Next Article in Journal
Retroviral Restriction Factors and Their Viral Targets: Restriction Strategies and Evolutionary Adaptations
Next Article in Special Issue
Nebulized Colistin in Ventilator-Associated Pneumonia and Tracheobronchitis: Historical Background, Pharmacokinetics and Perspectives
Previous Article in Journal
Combating Parasitic Nematode Infections, Newly Discovered Antinematode Compounds from Marine Epiphytic Bacteria
Previous Article in Special Issue
Performance of Population Pharmacokinetic Models in Predicting Polymyxin B Exposures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Combination of Colistin and Azidothymidine Demonstrates Synergistic Activity against Colistin-Resistant, Carbapenem-Resistant Klebsiella pneumoniae

1
Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
2
Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
3
Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
4
School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
5
Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
6
Center for Tropical Medicine and Infectious Disease Research, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
7
Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2020, 8(12), 1964; https://doi.org/10.3390/microorganisms8121964
Submission received: 12 November 2020 / Revised: 1 December 2020 / Accepted: 9 December 2020 / Published: 11 December 2020
(This article belongs to the Special Issue Polymyxins: New Insights into An 'Old' Class of Antibiotics)

Abstract

:
Carbapenem-resistant Enterobacteriaceae (CRE) is listed as an urgent threat by the World Health Organization because of the limited therapeutic options, rapid evolution of resistance mechanisms, and worldwide dissemination. Colistin is a common backbone agent among the “last-resort” antibiotics for CRE; however, its emerging resistance among CRE has taken the present dilemma to the next level. Azidothymidine (AZT), a thymidine analog used to treat human immunodeficiency virus/acquired immunodeficiency syndrome, has been known to possess antibacterial effects against Enterobacteriaceae. In this study, we investigated the combined effects of AZT and colistin in 40 clinical isolates of colistin-resistant, carbapenem-resistant K. pneumoniae (CCRKP). Eleven of the 40 isolates harbored Klebsiella pneumoniae carbapenemase. The in vitro checkerboard method and in vivo nematode killing assay both revealed synergistic activity between the two agents, with fractional inhibitory concentration indexes of ≤0.5 in every strain. Additionally, a significantly lower hazard ratio was observed for the nematodes treated with combination therapy (0.288; p < 0.0001) compared with either AZT or colistin treatment. Toxicity testing indicated potentially low toxicity of the combination therapy. Thus, the AZT–colistin combination could be a potentially favorable therapeutic option for treating CCRKP.

1. Introduction

Antimicrobial resistance (AMR) has been one of the most challenging public health problems during this era of expeditious medical advances. Multidrug-resistant organisms (MDROs) are recognized as an imminent global threat that impose substantial medical burdens and economic costs [1]. Various carbapenem-resistant Gram-negative bacteria (GNB) are listed as being critical and urgent MDRO threats by the World Health Organization and the US Centers for Disease Control and Prevention (CDC) [2,3] because of the rapid evolution of their resistance mechanisms and worldwide dissemination. Globally, the prevalence rates of carbapenem-resistant Enterobacteriaceae (CRE) have been increasing, especially those of Klebsiella pneumoniae and Escherichia coli [4]. Because of the limited antimicrobial options and an increased risk for horizontal transmission due to the presence of resistance genes in mobile genetic elements, the treatment and containment of CRE infections have become serious dilemmas in daily practice and in terms of infection control [5].
The phenotypic resistance to carbapenems in CRE typically originates from two main mechanisms: (1) the combined effects of β-lactamase(s) and structural mutations, and (2) the production of carbapenemases [6]. Therefore, CRE is frequently categorized into carbapenemase-producing (CP) CRE (CP-CRE) and non-CP CRE (non-CP-CRE). CP-CRE exhibits higher carbapenem minimum inhibitory concentrations (MICs), results in higher mortality, and has an increased risk for resistance transmission [7,8]. CP-K. pneumoniae (CPKP) is the most widely reported CRE species, with a much higher and increasing prevalence compared with E. coli [9]. Therefore, CPKP is the most extensively studied CRE regarding treatment response to various new and old antibiotics.
Among the list of “old antibiotics” for emerging MDROs, polymyxin B and E (colistin) are frequently used as the backbone of combination therapy for CRE [10]. In countries or regions where new β-lactam/β-lactamase inhibitors (BLBLIs) are unavailable, colistin resistance in CRE has a significant impact on patient survival [11]. In such a difficult situation, combining colistin with a synergistic agent would be a potential option and feasible approach. Azidothymidine (AZT; 3′-azido-3′-deoxythymidine), and also known as zidovudine, is a thymidine analog used as an antiretroviral agent to treat human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS). It has been found to possess antibacterial effects against GNB [12,13,14]. Several studies that were conducted to repurpose screens among FDA-approved drugs showed that AZT is a potential candidate for combination therapy of multidrug-resistant GNB, including CRE [14,15,16].
In this study, we performed synergistic analyses between AZT and colistin on 40 isolates of colistin-resistant, carbapenem-resistant K. pneumoniae (CCRKP) by using the checkerboard method. Meanwhile, an animal model using Caenorhabditis elegans was applied to evaluate the in vivo efficacy and safety of combination therapy. To the best of our knowledge, this study is the first to examine AZT–colistin synergism for concomitant colistin- and carbapenem-resistant K. pneumoniae isolates.

2. Materials and Methods

2.1. Bacterial Isolates

As a part of Taiwan’s nationwide surveillance, 40 isolates of CCRKP were collected between 2013 and 2015 from 11 hospitals in Taiwan [9,17]. Carbapenem resistance was defined as resistance to either imipenem or meropenem, according to the Clinical and Laboratory Standards Institute (CLSI) guidelines [18]. Colistin resistance was defined as MIC > 2 μg/mL, according to the European Committee on Antimicrobial Susceptibility Testing (EUCAST) criteria [19]. Among the 40 isolates, urine was the main source (11/40, 27.5%), followed by sputum (10/40, 25%), stool (4/40, 10%), pus/wound (3/40, 7.5%), abscess (3/40, 7.5%), blood (2/40, 5%), endotracheal aspirate (2/40, 5%), ascites (2/40, 5%), bile (2/40, 5%), and drainage (1/40, 2.5%) (Table 1).

2.2. Antimicrobial Susceptibility

The broth microdilution method (Sensititre, Trek Diagnostic Systems, Cleveland, OH, USA) was used to determine the isolates’ susceptibility profiles against 18 antimicrobial agents, according to the CLSI guidelines [18], including β-lactams (ampicillin, ceftazidime, cefazolin, cefepime, cefoxitin, ceftriaxone, imipenem, meropenem, doripenem, ertapenem, cefotaxime, and piperacillin-tazobactam), monobactams (aztreonam), aminoglycosides (amikacin and gentamicin), quinolones (ciprofloxacin and levofloxacin), and folate inhibitors (trimethoprim/sulfamethoxazole). For tigecycline and colistin, the Enterobacteriaceae breakpoints were adopted, according to the Food and Drug Administration (FDA) and EUCAST guidelines, respectively. Furthermore, a standard broth microdilution method was applied in accordance with CLSI guidelines to evaluate the MICs for colistin and AZT.

2.3. Synergistic Analysis

The synergism between AZT and colistin was investigated through the checkerboard method, as described previously [20]. In brief, the bacterial suspension of each strain was prepared in cation-adjusted Mueller–Hinton broth (CAMHB) and was added to wells at a final concentration of 5 × 105 CFU/mL. Two-fold serial dilutions of AZT and colistin were prepared and added to the wells containing bacteria. The synergistic effects were determined according to the fractional inhibitory concentration (FIC) index, which was calculated as follows:
(MIC of drug A tested in combination)/(MIC of drug A tested alone) +
(MIC of drug B tested in combination)/(MIC of drug B tested alone).
Synergy is defined as an FIC index ≤ 0.5. An FIC index between 0.5 and 4.0 is interpreted as there being no interaction, and antagonism is defined as an FIC index of >4.

2.4. In Vivo Study

C. elegans, strain N2, was used for both in vivo toxicity and nematode-killing assays. Nematode growth medium (NGM) agar plates with bacterial lawns of E. coli, laboratory strain OP50, were used as food sources for the maintenance of nematodes at 20 °C. The procedures were carried out as described in our previous study [21]. NGM agar plates were prepared with the following antibiotic concentrations: colistin, 1 μg/mL; AZT, 0.15 μg/mL; a combination of colistin and AZT, 1 and 0.15 μg/mL, respectively. Clinical isolates of CCRKP strain 1336 or E. coli OP50 were spread onto the plates. Growth-synchronized L4-stage nematodes (40 for each group) were transferred onto the plates containing different antimicrobial agents and bacterial lawns. All plates were maintained at 25 °C, and the survival of nematodes was recorded daily. For the toxicity assay, treatment plates that were supplemented with a vehicle served as the controls. For the killing assay, the plates containing only a lawn of E coli OP50 served as the negative control.

2.5. Polymerase Chain Reaction Detection

Polymerase chain reactions were used to detect the presence of extended-spectrum β-lactamase (ESBL) genes (blaSHV, blaTEM, blaOXA, blaCTX-M-G1, blaCTX-M-G2, and blaCTX-M-G9), plasmid-mediated AmpC genes (blaDHA and blaCMY), carbapenemase genes (blaKPC, blaNDM, blaIMP, blaNMC, blaSME, blaVIM, blaSPM-1, blaGIM-1, blaSIM-1, blaIMI, blaGES, and blaOXA-48), mcr-1 gene and outer membrane porin genes (ompK35 and ompK36) (Table S1) [21]. Reactions were performed using TaKaRa Taq™ (Cat. R001A, Takara Shuzo Co., Ltd., Tokyo, Japan) and prepared in a total volume of 25 μL, in accordance with the instruction manual. All analyses were performed with corresponding positive controls.

2.6. Statistical Analyses

The MIC distributions of colistin and AZT, in combination or alone, were constructed with GraphPad Prism software (v.7.0) and analyzed using paired Student’s t-tests.

3. Results

3.1. Distribution of Resistance Mechanisms and In Vitro Susceptibilities

Among the 40 isolates of CCRKP, Klebsiella pneumoniae carbapenemase (KPC) was identified in 11 of them and was the only carbapenemase. None of the strains harbored the mcr1 gene. For non-CP-CCRKP, 51.7% (15/29) had more than three types of resistance mechanisms, which most commonly involved DHA AmpC β-lactamase gene (20 isolates) and ESBL genes (TEM, 17 isolates; CTX-M, 15 isolates) (Figure 1a). It was determined that all except one isolate of CP-CCRKP had lost OmpK36 (Figure 1b). The antimicrobial susceptibilities to 19 common antimicrobial agents are listed in Table 2; tigecycline and amikacin demonstrated higher susceptibilities of 87.5% and 70%, respectively, compared with the other antimicrobials tested. The MICs for colistin ranged from 4 to 12 μg/mL, with the MIC50 being 64 μg/mL and MIC90 being 128 μg/mL. The MICs for AZT ranged from 0.125 to 16 μg/mL, with the MIC50 being 1 μg/mL and MIC90 being 2 μg/mL (Table 3). Table 3 also shows the changes to MICs after the two drugs were combined. MICs for both AZT and colistin decreased significantly after combining with the respective drug, ranging from 1 to 2 μg/mL for AZT and 0.03125 to 1 μg/mL for colistin. Figure 2 presents a comparison of the MICs of AZT and colistin as single agents and in combination. After combination, the MIC values of colistin fell below the breakpoint for resistance (MIC ≤ 2 μg/mL) in all strains. Moreover, the mean MIC value of AZT attained an average steady-state serum concentration of 0.19 μg/mL after combination with colistin [22].

3.2. Checkerboard Analysis and In Vivo C. elegans Toxicity and Killing Assay

To determine the combined effects of colistin and AZT, the checkerboard method was utilized and demonstrated synergistic activity (Table 4). The FIC indexes were ≤0.5 for 100% of both KPC producers and nonproducers, clearly showing synergism in the 40 strains of CCRKP, regardless of carbapenemase production (all KPC in this study).
For the toxicity testing of colistin, AZT, and their combination, C. elegans were fed with E. coli OP50 and treated with different regimens. As shown in Figure 3 and Table 5, no differences were observed between the groups of nematodes treated with colistin, AZT, or a combination thereof compared with the E. coli OP50 control group. This result suggests that the combination of AZT and colistin is safe. In addition, the synergistic activity between AZT and colistin was confirmed using the nematode killing assay. C. elegans fed with nontoxic E. coli OP50 (OP50-control) had a significantly longer median survival time (9 days; p < 0.0001) compared with C. elegans infected with a clinical strain of K. pneumoniae 1336 (1336-control) (Figure 4 and Table 5). There were no significant differences found between the 1336-control and the nematodes treated with either colistin at 1 μg/mL or AZT at 0.15 μg/mL, implying that neither of the antimicrobial agents could rescue the nematodes infected with a CCRKP. By contrast, the infected nematodes that were treated with a combination of colistin and AZT had a medium survival time that was significantly extended, from 6 days to 8.5 days (p < 0.0001). A significantly lower hazard ratio (HR) was observed for the nematodes treated with combination therapy (HR, 0.288; 95% confidence interval 0.17 to 0.50; p < 0.0001).

4. Discussion

CRE has become an imminent global hazard with increasing prevalence worldwide, leading to increased healthcare costs and higher mortality [23]. Among the carbapenemases with a high risk for transmission, KPC is the most prevalent and widely spread [24]. Despite the introduction of novel BLBLIs on the market in recent years, such as ceftazidime/avibactam, imipenem/relebactam, and meropenem/vaborbactam, polymyxins remain an important agent for the treatment of CRE, especially in regions where the new BLBLIs are unavailable or prevalent with metallo-β-lactamases-producing CRE. Nevertheless, the emerging resistance of CRE against “last-resort” antibiotics, such as colistin, tigecycline, and fosfomycin, has been increasingly reported [11,25,26,27]. The emergence of the colistin resistance gene mcr1 in CRE is of great concern, especially with its potential to spread into geographical areas beyond China and Southeast Asia, where it is most frequently reported, and into Enterobacteriaceae species other than E. coli and K. pneumoniae [28,29].
In this study, the mcr1 gene was not detected in any of the 40 clinical isolates of CCRKP. The extensive resistance primarily comes from the combination of multiple extensive-spectrum β-lactamases or carbapenemase (all KPC), predominantly combined with outer membrane protein deficiency (all Omp36K). AZT alone demonstrated relatively low MICs for the CCRKP (Table 3), and a similar observation was reported by Peyclit et al. for one strain of colistin-resistant, carbapenem-resistant K. pneumoniae, with AZT being highly effective at an MIC of 0.104 μg/mL [14]. Among the 12 strains of isolates with relatively higher AZT MICs, ranging from 2 to 16 μg/mL (nine isolates at 2 μg/mL, two isolates at 8 μg/mL, one isolate at 16 μg/mL; data not shown), only two strains harbored KPC, both with MICs at 2 μg/mL. Notably, all KPC-producing strains had lower MICs for AZT (0.25–2 μg/mL). Among the three isolates that exhibited the highest MICs for AZT, two had only one resistance gene (SHV (MIC = 8) and DHA (MIC = 16)), and one had multiple β-lactamase genes (TEM, DHA, and CTX-M (MIC = 8)). This observation is reasonable considering that the resistance mechanisms of AZT appear to involve the loss of thymidine kinase activity and/or poor cell membrane permeation [30,31] instead of the more commonly known mechanisms of β-lactamases. This suggests that AZT could be a favorable therapeutic candidate, even for carbapenemase-producing organisms.
We demonstrated the evident synergy between AZT and colistin using the checkerboard method (Table 4) and nematode killing assay (Table 5). The synergistic activity of polymyxins and AZT among E. coli and K. pneumoniae carrying various resistance mechanisms (Table 6) has been reported in several previous studies [32,33,34,35]. The most commonly investigated resistance genotypes are mcr1 and blaNDM-1. This study is the first to examine the combined effects of AZT and colistin in Enterobacteriaceae with concomitant colistin and carbapenem resistance. It also bears the important clinical implication that the colistin MIC in these 40 isolates decreased dramatically to a range of 1–2 μg/mL after combination with AZT, which is defined as susceptible by the EUCAST guidelines. AZT is a thymidine analog that acts as an inhibitor of HIV reverse transcriptase. Before being adopted as the first antiretroviral drug for HIV/AIDS, it had been noted to exhibit potent in vitro bactericidal activity against various bacteria of the family Enterobacteriaceae [36]. In Enterobacteriaceae, AZT is phosphorylated by thymidine kinase (when present) to its triphosphate metabolites, which are then incorporated into the bacterial DNA chain as a replication terminator [36]. Nonfermenters, such as Pseudomonas and Acinetobacter species, are unaffected by AZT because they naturally lack thymidine kinase [37]. In addition to the thymidine kinase levels of the bacteria, AZT susceptibility also correlates to cell permeability. Given that colistin lyses GNB by permeabilizing the outer membrane, it is hypothesized that this mechanism allows AZT to enter the cell at increased intracellular drug concentrations [38]. Lin et al. demonstrated that higher polymyxin B concentrations could increase outer membrane permeability, with simultaneous decreases in AZT MIC [33]. In addition, AZT can synergize with colistin by exerting a shared outer membrane disrupting effect [38].
The peak plasma AZT concentration (Cmax) of 1–5 μg/mL is achieved approximately 1 h after ingestion of 200–300 mg of AZT [39,40,41,42]. The pharmacokinetics (PK) of AZT has been most extensively investigated among HIV-infected individuals. In the works of Drew et al. [41] and Burger et al. [40], among HIV-infected patients, a Cmax of 0.73–1.3 and 0.8–1.1 μg/mL could be attained at 0.76 and 0.77 h, respectively. Wattanagoon et al. reported a Cmax of 17.98 μM (4.8 μg/mL) in healthy volunteers in Thailand after a single ingested dose of 300 mg AZT [39]. In agreement with a recent study [34], we found relatively low AZT MICs (0.03125–1 μg/mL) for colistin-resistant K. pneumoniae after combination with colistin, indicating that AZT could be a practical therapeutic candidate because of its clinically achievable concentrations. Loose et al. [37] investigated the serum bactericidal activity of combined intravenous (IV) colistin methanesulfonate (CMS) and AZT in colistin-resistant GNB (five isolates of mcr1-harboring E. coli). The trial was part of a phase 1 randomized, double-blinded study in healthy volunteers receiving multiple doses of IV CMS coadministrated with AZT. Seven volunteers received three IV infusions consisting of the following: first infusion, 4 million international units (MIU) of CMS and 200 mg of AZT; second and third infusions, 2 MIU of CMS and 100 mg of AZT. The study found the combination to be well tolerated, with only transient and manageable gastrointestinal side effects. It also revealed bactericidal, or at least bacteriostatic, activities for all strains tested. The results obtained with our in vivo C. elegans toxicity testing model are consistent with those reported by Loose et al. [37]. Although AZT appears to be important in the combined treatment of MDROs, the rapid emergence of stable high-level AZT resistance in Enterobacteriaceae has been well documented, which appears to be related to the loss of thymidine kinase activity [30,43]. Lin et al. demonstrated that the combination of polymyxin B and AZT has superior antimicrobial efficacy and minimizes the emergence of resistance to polymyxins [33]. The two-drug combination significantly increased bacterial killing and remained synergistic for up to 48 h in their study. It also significantly delayed bacterial regrowth compared with either monotherapy.

5. Conclusions

This study demonstrates the evident synergy between colistin and AZT in 40 clinical CCRKP isolates, regardless of the presence of carbapenemase. In vivo toxicity testing indicated low toxicity of the two-drug combination. The combined treatment also significantly increased the lifespan of C. elegans in the nematode killing assay. According to our findings, AZT could serve as a promising component in a combination regimen for CRE treatment. AZT has many beneficial characteristics, such as intravenous formulation, attainable clinical plasma concentration with effective central nervous system penetration, and known safety profiles for a wide range of populations, such as neonates or children and pregnant women. The AZT/colistin combination could be a potential therapeutic option for the treatment of CCRKP. Future investigation into the optimal dosage and frequency is necessary to achieve clinical efficacy and prevent the rapid emergence of resistance.

Supplementary Materials

The following are available online at https://www.mdpi.com/2076-2607/8/12/1964/s1, Table S1: Primers used in for PCR detection of antibiotic resistant genes.

Author Contributions

Conceptualization, Y.-J.H. and S.-P.T.; methodology, T.-Y.Y. and L.-C.W.; software, T.-Y.Y.; validation, T.-Y.Y., P.-L.L. and S.-Y.L.; formal analysis, Y.-T.C. and S.-F.W.; investigation, Y.-T.C. and T.-Y.Y.; resources, Y.-J.H. and S.-P.T.; data curation, Y.-J.H. and S.-P.T.; writing—original draft preparation, Y.-T.C., T.-Y.Y. and S.-P.T.; writing—review and editing, Y.-T.C. and S.-P.T.; visualization, S.-P.T.; supervision, Y.-J.H. and S.-P.T.; project administration, S.-P.T.; funding acquisition, Y.-J.H. and S.-P.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from Ministry of Science and Technology of Taiwan (MOST 109-2320-B-037-027), Kaohsiung Medical University Research Foundation (M109001), Kaohsiung Medical University Research Center Grant (KMU-TC109B02), and National Sun Yat-sen University-Kaohsiung Medical University (NSYSU-KMU) Joint Research Project (NSYSUKMU 109-I005).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marston, H.D.; Dixon, D.M.; Knisely, J.M.; Palmore, T.N.; Fauci, A.S. Antimicrobial Resistance. JAMA 2016, 316, 1193–1204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Global Priority List of Antibiotic-Resistant Bacteria to Guide Research, Discovery, and Development of New Antibiotics. Available online: https://www.who.int/medicines/publications/global-priority-list-antibiotic-resistant-bacteria/en/ (accessed on 11 November 2020).
  3. 2019 AR Threats Report. Available online: https://www.cdc.gov/drugresistance/biggest-threats.html?CDC_AA_refVal=https%3A%2F%2Fwww.cdc.gov%2Fdrugresistance%2Fbiggest_threats.html (accessed on 11 November 2020).
  4. Kelly, A.M.; Mathema, B.; Larson, E.L. Carbapenem-resistant Enterobacteriaceae in the community: A scoping review. Int. J. Antimicrob. Agents 2017, 50, 127–134. [Google Scholar] [CrossRef] [PubMed]
  5. 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] [PubMed] [Green Version]
  6. Bush, K.; Fisher, J.F. Epidemiological expansion, structural studies, and clinical challenges of new beta-lactamases from gram-negative bacteria. Annu. Rev. Microbiol. 2011, 65, 455–478. [Google Scholar] [CrossRef] [PubMed]
  7. Lasko, M.J.; Nicolau, D.P. Carbapenem-resistant Enterobacterales: Considerations for treatment in the era of new antimicrobials and evolving enzymology. Curr. Infect. Dis. Rep. 2020, 22, 6. [Google Scholar] [CrossRef] [PubMed]
  8. Tamma, P.D.; Goodman, K.E.; Harris, A.D.; Tekle, T.; Roberts, A.; Taiwo, A.; Simner, P.J. Comparing the outcomes of patients with carbapenemase-producing and non-carbapenemase-producing carbapenem-resistant Enterobacteriaceae bacteremia. Clin. Infect. Dis. 2017, 64, 257–264. [Google Scholar] [CrossRef] [Green Version]
  9. Chang, Y.T.; Siu, L.K.; Wang, J.T.; Wu, T.L.; Chen, Y.H.; Chuang, Y.C.; Lin, J.C.; Lu, P.L. Resistance mechanisms and molecular epidemiology of carbapenem-nonsusceptible Escherichia coli in Taiwan, 2012–2015. Infect. Drug Resist. 2019, 12, 2113–2123. [Google Scholar] [CrossRef] [Green Version]
  10. Rodriguez-Bano, J.; Gutierrez-Gutierrez, B.; Machuca, I.; Pascual, A. Treatment of infections caused by extended-spectrum-beta-lactamase-, ampC-, and carbapenemase-producing Enterobacteriaceae. Clin. Microbiol. Rev. 2018, 31. [Google Scholar] [CrossRef] [Green Version]
  11. Rojas, L.J.; Salim, M.; Cober, E.; Richter, S.S.; Perez, F.; Salata, R.A.; Kalayjian, R.C.; Watkins, R.R.; Marshall, S.; Rudin, S.D.; et al. Colistin resistance in carbapenem-resistant Klebsiella pneumoniae: Laboratory detection and impact on mortality. Clin. Infect. Dis. 2017, 64, 711–718. [Google Scholar] [CrossRef] [Green Version]
  12. Keith, B.R.; White, G.; Wilson, H.R. In vivo efficacy of zidovudine (3′-azido-3′-deoxythymidine) in experimental gram-negative-bacterial infections. Antimicrob. Agents Chemother. 1989, 33, 479–483. [Google Scholar] [CrossRef] [Green Version]
  13. Herrmann, J.L.; Lagrange, P.H. Intracellular activity of zidovudine (3′-azido-3′-deoxythymidine, AZT) against Salmonella typhimurium in the macrophage cell line J774-2. Antimicrob. Agents Chemother. 1992, 36, 1081–1085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Peyclit, L.; Baron, S.A.; Yousfi, H.; Rolain, J.M. Zidovudine: A salvage therapy for mcr-1 plasmid-mediated colistin-resistant bacterial infections? Int. J. Antimicrob. Agents 2018, 52, 11–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hind, C.K.; Dowson, C.G.; Sutton, J.M.; Jackson, T.; Clifford, M.; Garner, R.C.; Czaplewski, L. Evaluation of a library of FDA-approved drugs for their ability To potentiate antibiotics against multidrug-resistant Gram-negative pathogens. Antimicrob. Agents Chemother. 2019, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ng, S.M.S.; Sioson, J.S.P.; Yap, J.M.; Ng, F.M.; Ching, H.S.V.; Teo, J.W.P.; Jureen, R.; Hill, J.; Chia, C.S.B. Repurposing zidovudine in combination with tigecycline for treating carbapenem-resistant Enterobacteriaceae infections. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 141–148. [Google Scholar] [CrossRef] [PubMed]
  17. Chiu, S.K.; Ma, L.; Chan, M.C.; Lin, Y.T.; Fung, C.P.; Wu, T.L.; Chuang, Y.C.; Lu, P.L.; Wang, J.T.; Lin, J.C.; et al. Carbapenem nonsusceptible Klebsiella pneumoniae in Taiwan: Dissemination and increasing resistance of carbapenemase producers during 2012–2015. Sci. Rep. 2018, 8, 8468. [Google Scholar] [CrossRef] [Green Version]
  18. Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility Testing: Thirtieth Informational Supplement; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2020; Document M100-S30 CLSI. [Google Scholar]
  19. Clinical Breakpoints and Dosing of Antibiotics. Available online: https://www.eucast.org/clinical_breakpoints/ (accessed on 11 November 2020).
  20. Tseng, S.P.; Wang, S.F.; Ma, L.; Wang, T.Y.; Yang, T.Y.; Siu, L.K.; Chuang, Y.C.; Lee, P.S.; Wang, J.T.; Wu, T.L.; et al. The plasmid-mediated fosfomycin resistance determinants and synergy of fosfomycin and meropenem in carbapenem-resistant Klebsiella pneumoniae isolates in Taiwan. J. Microbiol. Immunol. Infect. 2017, 50, 653–661. [Google Scholar] [CrossRef]
  21. Yang, T.Y.; Wang, S.F.; Lin, J.E.; Griffith, B.T.S.; Lian, S.H.; Hong, Z.D.; Lin, L.; Lu, P.L.; Tseng, S.P. Contributions of insertion sequences conferring colistin resistance in Klebsiella pneumoniae. Int. J. Antimicrob. Agents 2020, 55, 105894. [Google Scholar] [CrossRef]
  22. Kakuda, T.N.; Page, L.M.; Anderson, P.L.; Henry, K.; Schacker, T.W.; Rhame, F.S.; Acosta, E.P.; Brundage, R.C.; Fletcher, C.V. Pharmacological basis for concentration-controlled therapy with zidovudine, lamivudine, and indinavir. Antimicrob. Agents Chemother. 2001, 45, 236–242. [Google Scholar] [CrossRef] [Green Version]
  23. Lutgring, J.D. Carbapenem-resistant Enterobacteriaceae: An emerging bacterial threat. Semin. Diagn. Pathol. 2019, 36, 182–186. [Google Scholar] [CrossRef] [PubMed]
  24. Porreca, A.M.; Sullivan, K.V.; Gallagher, J.C. The epidemiology, evolution, and treatment of KPC-producing organisms. Curr. Infect. Dis. Rep. 2018, 20, 13. [Google Scholar] [CrossRef] [PubMed]
  25. Kumar, M. Colistin and tigecycline resistance in carbapenem-resistant Enterobacteriaceae: Checkmate to our last line of defense. Infect. Control. Hosp. Epidemiol. 2016, 37, 624–625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Qamar, S.; Shaheen, N.; Shakoor, S.; Farooqi, J.; Jabeen, K.; Hasan, R. Frequency of colistin and fosfomycin resistance in carbapenem-resistant Enterobacteriaceae from a tertiary care hospital in Karachi. Infect. Drug Resist. 2017, 10, 231–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Li, Y.; Sun, Q.L.; Shen, Y.; Zhang, Y.; Yang, J.W.; Shu, L.B.; Zhou, H.W.; Wang, Y.; Wang, B.; Zhang, R.; et al. Rapid increase in prevalence of carbapenem-resistant Enterobacteriaceae (CRE) and emergence of colistin resistance gene mcr-1 in CRE in a hospital in Henan, China. J. Clin. Microbiol. 2018, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Du, H.; Chen, L.; Tang, Y.W.; Kreiswirth, B.N. Emergence of the mcr-1 colistin resistance gene in carbapenem-resistant Enterobacteriaceae. Lancet Infect. Dis. 2016, 16, 287–288. [Google Scholar] [CrossRef] [Green Version]
  29. Malchione, M.D.; Torres, L.M.; Hartley, D.M.; Koch, M.; Goodman, J.L. Carbapenem and colistin resistance in Enterobacteriaceae in Southeast Asia: Review and mapping of emerging and overlapping challenges. Int. J. Antimicrob. Agents 2019, 54, 381–399. [Google Scholar] [CrossRef]
  30. Doleans-Jordheim, A.; Bergeron, E.; Bereyziat, F.; Ben-Larbi, S.; Dumitrescu, O.; Mazoyer, M.A.; Morfin, F.; Dumontet, C.; Freney, J.; Jordheim, L.P. Zidovudine (AZT) has a bactericidal effect on enterobacteria and induces genetic modifications in resistant strains. Eur. J. Clin. Microbiol. Infect. Dis. 2011, 30, 1249–1256. [Google Scholar] [CrossRef]
  31. Jordheim, L.P.; Durantel, D.; Zoulim, F.; Dumontet, C. Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. Nat. Rev. Drug Discov. 2013, 12, 447–464. [Google Scholar] [CrossRef]
  32. Zhou, Y.F.; Liu, P.; Dai, S.H.; Sun, J.; Liu, Y.H.; Liao, X.P. Activity of tigecycline or colistin in combination with zidovudine against Escherichia coli harboring tet(X) and mcr-1. Antimicrob. Agents Chemother. 2020. [Google Scholar] [CrossRef]
  33. Lin, Y.W.; Abdul Rahim, N.; Zhao, J.; Han, M.L.; Yu, H.H.; Wickremasinghe, H.; Chen, K.; Wang, J.; Paterson, D.L.; Zhu, Y.; et al. Novel polymyxin combination with the antiretroviral zidovudine exerts synergistic killing against NDM-producing multidrug-resistant Klebsiella pneumoniae. Antimicrob. Agents Chemother. 2019, 63, e02176-18. [Google Scholar] [CrossRef] [Green Version]
  34. Falagas, M.E.; Voulgaris, G.L.; Tryfinopoulou, K.; Giakkoupi, P.; Kyriakidou, M.; Vatopoulos, A.; Coates, A.; Hu, Y.; Colistin—Azidothymidine Hellenic Study Group. Synergistic activity of colistin with azidothymidine against colistin-resistant Klebsiella pneumoniae clinical isolates collected from inpatients in Greek hospitals. Int. J. Antimicrob. Agents 2019, 53, 855–858. [Google Scholar] [CrossRef]
  35. Hu, Y.; Liu, Y.; Coates, A. Azidothymidine produces synergistic activity in combination with colistin against antibiotic-resistant Enterobacteriaceae. Antimicrob. Agents Chemother. 2019, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Elwell, L.P.; Ferone, R.; Freeman, G.A.; Fyfe, J.A.; Hill, J.A.; Ray, P.H.; Richards, C.A.; Singer, S.C.; Knick, V.B.; Rideout, J.L.; et al. Antibacterial activity and mechanism of action of 3′-azido-3′-deoxythymidine (BW A509U). Antimicrob. Agents Chemother. 1987, 31, 274–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Loose, M.; Naber, K.G.; Hu, Y.; Coates, A.; Wagenlehner, F.M.E. Serum bactericidal activity of colistin and azidothymidine combinations against mcr-1-positive colistin-resistant Escherichia coli. Int. J. Antimicrob. Agents 2018, 52, 783–789. [Google Scholar] [CrossRef] [PubMed]
  38. Schneider, E.K.; Reyes-Ortega, F.; Velkov, T.; Li, J. Antibiotic-non-antibiotic combinations for combating extremely drug-resistant Gram-negative ‘superbugs’. Essays Biochem. 2017, 61, 115–125. [Google Scholar] [PubMed]
  39. Wattanagoon, Y.; Na Bangchang, K.; Hoggard, P.G.; Khoo, S.H.; Gibbons, S.E.; Phiboonbhanakit, D.; Karbwang, J.; Back, D.J. Pharmacokinetics of zidovudine phosphorylation in human immunodeficiency virus-positive thai patients and healthy volunteers. Antimicrob. Agents Chemother. 2000, 44, 1986–1989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Burger, D.M.; Meenhorst, P.L.; ten Napel, C.H.; Mulder, J.W.; Neef, C.; Koks, C.H.; Bult, A.; Beijnen, J.H. Pharmacokinetic variability of zidovudine in HIV-infected individuals: Subgroup analysis and drug interactions. AIDS 1994, 8, 1683–1689. [Google Scholar] [CrossRef] [PubMed]
  41. Drew, R.H.; Weller, S.; Gallis, H.A.; Walmer, K.A.; Bartlett, J.A.; Blum, M.R. Bioequivalence assessment of zidovudine (Retrovir) syrup, solution, and capsule formulations in patients infected with human immunodeficiency virus. Antimicrob. Agents Chemother. 1989, 33, 1801–1803. [Google Scholar] [CrossRef] [Green Version]
  42. Hoetelmans, R.M.; Burger, D.M.; Meenhorst, P.L.; Beijnen, J.H. Pharmacokinetic individualisation of zidovudine therapy. Current state of pharmacokinetic-pharmacodynamic relationships. Clin. Pharmacokinet. 1996, 30, 314–327. [Google Scholar] [CrossRef]
  43. Lewin, C.S.; Allen, R.; Amyes, S.G. Zidovudine-resistance in Salmonella typhimurium and Escherichia coli. J. Antimicrob. Chemother. 1990, 25, 706–708. [Google Scholar] [CrossRef]
Figure 1. Distribution of the examined resistance mechanisms among the 40 isolates. (a) non-KPC producers; #, number of simultaneous resistance mechanisms (number of isolates) (b) KPC producers, with one isolate lacking data for outer membrane proteins.
Figure 1. Distribution of the examined resistance mechanisms among the 40 isolates. (a) non-KPC producers; #, number of simultaneous resistance mechanisms (number of isolates) (b) KPC producers, with one isolate lacking data for outer membrane proteins.
Microorganisms 08 01964 g001
Figure 2. MIC distributions of colistin (COL, hollow circles), azidothymidine (AZT, hollow circles), and their combination (filled circles). (a) Dotted line indicates the breakpoint for colistin resistance according to the EUCAST guidelines. (b) Dotted line represents the average steady-state concentration of AZT in plasma (0.19 μg/mL). ****, p < 0.0001 with the paired t-test.
Figure 2. MIC distributions of colistin (COL, hollow circles), azidothymidine (AZT, hollow circles), and their combination (filled circles). (a) Dotted line indicates the breakpoint for colistin resistance according to the EUCAST guidelines. (b) Dotted line represents the average steady-state concentration of AZT in plasma (0.19 μg/mL). ****, p < 0.0001 with the paired t-test.
Microorganisms 08 01964 g002
Figure 3. In vivo toxicity testing of colistin, AZT, or colistin/AZT combination using a C. elegans model. Nematodes (n = 40, each group) were fed with nontoxic E. coli laboratory strain OP50 and supplemented with a vehicle (control), colistin (COL, at 1 μg/mL), azidothymidine (AZT, at 0.15 μg/mL), or a combination (AZT + COL, at 0.15/1 μg/mL). ns, no significance.
Figure 3. In vivo toxicity testing of colistin, AZT, or colistin/AZT combination using a C. elegans model. Nematodes (n = 40, each group) were fed with nontoxic E. coli laboratory strain OP50 and supplemented with a vehicle (control), colistin (COL, at 1 μg/mL), azidothymidine (AZT, at 0.15 μg/mL), or a combination (AZT + COL, at 0.15/1 μg/mL). ns, no significance.
Microorganisms 08 01964 g003
Figure 4. In vivo killing assay of CCRKP strain 1336 using a C. elegans model. Forty nematodes were infected with CCRKP clinical strain 1336 (isolated from blood) and supplemented with a vehicle (control), colistin (COL, at 1 μg/mL), azidothymidine (AZT, at 0.15 μg/mL), or a combination (AZT + COL, at 0.15/1 μg/mL). ns, no significance; ****, p < 0.0001.
Figure 4. In vivo killing assay of CCRKP strain 1336 using a C. elegans model. Forty nematodes were infected with CCRKP clinical strain 1336 (isolated from blood) and supplemented with a vehicle (control), colistin (COL, at 1 μg/mL), azidothymidine (AZT, at 0.15 μg/mL), or a combination (AZT + COL, at 0.15/1 μg/mL). ns, no significance; ****, p < 0.0001.
Microorganisms 08 01964 g004
Table 1. Sources of the 40 isolates.
Table 1. Sources of the 40 isolates.
SourceNo. of Isolates
Urine11
Sputum10
Stool4
Pus/wound3
Abscess3
Blood2
Endotracheal 2
Ascites2
Bile2
Drainage1
Table 2. Antimicrobial susceptibilities against 19 common antimicrobial agents.
Table 2. Antimicrobial susceptibilities against 19 common antimicrobial agents.
Antimicrobial AgentsAntibiotic Susceptibility
SIR
Ampicillin 0.0%0.0%100.0%
Ceftazidime0.0%2.5%97.5%
Cefazolin 0.0%0.0%100.0%
Cefepime 5.0%7.5%87.5%
Cefoxitin 2.5%2.5%95.0%
Ceftriaxone 0.0%0.0%100.0%
Cefotaxime 2.5%0.0%97.5%
Imipenem 2.5%10.0%87.5%
Meropenem 7.5%0.0%92.5%
Doripenem 7.5%2.5%90.0%
Ertapenem 0.0%2.5%97.5%
Aztreonam7.5%2.5%90.0%
Piperacillin/Tazobactam5.0%0.0%95.0%
Tigecycline 87.5%10.0%2.5%
Ciprofloxacin 2.5%0.0%97.5%
Levofloxacin 2.5%0.0%97.5%
Gentamicin32.5%0.0%67.5%
Amikacin70.0%0.0%30.0%
Sulfamethoxazole-Trimethoprim12.5%0.0%87.5%
Table 3. MIC range, MIC50, MIC75, and MIC90 for colistin, AZT, and colistin/AZT combination.
Table 3. MIC range, MIC50, MIC75, and MIC90 for colistin, AZT, and colistin/AZT combination.
AgentsMIC (μg/mL)MIC in Combination (μg/mL)
RangeMIC50MIC75MIC90RangeMIC50MIC75MIC90
Colistin4–51264641281–2112
AZT 0.125–161220.03125–10.1250.1250.25
MIC, minimum inhibitory concentration; AZT, azidothymidine.
Table 4. In vitro synergistic analysis using the checkerboard method.
Table 4. In vitro synergistic analysis using the checkerboard method.
GroupActivity of the CombinationFICI CriteriaTotal No. (%) of Isolates
CCRKP (n = 40)KPC-producer (n = 11)synergy≤0.511 (100.0%)
No interaction>0.5–40
Antagonism>40
non-KPC-producer (n = 29)synergy≤0.529 (100.0%)
No interaction>0.5–10
Antagonism>40
FICI, fractional inhibitory concentration index; KPC, Klebsiella pneumoniae carbapenemase.
Table 5. Statistical analyses of in vivo C. elegans toxicity testing and killing assays.
Table 5. Statistical analyses of in vivo C. elegans toxicity testing and killing assays.
TestGroupMedian Survival
(Days)
p ValueRisk Ratio95% CI
LowerUpper
In vivo
toxicity assay
OP50-Control9-1--
OP50-AZT90.79010.9340.571.54
OP50-COL80.45730.8270.501.36
OP50-AZT + COL90.61771.130.691.87
In vivo
killing assay
OP50-Control9<0.00010.270.160.47
1336-Control6-1--
1336-AZT60.72541.0970.651.84
1336-COL60.85610.9530.561.61
1336-AZT + COL8.5<0.00010.2880.170.50
OP50, laboratory strain of nontoxic Escherichia coli OP50 as a food source for nematodes (representing the negative control); COL, colistin (concentration of 1 μg/mL); AZT, azidothymidine (concentration of 0.15 μg/mL); AZT + COL, combination (concentration of 0.15/1 μg/mL); 1336, clinical isolate of CCRKP; CI, confidence interval.
Table 6. Combination therapy against Enterobacteriaceae in different countries.
Table 6. Combination therapy against Enterobacteriaceae in different countries.
SpeciesCountryNo.Resistance PhenotypeResistance Mechanism(s)MIC of AZT (mg/L)MIC of Colistin (mg/L)Ref.
AloneCombinationAloneCombination
K. pneumoniaeGreece100ColistinNA0.125–40.0625–14–1280.25–16[34]
E. coliChina9Colistin
and tigecycline
tet(X) and
mcr-1
0.5–40.3–1.54–8<0.13–4[32]
K. pneumoniae,
E. coli, E. cloacae
UK7CarbapenemblaNDM-12–40.25–160.125–1Reduced
32–256 fold
[35]
E. coliUK13Colistinmcr-18–642–8Reduced
4–256 fold
[35]
K. pneumoniaeTaiwan40Colistin
and carbapenem
multiple,
blaKPC
0.125–160.03125–14–5121–2Present study
NA: not available; tet(X), a unique mobile tigecycline resistance gene [32].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chang, Y.-T.; Yang, T.-Y.; Lu, P.-L.; Lin, S.-Y.; Wang, L.-C.; Wang, S.-F.; Hsieh, Y.-J.; Tseng, S.-P. Combination of Colistin and Azidothymidine Demonstrates Synergistic Activity against Colistin-Resistant, Carbapenem-Resistant Klebsiella pneumoniae. Microorganisms 2020, 8, 1964. https://doi.org/10.3390/microorganisms8121964

AMA Style

Chang Y-T, Yang T-Y, Lu P-L, Lin S-Y, Wang L-C, Wang S-F, Hsieh Y-J, Tseng S-P. Combination of Colistin and Azidothymidine Demonstrates Synergistic Activity against Colistin-Resistant, Carbapenem-Resistant Klebsiella pneumoniae. Microorganisms. 2020; 8(12):1964. https://doi.org/10.3390/microorganisms8121964

Chicago/Turabian Style

Chang, Ya-Ting, Tsung-Ying Yang, Po-Liang Lu, Shang-Yi Lin, Liang-Chun Wang, Sheng-Fan Wang, Ya-Ju Hsieh, and Sung-Pin Tseng. 2020. "Combination of Colistin and Azidothymidine Demonstrates Synergistic Activity against Colistin-Resistant, Carbapenem-Resistant Klebsiella pneumoniae" Microorganisms 8, no. 12: 1964. https://doi.org/10.3390/microorganisms8121964

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