu, M.; Doi, Y.; Ferrer, R.; Lodise, T.P.; Naas, T.; Niki, Y.; Paterson, D.L.; et al. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant Gram-negative bacteria (CREDIBLE-CR): A randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect. Dis. 2021, 21, 226–240.
  35. Naseer, S.; Weinstein, E.A.; Rubin, D.B.; Suvarna, K.; Wei, X.; Higgins, K.; Goodwin, A.; Jang, S.H.; Iarikov, D.; Farley, J.; et al. US Food and Drug Administration (FDA): Benefit-risk considerations for cefiderocol (Fetroja®). Clin. Infect. Dis. 2021, 72, e1103–e1111.
  36. Wunderink, R.G.; Matsunaga, Y.; Ariyasu, M.; Clevenbergh, P.; Echols, R.; Kaye, K.S.; Kollef, M.; Menon, A.; Pogue, J.M.; Shorr, A.F.; et al. Cefiderocol versus high-dose, extended-infusion meropenem for the treatment of Gram-negative nosocomial pneumonia (APEKS-NP): A randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect. Dis. 2021, 21, 213–225.
  37. Babidhan, R.; Lewis, A.; Atkins, C.; Jozefczyk, N.J.; Nemecek, B.D.; Montepara, C.A.; Gionfriddo, M.R.; Zimmerman, D.E.; Covvey, J.R.; Guarascio, A.J. Safety and efficacy of cefiderocol for off-label treatment indications: A systematic review. Pharmacotherapy 2022, 42, 549–566.
  38. Morrissey, I.; Olesky, M.; Hawser, S.; Lob, S.H.; Karlowsky, J.A.; Corey, G.R.; Bassetti, M.; Fyfe, C. In vitro activity of eravacycline against Gram-negative bacilli isolated in clinical laboratories worldwide from 2013 to 2017. Antimicrob. Agents Chemother. 2020, 64, e01699-19.
  39. Solomkin, J.S.; Gardovskis, J.; Lawrence, K.; Montravers, P.; Sway, A.; Evans, D.; Tsai, L. 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, 921–929.
  40. Scott, C.J.; Zhu, E.; Jayakumar, R.A.; Shan, G.; Viswesh, V. Efficacy of eravacycline versus best previously available therapy for adults with pneumonia due to difficult-to-treat resistant (DTR) Acinetobacter baumannii. Ann. Pharmacother. 2022, 10600280221085551.
  41. Yahav, D.; Giske, C.G.; Grāmatniece, A.; Abodakpi, H.; Tam, V.H.; Leibovici, L. A new β-lactam-β-lactamase inhibitor combinations. Clin. Microbiol. Rev. 2020, 34, e00115-20.
  42. Sagan, O.; Yakubsevitch, R.; Yanev, K.; Fomkin, R.; Stone, E.; Hines, D.; O’Donnell, J.; Miller, A.; Isaacs, R.; Srinivasan, S. Pharmacokinetics and tolerability of intravenous sulbactam-durlobactam with imipenem-cilastatin in hospitalized adults with complicated urinary tract infections, including acute pyelonephritis. Antimicrob. Agents Chemother. 2020, 64, e01506-19.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 388
Revisions: 2 times (View History)
Update Date: 31 Oct 2022
1000/1000
Video Production Service