Eur J Clin Microbiol Infect Dis https://doi.org/10.1007/s10096-017-3114-5

ORIGINAL ARTICLE

Repurposing Zidovudine in combination with Tigecycline for treating carbapenem-resistant Enterobacteriaceae infections S. M. S. Ng 1 & J. S. P. Sioson 1 & J. M. Yap 1 & F. M. Ng 1 & H. S. V. Ching 1 & J. W. P. Teo 2 & R. Jureen 2 & J. Hill 1 & C. S. B. Chia 1

Received: 22 August 2017 / Accepted: 27 September 2017 # Springer-Verlag GmbH Germany 2017

Abstract The global emergence of carbapenem-resistant Enterobacteriaceae (CRE) presents a significant clinical concern, prompting the WHO to prioritize CRE as a top priority pathogen in their 2017 global antibiotic-resistant bacteria priority list. Due to the fast-depleting antibiotic arsenal, clinicians are now resorting to using once-abandoned, highly toxic antibiotics such as the polymyxins and aminoglycosides, creating an urgent need for new antibiotics. Drug repurposing, the application of an approved drug for a new therapeutic indication, is deemed a plausible solution to this problem. A total of 1,163 FDA-approved drugs were screened for activity against a clinical carbapenem- and multidrug-resistant E. coli isolate using a single-point 10 μM assay. Hit compounds were then assessed for their suitability for repurposing. The lead candidate was then tested against a panel of clinical CREs, a bactericidal/static determination assay, a time-kill assay and a checkerboard assay to evaluate its suitability for use in combination with Tigecycline against CRE infections. Three drugs were identified. The lead candidate was determined to be Zidovudine (azidothymidine/AZT), an oral anti-viral drug used for HIV treatment. Zidovudine was shown to be the most promising candidate for use in combination with Tigecycline to treat systemic CRE infections. Further experiments should involve the use of animal infection models.

S. M. S. Ng, J. S. P. Sioson and J. M. Yap contributed equally to this work. * C. S. B. Chia [email protected] 1

Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos #03-01, Singapore 138669, Singapore

2

Department of Laboratory Medicine, National University Hospital, 5 Lower Kent Ridge Road, Singapore 119074, Singapore

Keywords Drug repurposing . Antibiotic . Carbapenem-resistant Enterobacteriaceae . Zidovudine

Introduction Escherichia coli and Klebsiella pneumoniae are Enterobacteriaceae responsible for causing various human infections such as bacteremia, meningitis and pneumonia [1, 2]. Standard treatment involve the use of carbapenems, widely considered as last-line antibiotics with favorable safety profiles [3–7]. However, their extensive use has led to the emergence of carbapenem-resistant Enterobacteriaceae (CRE), presenting a global health threat [1, 8–15]. Carbapenem resistance is caused by a variety of mechanisms. Of particular concern are carbapenemase-producing Enterobacteriaceae where their plasmid-borne carbapenemase genes are easily transferred among Enterobacteriaceae, resulting in rapid dissemination [3, 16–18]. This, coupled to the dearth of new antibiotics entering the market has created a global concern, prompting the US CDC and the WHO to redflag CRE as a high-priority pathogen [19, 20]. Treatment options for CRE infections is limited [21–24]. This includes the off-label use of Tigecycline, a tetracycline analog originally indicated for skin and soft tissue infections [21, 22, 25–27]. Since its introduction in 2005, Tigecyclineresistant CRE strains have emerged [28–30] and in 2010, the FDA issued a black box warning due to an increased incidence of deaths in patients on Tigecycline therapy [31–33]. The lack of new antibiotics has prompted clinicians to resort to old antibiotics like the polymyxins (e.g. Colistin) and aminoglycosides (e.g. Gentamicin) which were abandoned in favor of new antibiotics with better safety profiles like the quinolones (e.g. Ciprofloxacin) and the carbapenems (e.g. Meropenem) [22, 24, 26, 34–36]. For example, Colistin,

Eur J Clin Microbiol Infect Dis

introduced in 1958, is nephrotoxic and neurotoxic [21, 23, 26, 37–40], while Gentamicin, introduced in 1963, is nephrotoxic and ototoxic [26, 37, 41–44]. Furthermore, the past use of these old antibiotics has also resulted in the emergence of drug resistance, further amplifying the need for new antibiotics [45–56]. The dearth of novel antibiotics entering the market has been attributed to regulatory hurdles, high research costs and low investment returns [57–60]. A possible solution is drug repurposing: the application of an approved drug for a new therapeutic indication [61–63]. Since the safety, pharmacological profile and manufacturing process of an approved drug is known, it can be rapidly made available for a new disease indication [62]. Based on this, we screened a library of 1,163 FDA-approved drugs for antibacterial activity against carbapenem-resistant E. coli using a single-concentration (10 μM) assay. After identifying hit compounds and removing known antibiotics, the remaining compounds were subjected to a minimum inhibitory concentration (MIC) assay followed by a toxicity and pharmacological assessment to determine their suitability for repurposing as an antibiotic. Next, the most promising candidate was tested against a CRE panel, followed by a bactericidal/ static determination assay and a time-kill assay. Lastly, a combination (checkerboard) assay was conducted with Tigecycline to determine synergism and suitability for repurposing as an antibiotic to treat CRE infections.

Materials and methods

containing the test compounds in duplicates. The final DMSO concentration was kept at 2.5% in each well. After an overnight incubation at 35 °C, optical density (OD600) of e ac h w el l w a s d et er m i n ed u s i n g t h e M i c r o p l a t e Spectrophotometer (Molecular Devices Spectra Max Plus, USA). Minimum inhibitory concentration (MIC) determination assay MICs of antibiotics and test compounds were determined using the microdilution method from the Clinical and Laboratory Standards Institute (CLSI) guidelines [64]. Briefly, bacteria were grown fresh from frozen stock in MH2 agar at 37 °C. After an overnight incubation, five bacteria colonies were selected to grow in cation-adjusted MH2 broth (BD Biosciences) in a shaker incubator at 37 °C and 220 RPM. Cells were grown to an optical density (OD600) of 0.28– 0.30 using a Microplate Spectrophotometer (Molecular Devices Spectra Max Plus), which corresponds to 1– 2 × 108 CFU/mL. Test compounds were constituted into 4 mM DMSO stock solutions and then subjected to two-fold serial dilution in a 96-well plate with concentrations ranging from 100 to 0.2 μM in duplicates. The amount of 50 μL of microbial culture containing ~1–2 × 106 CFU/mL of microbes in the respective broths was introduced into each well containing 50 μL of compound. After an overnight incubation at 35 °C, OD600 measurements were conducted using the Microplate Spectrophotometer. The MIC was defined as the lowest compound concentration (μM) required to stop bacterial growth.

Drug library, antibiotics and bacteria Bactericidal/static determination assay The FDA-approved drug library was purchased from Selleckchem (catalog #L1300). All antibiotics listed in Table 1 were purchased from Sigma-Aldrich. Bacteria were either purchased from ATCC (USA) or obtained from the Singapore National University Hospital (NUH) clinical microbiology unit. Single-concentration bacteria growth inhibition screening assay All bacteria were grown from frozen glycerol stock on Mueller Hinton 2 (MH2) agar overnight at 37 °C. Five colonies were selected from the agar plate and grown in cationadjusted MH2 broth (BD Biosciences) in a shaker incubator at 37 °C and 220 RPM. Cells were grown to 1–2 × 108 CFU/mL and diluted 1:100 in MH2 broth. 50 μL of test compounds pre-dissolved in MH2 broth at 20 μM concentrations were added in duplicates into each well in a 96-well plate. A total of 50 μL of bacteria inoculums containing ~1 × 106 CFU/mL of bacteria in MH2 broth was introduced into each well

After the MICs of test compounds were determined using the microdilution method described earlier, the entire well contents (100 μL) corresponding to 1×, 2×, 4× and 8× the MICs of the compound were transferred and spread on fresh sterile MH2 agar plates. The plates were incubated overnight at 37 °C and the number of CFU were observed the next day. The minimum bactericidal concentration (MBC) of a compound was defined as the lowest compound concentration required to cause ≥99.99% cell death within 24 h and a compound was classified as bactericidal if its MBC lies between 1× to 4× of its MIC [65]. Time-kill assay Bacteria cells were grown to 1 × 108 CFU/mL in MH2 broth and then diluted 200-fold into 5 mL aliquots to a density of 5 × 105 CFU/mL in a flask. Test compounds pre-dissolved in DMSO corresponding to 4× their MICs were then added into each flask with a final DMSO concentration of 2.5% v/v. The

Eur J Clin Microbiol Infect Dis Table 1

MICs (μM) of hit compounds and FDA-approved antibiotics against carbapenem-resistant E. coli (ATCC-BAA-2469)

Hit compounds

Original indication

Administration route(s)

MIC (μM)

MBC (μM)

Azacitidine

Cancer

Intravenous, subcutaneous

6.25

50

Floxuridine

Cancer

Intraarterial

6.25

50

Zidovudine

Anti-viral

Intravenous, oral

3.125

6.25

Colistin

Antibiotic

Intramuscular, intravenous, topical

0.78

0.78

Ciprofloxacin Gentamicin

Antibiotic Antibiotic

Ophthalmic, oral Intramuscular, intravenous, ophthalmic, topical

>100 >100

Not applicable Not applicable

Meropenem Tigecycline

Antibiotic Antibiotic

Intravenous Intravenous

>100 0.78

Not applicable 3.125

Approved antibiotics

negative control was 2.5% DMSO. The flasks were incubated in a shaking incubator (220 RPM) at 37 °C. Samples (100 μL) were taken hourly from the flasks between t = 0 and t = 6 h. The samples were serially diluted in MH2 broth before plating on sterile MH2 agar and incubated overnight at 37 °C. A bacteria count for each plate was conducted the next day. A graph of Log CFU/mL was plotted against time to obtain the time-kill graph. Combination (checkerboard) assay Antibiotic synergism against carbapenem-resistant E. coli (ATCC-BAA-2469) was determined using the checkerboard method described previously [66]. Test compounds were tested over a concentration range: Tigecycline (0.00–6.25 μM), Zidovudine (0.00–12.5 μM). 50 mL of 1 × 106 CFU/mL of E. coli were inoculated into the wells to make a final volume of 100 μL containing 5 × 105 CFU/mL per well followed by an overnight incubation at 37 °C. The fractional inhibitory concentration index (FICI) was calculated for all the clear interface wells using the formula: FICI = FICT + FICZ where FICT is the MIC of Tigecycline used in combination/MIC of Tigecycline alone and FICZ is the MIC of Zidovudine used in combination/MIC of Zidovudine alone. A particular drug combination is considered synergistic when the FICI is ≤0.5; indifferent when the FICI is >0.5 but ≤4; and antagonistic when the FICI is >4 [66].

MIC determination The MICs for all three hit compounds against carbapenemresistant E. coli (ATCC-BAA-2469) ranged between 3.125 and 6.25 μM (Table 1). In comparison, FDA-approved antibiotics Colistin and Tigecycline were found to be equipotent (MICs 0.78 μM), in agreement with a previous report [67] while Ciprofloxacin, Gentamicin and Meropenem were found to be inactive (MICs >100 μM). Bactericidal/static determination assay The three hit compounds were subjected to a bactericidal/ static determination assay with Colistin and Tigecycline as 1x MIC

2x MIC

4x MIC

8x MIC

Azacitidine

Floxuridine

Zidovudine

Colistin

Results Drug library screen

Tigecycline

The 1163 drug library screen yielded 11 hits. After removing known antibiotics, three antibacterial compounds were identified: Azacitidine, Floxuridine and Zidovudine.

Fig. 1 Overnight growth of carbapenem-resistant E. coli (ATCC-BAA2469) after treatment with Azacitidine, Floxuridine, Zidovudine, Colistin and Tigecycline at 1×, 2×, 4× and 8× MIC

Log (CFU/ml)

Eur J Clin Microbiol Infect Dis 9 8 7 6 5 4 3 2 1 0

DMSO

Combination (checkerboard) assay

Tigecycline 4x MIC

Zidovudine 4x MIC Colistin 4x MIC 0

1

2

3

4

5

6

A checkerboard assay was conducted based on an established protocol using Zidovudine and Tigecycline [66]. Colistin was not considered due to its toxic side-effects [21, 26, 37–40]. Results revealed Zidovudine and Tigecycline to be indifferent at various concentrations with FICI values ranging between 0.63 and 1.25 (Fig. 3).

Time (h.)

Fig. 2 Time-kill assay of Colistin, Tigecycline and Zidovudine using carbapenem-resistant E. coli (ATCC-BAA-2469)

Discussion comparators (Fig. 1). The MBCs for Azacitidine and Floxuridine were found to be 50 μM (8× MIC), indicating both were bacteriostatic while Zidovudine was 6.25 μM (2× MIC), indicating that it was bactericidal [65]. The MBCs of Colistin and Tigecycline were 0.78 and 3.125 μM (1× and 4× MIC respectively), indicating both were also bactericidal (Table 1).

By screening a 1163 drug library, 11 FDA-approved drugs were identified to be active against carbapenem-resistant E. coli. After excluding known antibiotics, three drugs: Azacitidine, Floxuridine and Zidovudine were identified for further investigation (MICs 6.25 μM; Table 1). Azacitidine

CRE panel MICs The bioactivity of Zidovudine against a panel of E. coli and K. pneumoniae clinical isolates is summarized in Table 2. Zidovudine, Colistin and Tigecycline were found to be active against all clinical isolates with MICs ranging from 0.39 to 6.25 μM. In contrast, Ciprofloxacin and Gentamicin were inactive (MIC ≥50 μM) against some isolates. Time-kill assay E. coli killing kinetics of Zidovudine was performed using a time-kill assay following a previous protocol [68]. Results revealed Zidovudine to be rapidly bactericidal, achieving a 6-log kill within 3 h (Fig. 2). Table 2

Azacitidine (5-azacytidine/5AZC/Vidaza™) is a nucleoside analogue approved in 2004 for treating various blood cancers [69–71]. The recommended daily dose is 75 mg/m2 (~120 mg for a 60 kg adult) for 7 days [69, 72]. After intravenous administration, Azacitidine is rapidly hydrolysed with a plasma half-life of ~22 min [69, 73]. In comparison, Colistin, in its colistimethate pro-drug form, and Tigecycline have plasma half-lives exceeding 4 and 37 h, respectively [74, 75]. In our view, Azacitidine’s short half-life is unsuitable for repurposing as an antibacterial agent. In addition, the major concern using Azacitidine as an antibacterial agent is its myelosuppressive side-effect manifested by neutropenia which diminishes a patient’s ability to fight bacterial infections [69]. Indeed, the European Medicines Agency (EMA) reported patients on

MICs (μM) of FDA-approved antibiotics and zidovudine against various Enterobacteriaceae clinical isolates

Bacteria

ATCC or NUH number

Ciprofloxacin

Colistin

Gentamicin

Meropenem

Tigecycline

Zidovudine

E. coli E. coli E. coli E. coli E. coli E. coli E. coli K. pneumoniae

BAA-2355 BAA-2340 BAA-2469 BAA-2523 NUH-376 NUH-1289 NUH-5684 700603

>100 >100 >100 0.78 50 >100 25 0.78

0.78 0.78 0.78 0.78 3.125 0.78 0.78 0.78

>100 3.125 >100 >100 >100 1.56 1.56 12.5

0.39 25 >100 3.125 >100 >100 50 0.39

0.78 0.39 0.78 0.78 1.56 1.56 1.56 6.25

3.125 3.125 3.125 1.56 6.25 1.56 0.39 6.25

K. pneumoniae K. pneumoniae K. pneumoniae K. pneumoniae K. pneumoniae

BAA-2342 BAA-2524 NUH-34 NUH-2751 NUH-6751

>100 0.2 >100 >100 0.2

1.56 0.39 1.56 1.56 0.78

12.5 0.78 >100 0.78 3.125

>100 25 >100 >100 50

3.125 3.125 3.125 6.25 0.78

3.125 6.25 6.25 6.25 3.125

0.78 0.39

1.06

1.00

0.20

0.75

1.25

0.10

0.63

1.13

0.00

Tigecycline ( M)

1.56

3.13

6.25

Eur J Clin Microbiol Infect Dis

0.00

0.20

0.39

0.78 1.56 Zidovudine ( M)

3.13

6.25

12.5

Fig. 3 Schematic diagram of the Tigecycline-Zidovudine checkerboard assay using carbapenem-resistant E. coli (ATCC-BAA-2469). Grey areas represent cloudy wells (viable bacteria) while white areas represent clear wells. Numbers in bold text represent the FICI value of each individual well. A drug combination is classified synergistic when its FICI is ≤0.5

Azacitidine treatment very commonly contracted pneumonia (≥1 in 10) in their public assessment report [76]. Lastly, Azacitidine was observed to be bacteriostatic against E. coli. (Fig. 1). With these unfavorable characteristics, Azacitidine lacks potential for repurposing as an antibacterial drug. Floxuridine Floxuridine (5-fluorodeoxyuridine/5FDU/FUDR®) is an intraarterially-administered nucleoside analog approved in 1970 for treating colorectal liver metastases and has been reported to act synergistically with Zidovudine against Enterobacteriaceae [77, 78]. Floxuridine inhibits thymidylate synthase, blocking thymidine production and consequently stopping DNA and RNA synthesis, resulting in cell death [77]. The drug is highly toxic with a recommended daily dose of 0.3 mg/kg (18 mg for a 60 kg adult) over 14 days [79] and possesses a very short plasma half-life of ~15 min [80]. Like Azacitidine, it is myelosuppressive and was observed to be bacteriostatic against E. coli. (Fig. 1). Based on these unfavorable characteristics, Floxuridine lacks potential for repurposing as an antibacterial drug. Zidovudine Zidovudine (azidothymidine/AZT/Retrovir®) is an oral or intravenously-administered nucleoside analog approved in 1987 for treating human immunodeficiency virus (HIV) infection [81, 82]. In an infected cell, Zidovudine is phosphorylated

by cellular kinases to form Zidovudine triphosphate which functions as an HIV reverse transcriptase inhibitor, arresting viral DNA production and ultimately viral replication [81–84]. Zidovudine was found to be bactericidal against Enterobacteriaceae with reported MICs against E. coli and K. pneumoniae of 0.0025 to 1.0 μg/mL (0.01 to 3.7 μM) and 0.025 to 3.1 μg/mL (0.1 to 11.6 μM) [85, 86], in agreement with our experimental results (Table 2 and Fig. 1). Zidovudine’s antibacterial activity is thought to be derived from its ability to act as a chain terminator following bacterial DNA incorporation after being phosphorylated by bacterial kinases [85, 86]. Compared to Azacitidine and Floxuridine, Zidovudine has a much better safety profile with a recommended daily dose of 600 mg over a patient’s life-time due to its short plasma halflife (1.1 h) and 100-fold selectivity of its triphosphate analog for HIV reverse transcriptase over human DNA polymerase [81, 82, 84, 87, 88]. The most commonly reported side-effects are anorexia, headaches, myalgia, nausea and vomiting [84]. Although long-term dosing side-effects include anemia, arthralgia, myopathy and neutropenia [84], antibacterial therapy are short-term (< 2 weeks) so these side-effects are unlikely to be of concern. In mice, the median oral lethal dose was found to be 3000 mg/kg [89]. In humans, two cases of oral overdosages of 20 g were reported without adverse effects or bonemarrow suppression [90, 91], suggesting that Zidovudine can be repurposed as an oral antibacterial agent for treating CRE bacteremia. In addition, Zidovudine also penetrates the bloodbrain barrier and can thus potentially be used for treating CRE meningitis [87, 88]. Mouse model E. coli systemic infection studies involving orally-administered Zidovudine (10 mg/kg body weight) showed that the drug was therapeutically efficacious [92]. Zidovudine levels in mouse plasma were found to be approximately 8 μg/mL (30 μM) an hour after oral administration, exceeding the 0.39–6.25 μM in vitro MICs against carbapenem-resistant E. coli and K. pneumoniae (Table 2). This suggests that therapeutically-active concentrations of Zidovudine can be achieved after oral administration [92]. An ideal antibacterial drug should kill bacteria quickly as this reduces the chances of resistance development and can potentially reduce treatment duration. Our time-kill assay (Fig. 2) revealed Zidovudine to be rapidly bactericial against E. coli, achieving a 6-log kill at 4× MIC within 3 h, in agreement with the literature [86]. Although Colistin exhibited faster killing kinetics (6-log kill at 4× MIC) in 1 h, its nephrotoxic and neurotoxic side-effects must be taken into consideration, especially for patients with impaired renal function [37–40]. Drugs that target a specific enzyme are known to be prone to resistance development [93]. Indeed, Zidovudine-resistant E. coli has been reported in HIV patients on Zidovudine

Eur J Clin Microbiol Infect Dis

therapy [94]. In vitro experiments revealed E. coli can become resistant within 24 h due to the loss of thymidine kinase activity which abrogates the bacteria’s ability to activate Zidovudine into the biologically-active triphosphate form [95, 96]. A strategy to circumvent resistance development is to administer Zidovudine in combination with another antibiotic. Earlier antibiotic combination studies have shown that Zidovudine acted synergistically with Colistin or Gentamicin against E. coli [83, 97]. Colistin and Tigecycline became potential combination candidates with Zidovudine as they were the only drugs active against all CRE isolates in Table 2. However, we decided to take Colistin out due to its nephroand neurotoxic side-effects. Thus, Zidovudine was paired with Tigecycline for the checkerboard assay. Results revealed that various Zidovudine-Tigecycline combinations were indifferent (FICI between 0.5 and 4.0) at various concentrations (Fig. 3), suggesting that Zidovudine and Tigecycline can be administered in combination to treat CRE infections. In conclusion, our drug library screen identified Zidovudine as a plausible antibacterial agent that can potentially be repurposed as an antibacterial agent. If so, to reduce the chances of resistance development, we would recommend Zidovudine be used in combination with a drug for which the bacteria has tested susceptible, for example, Tigecycline, when treating infections caused by CREs.

5.

6. 7.

8.

9.

10.

11.

12.

13.

14. Acknowledgements The authors thank the Agency for Science, Technology and Research (A*STAR) Biomedical Research Council for funding.

15.

Compliance with ethical standards 16. Ethical statement Not required as no humans or animals were involved in this study. 17. Conflicts of interest The authors of this paper declare no conflicts of interest.

References Gupta N, Limbago BM, Patel JB, Kallen AJ (2011) Carbapenemresistant Enterobacteriaceae: epidemiology and prevention. Clin Infect Dis 53:60–67 2. Satlin MJ, Chen L, Patel G, Gomez-Simmonds A, Weston G et al (2017) Multicenter clinical and molecular epidemiological analysis of bacteremia due to carbapenem-resistant Enterobacteriaceae (CRE) in the CRE epicenter of the United States. Antimicrob Agents Chemother 61:e02349–e02316 3. Nordmann P (2014) Carbapenemase-producing Enterobacteriaceae: overview of a major public health challenge. Med Mal Infect 44:51–56 4. Baldwin CM, Lyseng-Williamson KA, Keam SJ (2008) Meropenem: a review of its use in the treatment of serious bacterial infections. Drugs 68:803–838

18.

19.

1.

20.

21.

22.

23.

Morrill HJ, Pogue JM, Kaye KS, LaPlante KL (2015) Treatment options for carbapenem-resistant Enterobacteriaceae infections. Open Forum Infect Dis 2:ofv050 Vergidis PI, Falagas ME (2008) New antibiotic agents for bloodstream infections. Int J Antimicrob Agents 32:S60–S65 El-Gamal MI, Brahim I, Hisham N, Aladdin R, Mohammed H et al (2017) Recent updates of carbapenem antibiotics. Eur J Med Chem 131:185–195 Hsu LY, Apisarnthanarak A, Khan E, Suwantarat N, Ghafur A et al (2017) Carbapenem-resistant Acinetobacter baumannii and Enterobacteriaceae in South and Southeast Asia. Clin Microbiol Rev 30:1–22 Jain A, Hopkins KL, Turton J, Doumith M, Hill R et al (2014) NDM carbapenemases in the United Kingdom: an analysis of the first 250 cases. J Antimicrob Chemother 69:1777–1784 Kazmierczak KM, Rabine S, Hackel M, McLaughlin RE, Biedenbach DJ et al (2015) Multiyear, multinational survey of the incidence and global distribution of metallo-β-lactamaseproducing Enterobacteriaceae and Pseudomonas aeruginosa. Antimicrob Agents Chemother 60:1067–1078 Ling ML, Tee YM, Tan SG, Amin IM, How KB et al (2015) Risk factors for acquisition of carbapenem resistant Enterobacteriaceae in an acute tertiary care hospital in Singapore. Antimicrob Resist Infect Control 4:26 Mataseje LF, Abdesselam K, Vachon J, Mitchel R, Bryce E et al (2010) Results from the Canadian Nosocomial Infection Surveillance Program on carbapenemase-producing Enterobacteriaceae, 2010 to 2014. Antimicrob Agents Chemother 60:6787–6794 Pitout JD, Nordmann P, Poirel L (2015) Carbapenemase-producing Klebsiella pneumoniae, a key pathogen set for global nosocomial dominance. Antimicrob Agents Chemother 59:5873–5884 Xu Y, Gu B, Huang M, Liu H, Xu T et al (2015) Epidemiology of carbapenem resistant Enterobacteriaceae (CRE) during 2000-2012 in Asia. J Thorac Dis 7:376–385 Xu A, Zheng B, Xu YC, Huang ZG, Zhong NS et al (2016) National epidemiology of carbapenem-resistant and extensively drug-resistant Gram-negative bacteria isolated from blood samples in China in 2013. Clin Microbiol Infect 22:S1–S8 Nordmann P, Naas T, Poirel L (2011) Global Spread of carbapenemase-producing Enterobacteriaceae. Emerg Infect Dis 17:1791–1798 Logan LK, Weinstein RA (2017) The epidemiology of carbapenem-resistant Enterobacteriaceae: the impact and evolution of a global menace. J Infect Dis 15:S28–S36 Temkin E, Adler A, Lerner A, Carmeli Y (2014) Carbapenemresistant Enterobacteriaceae: biology, epidemiology, and management. Ann NY Acad Sci 1323:22–42 Frieden T (2013) Antibiotic Resistance Threats in the United States, 2013. Executive Summary. Centers for Disease Control and Prevention, Atlanta, GA, USA. Available online: http://www.cdc. gov/drugresistance/threat-report-2013/ . Accessed 1 July 2017 WHO (2017) Global priority list of antibiotic-resistant bacteria to guide research, discovery, and development of new antibiotics. http://www.who.int/medicines/publications/global-priority-listantibiotic-resistant-bacteria/en/ . Accessed 1 July 2017 Falagas ME, Kasiakou SK (2005) Colistin: the revival of polymyxins for the management of multidrug-resistant gram-negative bacterial infections. Clin Infect Dis 40:1333–1341 Falagas ME, Lourida P, Poulikakos P, Rafailidis PI, Tansarli GS (2014) Antibiotic treatment of infections due to carbapenemresistant Enterobacteriaceae: systematic evaluation of the available evidence. Antimicrob Agents Chemother 58:654–663 Karaiskos I, Giamarellou H (2014) Multidrug-resistant and extensively drug-resistant Gram-negative pathogens: current and

Eur J Clin Microbiol Infect Dis

24.

25.

26.

27. 28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

emerging therapeutic approaches. Expert Opin Pharmacother 15: 1351–1370 Zayyad H, Eliakim-Raz N, Leibovici L, Paul M (2017) Revival of old antibiotics: needs, the state of evidence and expectations. Int J Antimicrob Agents 49:536–541 Brust K, Evans A, Plemmons R (2014) Tigecycline in treatment of multidrug-resistant Gram-negative bacillus urinary tract infections: a systematic review. J Antimicrob Chemother 69:2606–1260 Morrill HJ, Pogue JM, Kaye KS, LaPlante KL (2015) Treatment options for carbapenem-resistant Enterobacteriaceae infections. Open Forum Infect Dis 2:1–15 Noskin GA (2005) Tigecycline: a new glycylcycline for treatment of serious infections. Clin Infect Dis 41:S303–S314 Sader HS, Castanheira M, Flamm RK, Mendes RE, Farrell DJ et al (2015) Tigecycline activity tested against carbapenem-resistant Enterobacteriaceae from 18 European nations: results from the SENTRY surveillance program (2010-2013). Diagn Microbiol Infect Dis 83:183–186 Chiu SK, Chan MC, Huang LY, Lin YT, Lin JC et al (2017) Tigecycline resistance among carbapenem-resistant Klebsiella pneumoniae: clinical characteristics and expression levels of efflux pump genes. PLoS One 12:e0175140 Rodríguez-Avial C, Rodríguez-Avial I, Merino P, Picazo JJ (2012) Klebsiella pneumoniae: development of a mixed population of carbapenem and tigecycline resistance during antimicrobial therapy in a kidney transplant patient. Clin Microbiol Infect 18:61–66 FDA (2013) FDA Drug Safety Communication: FDA warns of increased risk of death with IV antibacterial Tygacil (tigecycline) and approves new boxed warning. FDA, Rockville, MD. https:// www.fda.gov/drugs/drugsafety/ucm369580.htm. Accessed 1 July 2017 Dixit D, Madduri RP, Sharma R (2014) The role of tigecycline in the treatment of infections in light of the new black box warning. Expert Rev Anti-Infect Ther 12:397–400 Prasad P, Sun J, Danner RL, Natanson C (2012) Excess deaths associated with tigecycline after approval based on noninferiority trials. Clin Infect Dis 54:1699–1709 Falagas ME, Grammatikos AP, Michalopoulos A (2008) Potential of old-generation antibiotics to address current need for new antibiotics. Expert Rev Anti-Infect Ther 6:593–600 Li J, Nation RL, Milne RW, Turnidge JD, Coulthard K (2005) Evaluation of colistin as an agent against multi-resistant Gram-negative bacteria. Int J Antimicrob Agents 25:11–25 Li J, Nation RL, Turnidge JD, Milne RW, Coulthard K et al (2006) Colistin: the re-emerging antibiotic for multidrug-resistant Gramnegative bacterial infections. Lancet Infect Dis 6:589–601 Kaye KS, Gales AC, Dubourg G (2017) Old antibiotics for multidrug-resistant pathogens: from in vitro activity to clinical outcomes. Int J Antimicrob Agents 49:542–548 Pogue JM, Ortwine JK, Kaye KS (2016) Are there any ways around the exposure-limiting nephrotoxicity of the polymyxins? Int J Antimicrob Agents 48:622–626 Tran TB, Velkov T, Nation RL, Forrest A, Tsuji BT et al (2016) Pharmacokinetics/ pharmacodynamics of colistin and polymyxin B: are we there yet? Int J Antimicrob Agents 48:592–597 Yun B, Azad MA, Wang J, Nation RL, Thompson PE et al (2015) Imaging the distribution of polymyxins in the kidney. J Antimicrob Chemother 70:827–829 Aran JM, Erre JP, Lima da Costa D, Debbarh I et al (1999) Acute and chronic effects of aminoglycosides on cochlear hair cells. Ann NY Acad Sci 884:60–68 Huth ME, Ricci AJ, Cheg AG (2011) Mechanisms of aminoglycoside ototoxicity and targets of hair cell protection. Int J Otolarynol 2011:937861 Cosgrove SE, Vigliani GA, Fowler VG Jr, Abrutyn E, Corey GR et al (2009) Initial low-dose gentamicin for Staphylococcus aureus

44. 45. 46. 47.

48. 49.

50.

51. 52.

53. 54.

55.

56.

57.

58.

59. 60.

61.

62. 63. 64.

65.

bacteremia and endocarditis is nephrotoxic. Clin Infect Dis 48:713– 721 Selimoglu E (2007) Aminoglycoside-induced ototoxicity. Curr Pharm Des 13:119–126 Kaye KS, Pogue JM, Tran TB, Nation RL, Li J (2016) Agents of last resort: polymyxin resistance. Infect Dis Clin N Am 30:391–414 Hu Y, Liu F, Lin IY, Gao GF, Zhu B (2016) Dissemination of the mcr-1 colistin resistance gene. Lancet Infect Dis 16:146–147 Olaitan AO, Morand S, Rolain JM (2016) Emergence of colistinresistant bacteria in humans without colistin usage: a new worry and cause for vigilance. Int J Antimicrob Agents 47:1–3 Olaitan AO (2016) Emergence of polymyxin resistance in Gramnegative bacteria. Int J Antimicrob Agents 48:581–582 Garneau-Tsodikova S, Labby KJ (2016) Mechanisms of resistance to aminoglycoside antibiotics: overview and perspectives. Med Chem Commun 7:11–27 Poirel L, Jayol A, Nordmann P (2017) Polymyxins: antibacterial activity, susceptibility testing, and resistance mechanisms encoded by plasmids or chromosomes. Clin Microbiol Rev 30:557–596 Rolain JM, Olaitan AO (2016) Plasmid-mediated colistin resistance: the final blow to colistin? Int J Antimicrob Agents 47:4–5 Jayol A, Poirel L, Dortet L, Nordmann P (2016) National survey of c o l i s t i n r e s i s t a n c e a m o n g c a r ba pe ne m a s e - p r o d u c i n g Enterobacteriaceae and outbreak caused by colistin-resistant OXA-48-producing Klebsiella pneumoniae, France, 2014. Euro Surveill 21 Jeannot K, Bolard A, Plésiat P (2017) Resistance to polymyxins in Gram-negative organisms. Int J Antimicrob Agents 49:526–535 Quan J, Li X, Chen Y, Jiang Y, Zhou Z et al (2017) Prevalence of mcr-1 in Escherichia coli and Klebsiella pneumoniae recovered from bloodstream infections in China: a multicentre longitudinal study. Lancet Infect Dis 17:400–410 Wang Y, Tian GB, Zhang R, Shen Y, Tyrrell JM et al (2017) Prevalence, risk factors, outcomes, and molecular epidemiology of mcr-1-positive Enterobacteriaceae in patients and healthy adults from China: an epidemiological and clinical study. Lancet Infect Dis 17:390–399 Zowawi HM, Harris PN, Roberts MJ, Tambyah PA, Schembri MA et al (2015) The emerging threat of multidrug-resistant Gram-negative bacteria in urology. Nat Rev Urol 12:570–584 Kinch MS, Patridge E, Plummer M, Hoyer D (2014) An analysis of FDA-approved drugs for infectious disease: Antibacterial agents. Drug Discov Today 19:1283–1287 Brenner R, Ellis-Grosse EJ, Echols R (2006) Moving goalposts regulatory oversight of antibacterial drugs. Nat Biotechnol 24: 1515–1520 Fernandes P (2006) Antibacterial discovery and development - The failure of success? Nat Biotechnol 24:1497–1503 Nambiar S, Laessig K, Toerner J, Farley J, Cox E (2014) Antibacterial drug development: Challenges, recent developments, and future considerations. Clin Pharmacol Ther 96:147–149 Ashburn TT, Thor KB (2004) Drug repositioning: Identifying and developing new uses for existing drugs. Nat Rev Drug Discov 3: 673–683 Nosengo N (2016) New tricks for old drugs. Nature 534:314–316 Oprea TI, Mestres J (2012) Drug repurposing: Far beyond new targets for old drugs. AAPS J 14:759–763 Clinical and Laboratory Standards Institute (2012) Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically, approved standard, 9th ed, vol 32, no. 2. Document M07A9. CLSI, Wayne, PA, USA French GL (2006) Bactericidal agents in the treatment of MRSA infections - the potential role of daptomycin. J Antimicrob Chemother 58:1107–1117

Eur J Clin Microbiol Infect Dis 66.

Hsieh MH, Yu CM, Yu VL, Chow JW (1993) Synergy assessed by checkerboard. A critical analysis. Diagn Microbiol Infect Dis 16: 343–349 67. Sader HS, Farrell DJ, Flamm RK, Jones RN (2014) Variation in potency and spectrum of tigecycline activity against bacterial strains from U.S. medical centers since its approval for clinical use (2006 to 2012). Antimicrob Agents Chemother 58:2274–2280 68. Small PM, Chambers HF (1990) Vancomycin for Staphylococcus aureus endocarditis in intravenous drug users. Antimicrob Agents Chemother 34:1227–1231 69. Kaminskas E, Farrell AT, Wang YC, Sridhara R, Pazdur R (2005) FDA drug approval summary: azacitidine (5-azacytidine, Vidaza) for injectable suspension. Oncologist 10:176–182 70. Jones PA, Taylor SM, Wilson VL (1983) Inhibition of DNA methylation by 5-azacytidine. Recent Results Cancer Res 84:202–211 71. Uchida T, Kinoshita T, Nagai H, Nakahara Y, Saito H et al (1997) Hypermethylation of the p15INK4B gene in myelodysplastic syndromes. Blood 90:1403–1409 72. Freireich EJ, Gehan EA, Rall DP, Schmidt LH, Skipper HE (1966) Quantitative comparison of toxicity of anticancer agents in mouse, rat, hamster, dog, monkey, and man. Cancer Chemother Rep 50: 219–244 73. Chabner BA, Drake JC, Johns DG (1973) Deamination of 5azacytidine by a human leukemia cell cytidine deaminase. Biochem Pharmacol 22:2763–2765 74. Michalopoulos AS, Falagas ME (2011) Colistin: recent data on pharmacodynamics properties and clinical efficacy in critically ill patients. Ann Intensive Care 1:30–35 75. Muralidharan G, Micalizzi M, Speth J, Raible D, Troy S (2005) Pharmacokinetics of tigecycline after single and multiple doses in healthy subjects. Antimicrob Agents Chemother 49:220–229 76. EMA (2017) EMA public assessment report on Vidaza. Document number: Vidaza -EMEA/H/C/000978 -IB/0040. Date: 20/03/ 2017.www.ema.europa.eu/docs/en_GB/document_library/EPAR__Product_Information/human/000978/WC500050239.pdf. Accessed 4 October 2017 77. Power DG, Kemeny NE (2009) The role of floxuridine in metastatic liver disease. Mol Cancer Ther 8:1015–1025 78. Wambaugh MA, Shakya VPS, Lewis AJ, Mulvey MA, Brown JCS (2017) High-throughput identification and rational design of synergistic small-molecule pairs for combating and bypassing antibiotic resistance. PLoS Biol 15:e2001644 79. Chang AE, Schneider PD, Sugarbaker PH, Simpson C, Culnane M et al (1987) A prospective randomized trial of regional versus systemic continuous 5-fluorodeoxyuridine chemotherapy in the treatment of colorectal liver metastases. Ann Surg 206:685–693 80. Leichman CG (1994) Prolonged infusion of fluorinated pyrimidines in gastrointestinal malignancies: a review of recent clinical trials. Cancer Investig 12:166–175 81. Mitsuya H, Weinhold K, Furman P, St Clair MH, Lehrman SN et al (1985) 3′-Azido-3′-deoxythymidine (BW A509U): an antiviral

agent that inhibits the infectivity and cytopathic effect of human T-lymphotropic virus type III/lymphadenopathy-associated virus in vitro. Proc Natl Acad Sci USA 82:7096–7100 82. Furman P, Fyfe J, St Clair MH, Weinhold K, Rideout J et al (1986) Phosphorylation of 3′-azido-3′-deoxythymidine and selective interaction of the 5′-triphosphate with human immunodeficiency virus reverse transcriptase. Proc Natl Acad Sci USA 83:8333–8337 83. Doléans-Jordheim A, Bergeron E, Bereyziat F, Ben-Larbi S, Dumitrescu O et al (2011) Zidovudine (AZT) has a bactericidal effect on enterobacteria and induces genetic modifications in resistant strains. Eur J Clin Microbiol Infect Dis 30:1249–1256 84. Beach JW (1998) Chemotherapeutic agents for human immunodeficiency virus infection: mechanism of action, pharmacokinetics, metabolism, and adverse reactions. Clin Ther 20:2–25 85. Elwell LP, Ferone R, Freeman GA, Fyfe JA, Hill JA et al (1987) Antibacterial activity and mechanism of action of 3′-azido-3′deoxythymidine (BW A509U). Antimicrob Agents Chemother 31:274–280 86. Lewin CS, Amyes SG (1989) Conditions required for the antibacterial activity of zidovudine. Eur J Clin Microbiol Infect Dis 8:737– 741 87. Blum MR, Liao SH, Good SS, de Miranda P (1988) Pharmacokinetics and bioavailability of zidovudine in humans. Am J Med 85:189–194 88. Klecker RW Jr, Collins JM, Yarchoan R, Thomas R, Jenkins JF et al (1987) Plasma and cerebrospinal fluid pharmacokinetics of 3′-azido-3′-deoxythymidine: a novel pyrimidine analog with potential application for the treatment of patients with AIDS and related diseases. Clin Pharmacol Ther 41:407–412 89. Ayers KM (1988) Preclinical toxicology of zidovudine. An overview. Am J Med 85(2A):186–188 90. Hargreaves M, Fuller G, Costello C, Gazzard B (1988) Zidovudine overdose. Lancet 332:509 91. Pickus OB (1988) Overdose of zidovudine. N Engl J Med 318:1206 92. Keith BR, White G, Wilson HR (1989) In vivo efficacy of zidovudine (3′-azido-3′-deoxythymidine) in experimental gram-negativebacterial infections. Antimicrob Agents Chemother 33:479–483 93. Spratt BG (1994) Resistance to antibiotics mediated by target alterations. Science 264:388–393 94. Lewin CS, Watt B, Paton R, Amyes SGB (1990) Isolation of zidovudine resistant Escherichia coli from AIDS patients. FEMS Microbiol Lett 58:141–143 95. Lewin CS, Allen R, Amyes SGB (1990) Zidovudine-resistance in Salmonella typhimurium and Escherichia coli. J Antimicrob Chemother 25:706–708 96. Lewin CS, Allen RA, Amyes SGB (1990) Mechanisms of zidovudine resistance in bacteria. J Med Microbiol 33:235–238 97. Hu Y, Coates A (2014) Combination comprising zidovudine and polymyxin. WO 2014147405 A1

Repurposing Zidovudine in combination with Tigecycline for treating carbapenem-resistant Enterobacteriaceae infections.

The global emergence of carbapenem-resistant Enterobacteriaceae (CRE) presents a significant clinical concern, prompting the WHO to prioritize CRE as ...
735KB Sizes 0 Downloads 9 Views