1
|
Sedaghati M, Akbari R, Lotfollahi Hagghi L, Yousefi S, Mesbahi T, Delfi M. Survey of probable synergism between melittin and ciprofloxacin, rifampicin, and chloramphenicol against multidrug-resistant Pseudomonas aeruginosa. Front Microbiol 2024; 15:1480299. [PMID: 39640853 PMCID: PMC11617520 DOI: 10.3389/fmicb.2024.1480299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Background The emergence of multidrug-resistant bacteria and also biofilm-associated infections is a great health concern due to the failure of available antibiotics. This has alerted scientists to developing alternative antibiotics. Melittin as an antimicrobial peptide has antibacterial synergistic activity in combining with conventional antibiotics against pathogenic bacteria. Accordingly, this study aimed to assess the synergistic effect of melittin in combination with Ciprofloxacin, Rifampicin, and Chloramphenicol against MDR strains of P. aeruginosa. Materials and methods Fifty strains of P. aeruginosa were isolated from clinical specimens. The antibiotic susceptibility of isolates was evaluated by the disk diffusion method. The MIC and MBC of melittin and melittin-antibiotics combination against isolated strains were examined by microdilution method. The probable synergism between melittin and antibiotics was assayed using the FIC protocol. Time-killing kinetics and anti-biofilm effects of melittin and melittin-antibiotics combination were evaluated using time-kill kinetics and crystal violet staining method, respectively. The toxicity of the melittin-antibiotics combination on the HEK293 cell line was also assessed by the MTT assay method. Results Out of 50 isolates of P. aeruginosa, 15 strains are considered to be multidrug strains. Among MDR strains of P. aeruginosa, 42.85% were resistant to cefepime and ceftazidime and all urine-originate isolates were resistant to cotrimoxazole. A combination of MIC dose of ciprofloxacin and melittin decreased resistance against ciprofloxacin up to 33%. The ciprofloxacin-melittin combination showed a favorable synergism and anti-biofilm effect and was also 30.3% less toxic than melittin alone at 4 μg/ml against the HEK293 cell line. In contrast to ciprofloxacin, with the melittin-rifampicin and melittin-chloramphenicol combinations, an addition effect occurred, respectively, in 86.66 and 53.33% of MDR strains of P. aeruginosa. Conclusion Combining melittin's antibacterial and anti-biofilm properties with traditional antibiotics may offer a novel strategy to address antibiotic resistance in P. aeruginosa. The simultaneous administration of melittin and ciprofloxacin in a single dose has shown a marked increase in antibacterial effectiveness while minimizing toxicity to the HEK293 cell line. It is advisable to conduct additional research to explore the combined antibacterial effects of melittin and ciprofloxacin in a wider range of clinical samples, animal models, and clinical trial settings.
Collapse
Affiliation(s)
| | - Reza Akbari
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Lida Lotfollahi Hagghi
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | | | | |
Collapse
|
2
|
Aktas Z, Sonmez N, Oksuz L, Boral O, Issever H, Oncul O. Efficacy of antibiotic combinations in an experimental sepsis model with Pseudomonas aeruginosa. Braz J Microbiol 2023; 54:2817-2826. [PMID: 37828396 PMCID: PMC10689617 DOI: 10.1007/s42770-023-01141-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023] Open
Abstract
This study aimed to compare the efficacy of fosfomycin, colistin, tobramycin and their dual combinations in an experimental sepsis model. After sepsis was established with a Pseudomonas aeruginosa isolate (P1), antibiotic-administered rats were divided into six groups: Fosfomycin, tobramycin, colistin and their dual combinations were administered by the intravenous or intraperitoneal route to the groups. The brain, heart, lung, liver, spleen and kidney tissues of rats were cultured to investigate bacterial translocation caused by P1. Given the antibiotics and their combinations, bacterial colony counts in liver tissues were decreased in colistin alone and colistin plus tobramycin groups compared with control group, but there were no significant differences. In addition, a non-statistical decrease was found in the spleen tissues of rats in the colistin plus tobramycin group. There was a > 2 log10 CFU/ml decrease in the number of bacterial colonies in the kidney tissues of the rats in the fosfomycin group alone, but the decrease was not statistically significant. However, there was an increase in the number of bacterial colonies in the spleen and kidney samples in the group treated with colistin as monotherapy compared to the control group. The number of bacterial colonies in the spleen samples in fosfomycin plus tobramycin groups increased compared to the control group. Bacterial colony numbers in all tissue samples in the fosfomycin plus colistin group were found to be close to those in the control group. Colistin plus tobramycin combinations are effective against P. aeruginosa in experimental sepsis, and clinical success may be achieved. New in vivo studies demonstrating the ability of P. aeruginosa to biofilm formation in tissues other than the lung are warranted in future.
Collapse
Affiliation(s)
- Zerrin Aktas
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Capa, Istanbul, Türkiye
| | - Nese Sonmez
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Capa, Istanbul, Türkiye
| | - Lutfiye Oksuz
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Capa, Istanbul, Türkiye.
| | - Ozden Boral
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Capa, Istanbul, Türkiye
| | - Halim Issever
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Capa, Istanbul, Türkiye
| | - Oral Oncul
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Capa, Istanbul, Türkiye
| |
Collapse
|
3
|
Badawy MSEM, Elkhatib WF, Shebl RI. Mathematical pharmacodynamic modeling for antimicrobial assessment of ceftazidime/colistin versus gentamicin/meropenem combinations against carbapenem-resistant Pseudomonas aeruginosa biofilm. Ann Clin Microbiol Antimicrob 2023; 22:53. [PMID: 37394468 DOI: 10.1186/s12941-023-00597-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/29/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Carbapenem-resistant Pseudomonas aeruginosa (CRPA) represents an escalating healthcare hazard with high mortality worldwide, especially in presence of biofilm. The current study aimed to evaluate the anti-biofilm potentials of ceftazidime, colistin, gentamicin, and meropenem alone and in combinations against biofilm-forming CRPA. METHODS Biofilm killing and checkerboard assay were performed to detect the effectiveness of combined antibiotics against biofilms and planktonic cells, respectively. The bacterial bioburden retrieved from the established biofilms following treatment with combined antibiotics was utilized to construct a three-dimensional response surface plot. A sigmoidal maximum effect model was applied to determine the pharmacodynamic parameters (maximal effect, median effective concentration, and Hill factor) of each antibiotic to create a mathematical three-dimensional response surface plot. RESULTS Data revealed statistically significant (p < 0.05) superior anti-biofilm potential in the case of colistin followed by a lower effect in the case of gentamicin and meropenem, while ceftazidime exhibited the least anti-biofilm activity. The fractional inhibitory concentration index (FICI ≤ 0.5) indicated synergism following treatment with the combined antibiotics. An elevated anti-biofilm activity was recorded in the case of gentamicin/meropenem compared to ceftazidime/colistin. Synergistic anti-biofilm potentials were also detected via the simulated pharmacodynamic modeling, with higher anti-biofilm activity in the case of the in vitro observation compared to the simulated anti-biofilm profile. CONCLUSIONS The present study highlighted the synergistic potentials of the tested antibiotic combinations against P. aeruginosa biofilms and the importance of the mathematical pharmacodynamic modeling in investigating the efficacy of antibiotics in combination as an effective strategy for successful antibiotic therapy to tackle the extensively growing resistance to the currently available antibiotics.
Collapse
Affiliation(s)
- Mona Shaban E M Badawy
- Department of Microbiology and Immunology, Faculty of Pharmacy (Girls), El-Azhar University, Cairo, Egypt
| | - Walid F Elkhatib
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo, 11566, Egypt.
- Department of Microbiology & Immunology, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt.
| | - Rania I Shebl
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, 6th October city, 4th industrial zone, Giza, 12451, Egypt.
| |
Collapse
|
4
|
Truşcă BS, Gheorghe-Barbu I, Manea M, Ianculescu E, Barbu IC, Măruțescu LG, Dițu LM, Chifiriuc MC, Lazăr V. Snapshot of Phenotypic and Molecular Virulence and Resistance Profiles in Multidrug-Resistant Strains Isolated in a Tertiary Hospital in Romania. Pathogens 2023; 12:pathogens12040609. [PMID: 37111495 PMCID: PMC10145626 DOI: 10.3390/pathogens12040609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
A current major healthcare problem is represented by antibiotic resistance, mainly due to multidrug resistant (MDR) Gram negative bacilli (GNB), because of their extended spread both in hospital facilities and in the community's environment. The aim of this study was to investigate the virulence traits of Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa MDR, XDR, and PDR strains isolated from various hospitalized patients. These GNB strains were investigated for the presence of soluble virulence factors (VF), such as hemolysins, lecithinase, amylase, lipase, caseinase, gelatinase, and esculin hydrolysis, as well as for the presence of virulence genes encoding for VF involved in adherence (TC, fimH, and fimA), biofilm formation (algD, ecpRAB, mrkA, mrkD, ompA, and epsA), tissue destruction (plcH and plcN), and in toxin production (cnfI, hlyA, hlyD, and exo complex). All P. aeruginosa strains produced hemolysins; 90% produced lecithinase; and 80% harbored algD, plcH, and plcN genes. The esculin hydrolysis was detected in 96.1% of the K. pneumoniae strains, whereas 86% of them were positive for the mrkA gene. All of the A. baumannii strains produced lecithinase and 80% presented the ompA gene. A significant association was found between the number of VF and the XDR strains, regardless of the isolation sources. This study opens new research perspectives related to bacterial fitness and pathogenicity, and it provides new insights regarding the connection between biofilm formation, other virulence factors, and antibiotic resistance.
Collapse
Affiliation(s)
- Bianca Simona Truşcă
- Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
| | - Irina Gheorghe-Barbu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 050567 Bucharest, Romania
| | - Marina Manea
- Fundeni Clinical Institute, 022328 Bucharest, Romania
- Fundeni Clinical Institute, University of Medicine and Pharmacy "Carol Davila" Bucharest, 020021 Bucharest, Romania
| | | | - Ilda Czobor Barbu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 050567 Bucharest, Romania
| | - Luminița Gabriela Măruțescu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 050567 Bucharest, Romania
| | - Lia-Mara Dițu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 050567 Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 050567 Bucharest, Romania
- Romanian Academy, 050045 Bucharest, Romania
| | - Veronica Lazăr
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
| |
Collapse
|
5
|
Liu Y, Ma W, Li M, Wu J, Sun L, Zhao W, Sun S. Antibacterial and anti-biofilm activities of fosfomycin combined with rifampin against Carbapenem-resistant Pseudomonas aeruginosa. Lett Appl Microbiol 2022; 75:1559-1568. [PMID: 36036376 DOI: 10.1111/lam.13822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Abstract
Increasing prevalence of carbapenem-resistant Pseudomonas aeruginosa(CRPA)strains in the hospital setting represents an emerging challenge to clinical treatment for Pseudomonas aeruginosa (PA) infections, as the range of therapeutic agents active against these pathogens becomes increasingly constrained. This study demonstrated for the first time that fosfomycin (FOS) combined with rifampin (RIF) showed strong synergistic effects against CRPA and carbapenem-susceptible PA, with 100% synergistic rates. Additionally, time-killing curve further proves dynamic antibacterial activity of FOS+RIF against CRPA. Further experiments determined that antibacterial mechanisms of FOS+RIF might be inhibition of biofilm formation and eradication of pre-formed biofilm. The results of inhibition biofilm formation assay demonstrated that RIF and FOS at 1/8MIC, 1/16MIC and 1/32MIC have better inhibitory effects on CRPA biofilm formation VS FOS alone (96%, 90% and 78% VS 29%, 24% and 22%) (p<0.0001) or RIF alone (96%, 90% and 78% VS 86%, 67% and 29%) (p<0.01). The rates of eradicating pre-formed biofilm with combination therapy at 1/2MIC, 1/4MIC and 1/8MIC of both antibiotics, increased 46%, 61% and 55% compared with FOS alone (p<0.001) and 37%, 33% and 46% compared with RIF alone (p<0.01). This finding will provide new insights for the treatment of bacterial infection caused by CRPA, which can be further explored in the clinical practice.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Clinical Pharmacy, the First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China
| | - Wenli Ma
- Department of Clinical Pharmacy, the First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China.,Department of Clinical Pharmacy, ShanDong Healthcare Group FeiCheng Hospital, Taian, People's Republic of China
| | - Min Li
- Department of Clinical Pharmacy, the First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China
| | - Jiyong Wu
- Department of Pharmacy, Shandong Second Provincial General Hospital, 250022, Jinan, Shandong, P.R. China
| | - Licui Sun
- Department of Clinical Pharmacy, ShanDong Healthcare Group FeiCheng Hospital, Taian, People's Republic of China
| | - Wei Zhao
- Department of Clinical Pharmacy, ShanDong Healthcare Group FeiCheng Hospital, Taian, People's Republic of China
| | - Shujuan Sun
- Department of Clinical Pharmacy, the First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China.,Department of Pharmacy, Shandong Second Provincial General Hospital, 250022, Jinan, Shandong, P.R. China
| |
Collapse
|
6
|
Tait JR, Bilal H, Rogers KE, Lang Y, Kim TH, Zhou J, Wallis SC, Bulitta JB, Kirkpatrick CMJ, Paterson DL, Lipman J, Bergen PJ, Roberts JA, Nation RL, Landersdorfer CB. Effect of Different Piperacillin-Tazobactam Dosage Regimens on Synergy of the Combination with Tobramycin against Pseudomonas aeruginosa for the Pharmacokinetics of Critically Ill Patients in a Dynamic Infection Model. Antibiotics (Basel) 2022; 11:101. [PMID: 35052977 PMCID: PMC8772788 DOI: 10.3390/antibiotics11010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 12/10/2022] Open
Abstract
We evaluated piperacillin-tazobactam and tobramycin regimens against Pseudomonas aeruginosa isolates from critically ill patients. Static-concentration time-kill studies (SCTK) assessed piperacillin-tazobactam and tobramycin monotherapies and combinations against four isolates over 72 h. A 120 h-dynamic in vitro infection model (IVM) investigated isolates Pa1281 (MICpiperacillin 4 mg/L, MICtobramycin 0.5 mg/L) and CR380 (MICpiperacillin 32 mg/L, MICtobramycin 1 mg/L), simulating the pharmacokinetics of: (A) tobramycin 7 mg/kg q24 h (0.5 h-infusions, t1/2 = 3.1 h); (B) piperacillin 4 g q4 h (0.5 h-infusions, t1/2 = 1.5 h); (C) piperacillin 24 g/day, continuous infusion; A + B; A + C. Total and less-susceptible bacteria were determined. SCTK demonstrated synergy of the combination for all isolates. In the IVM, regimens A and B provided initial killing, followed by extensive regrowth by 72 h for both isolates. C provided >4 log10 CFU/mL killing, followed by regrowth close to initial inoculum by 96 h for Pa1281, and suppressed growth to <4 log10 CFU/mL for CR380. A and A + B initially suppressed counts of both isolates to <1 log10 CFU/mL, before regrowth to control or starting inoculum and resistance emergence by 72 h. Overall, the combination including intermittent piperacillin-tazobactam did not provide a benefit over tobramycin monotherapy. A + C, the combination regimen with continuous infusion of piperacillin-tazobactam, provided synergistic killing (counts <1 log10 CFU/mL) of Pa1281 and CR380, and suppressed regrowth to <2 and <4 log10 CFU/mL, respectively, and resistance emergence over 120 h. The shape of the concentration-time curve was important for synergy of the combination.
Collapse
Affiliation(s)
- Jessica R. Tait
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.R.T.); (K.E.R.); (R.L.N.)
| | - Hajira Bilal
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (H.B.); (C.M.J.K.); (P.J.B.)
| | - Kate E. Rogers
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.R.T.); (K.E.R.); (R.L.N.)
| | - Yinzhi Lang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (Y.L.); (J.Z.); (J.B.B.)
| | - Tae-Hwan Kim
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Korea;
| | - Jieqiang Zhou
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (Y.L.); (J.Z.); (J.B.B.)
| | - Steven C. Wallis
- The University of Queensland Center for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (S.C.W.); (D.L.P.); (J.L.); (J.A.R.)
| | - Jürgen B. Bulitta
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (Y.L.); (J.Z.); (J.B.B.)
| | - Carl M. J. Kirkpatrick
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (H.B.); (C.M.J.K.); (P.J.B.)
| | - David L. Paterson
- The University of Queensland Center for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (S.C.W.); (D.L.P.); (J.L.); (J.A.R.)
| | - Jeffrey Lipman
- The University of Queensland Center for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (S.C.W.); (D.L.P.); (J.L.); (J.A.R.)
- Intensive Care Unit, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
- Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, 30900 Nîmes, France
- Jamieson Trauma Institute, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
| | - Phillip J. Bergen
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (H.B.); (C.M.J.K.); (P.J.B.)
| | - Jason A. Roberts
- The University of Queensland Center for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (S.C.W.); (D.L.P.); (J.L.); (J.A.R.)
- Intensive Care Unit, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
- Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, 30900 Nîmes, France
| | - Roger L. Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.R.T.); (K.E.R.); (R.L.N.)
| | - Cornelia B. Landersdorfer
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.R.T.); (K.E.R.); (R.L.N.)
| |
Collapse
|
7
|
Antimicrobial susceptibility of multidrug-resistant Pseudomonas aeruginosa isolated from drinking water and hospitalized patients in Jordan. Acta Trop 2021; 217:105859. [PMID: 33582141 DOI: 10.1016/j.actatropica.2021.105859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/28/2022]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is an important environmental, opportunistic and nosocomial pathogen with a significant threat to public health. The objectives of this study were to determine the in vitro antimicrobial susceptibility patterns of, and antibiotic drug combinations with synergistic effects against P. aeruginosa isolated from drinking water and hospitalized patients in Jordan. A total of 16 P. aeruginosa isolates were obtained from hospitalized patients and 15 were isolated from bottled drinking water were used in the study. Bacterial isolation and identification was performed using routine microbiological methods and confirmed using PCR technique targeting the 16S rDNA gene. The antimicrobial susceptibility patterns were determined by measuring the minimum inhibitory concentration (MIC) using the 2-fold microdilution method. Synergy interaction between various antimicrobials was determined using the checkerboard method and fractional inhibitory concentration index (FICI). The majority of water isolates were sensitive to gentamicin (93.3%), ticarcillin (86.7%) and ciprofloxacin, levofloxacin, amikacin, colistin, piperacillin, azlocillin, aztreonam, ceftazidime and imipenem (100% each). All water isolates (100%) were resistant to amoxicillin, oxytetracycline and doxycycline (93.3% and 86.7, respectively). For the clinical isolates, all (100%) were sensitive to ceftazidime, 81.3% were sensitive to aztreonam, while 62.5% were sensitive to ciprofloxacin, levofloxacin, gentamicin, amikacin, colistin, piperacillin, ticracillin, azlocillin, and imipenem. All clinical isolates (100%) were resistant to oxytetracycline, doxycycline and amoxicillin. Analysis of the checkerboard synergy assay of multi-drug resistant isolates (n=26) showed significant synergism (P ≤ 0.05) when ciprofloxacin or gentamicin were included in the combination. There were no significant differences in synergistic activity between ciprofloxacin and levofloxacin when combined with other antimicrobial agents of the beta-lactams or aminoglycosides classes. There were no significant differences in the synergistic activities between beta lactams - aminoglycoside and beta lactams - fluoroquinolone combinations. Results of this study indicate an alarming widespread presence of multidrug-resistant P. aeruginosa associated with chronic suppurative infections in hospitalized patients and apparently clean drinking water in Jordan. Treatment of clinical suppurative lesions must be based on culture and in vitro susceptibility testing using potent antimicrobial combinations to avoid emergence of resistant strains and to improve the clinical outcome of treated patients.
Collapse
|
8
|
Liu J, Chen F, Wang X, Peng H, Zhang H, Wang KJ. The Synergistic Effect of Mud Crab Antimicrobial Peptides Sphistin and Sph 12-38 With Antibiotics Azithromycin and Rifampicin Enhances Bactericidal Activity Against Pseudomonas Aeruginosa. Front Cell Infect Microbiol 2020; 10:572849. [PMID: 33194811 PMCID: PMC7645104 DOI: 10.3389/fcimb.2020.572849] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022] Open
Abstract
Overuse or abuse of antibiotics has undoubtedly accelerated the increasing prevalence of global antibiotic resistance crisis, and thus, people have been trying to explore approaches to decrease dosage of antibiotics or find new antibacterial agents for many years. Antimicrobial peptides (AMPs) are the ideal candidates that could kill pathogens and multidrug-resistant bacteria either alone or in combination with conventional antibiotics. In the study, the antimicrobial efficacy of mud crab Scylla paramamosain AMPs Sphistin and Sph12−38 in combination with eight selected antibiotics was evaluated using a clinical pathogen, Pseudomonas aeruginosa. It was interesting to note that the in vitro combination of rifampicin and azithromycin with Sphistin and Sph12−38 showed significant synergistic activity against P. aeruginosa. Moreover, an in vivo study was carried out using a mouse model challenged with P. aeruginosa, and the result showed that the combination of Sph12−38 with either rifampicin or azithromycin could significantly promote the healing of wounds and had the healing time shortened to 4–5 days compared with 7–8 days in control. The underlying mechanism might be due to the binding of Sphistin and Sph12−38 with P. aeruginosa lipopolysaccharides (LPS) and subsequent promotion of the intracellular uptake of rifampicin and azithromycin. Taken together, the significant synergistic antibacterial effect on P. aeruginosa in vitro and in vivo conferred by the combination of low dose of Sphistin and Sph12−38 with low dose of rifampicin and azithromycin would be beneficial for the control of antibiotic resistance and effective treatment of P. aeruginosa-infected diseases in the future.
Collapse
Affiliation(s)
- Jie Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Xiaofei Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Hui Peng
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Hua Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
9
|
Antonello RM, Principe L, Maraolo AE, Viaggi V, Pol R, Fabbiani M, Montagnani F, Lovecchio A, Luzzati R, Di Bella S. Fosfomycin as Partner Drug for Systemic Infection Management. A Systematic Review of Its Synergistic Properties from In Vitro and In Vivo Studies. Antibiotics (Basel) 2020; 9:antibiotics9080500. [PMID: 32785114 PMCID: PMC7460049 DOI: 10.3390/antibiotics9080500] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/31/2022] Open
Abstract
Fosfomycin is being increasingly prescribed for multidrug-resistant bacterial infections. In patients with systemic involvement, intravenous fosfomycin is usually administered as a partner drug, as part of an antibiotic regimen. Hence, the knowledge of fosfomycin pharmacodynamic interactions (synergistic, additive, indifferent and antagonistic effect) is fundamental for a proper clinical management of severe bacterial infections. We performed a systematic review to point out fosfomycin’s synergistic properties, when administered with other antibiotics, in order to help clinicians to maximize drug efficacy optimizing its use in clinical practice. Interactions were more frequently additive or indifferent (65.4%). Synergism accounted for 33.7% of total interactions, while antagonism occurred sporadically (0.9%). Clinically significant synergistic interactions were mostly distributed in combination with penicillins (51%), carbapenems (43%), chloramphenicol (39%) and cephalosporins (33%) in Enterobactaerales; with linezolid (74%), tetracyclines (72%) and daptomycin (56%) in Staphylococcus aureus; with chloramphenicol (53%), aminoglycosides (43%) and cephalosporins (36%) against Pseudomonas aeruginosa; with daptomycin (97%) in Enterococcus spp. and with sulbactam (75%) and penicillins (60%) and in Acinetobacter spp. fosfomycin-based antibiotic associations benefit from increase in the bactericidal effect and prevention of antimicrobial resistances. Taken together, the presence of synergistic interactions and the nearly total absence of antagonisms, make fosfomycin a good partner drug in clinical practice.
Collapse
Affiliation(s)
- Roberta Maria Antonello
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
| | | | - Alberto Enrico Maraolo
- First Division of Infectious Diseases, Cotugno Hospital, AORN dei Colli, 80131 Naples, Italy;
| | | | - Riccardo Pol
- Department of Infectious Diseases, Udine University, 33100 Udine, Italy;
| | - Massimiliano Fabbiani
- Department of Medical Sciences, Tropical and Infectious Diseases Unit, University Hospital of Siena, 53100 Siena, Italy; (M.F.); (F.M.)
| | - Francesca Montagnani
- Department of Medical Sciences, Tropical and Infectious Diseases Unit, University Hospital of Siena, 53100 Siena, Italy; (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Antonio Lovecchio
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
| | - Roberto Luzzati
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
- Correspondence:
| |
Collapse
|
10
|
Garza-Cervantes JA, Meza-Bustillos JF, Resendiz-Hernández H, Suárez-Cantú IA, Ortega-Rivera OA, Salinas E, Escárcega-González CE, Morones-Ramírez JR. Re-sensitizing Ampicillin and Kanamycin-Resistant E. coli and S. aureus Using Synergistic Metal Micronutrients-Antibiotic Combinations. Front Bioeng Biotechnol 2020; 8:612. [PMID: 32671033 PMCID: PMC7327704 DOI: 10.3389/fbioe.2020.00612] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/19/2020] [Indexed: 01/19/2023] Open
Abstract
Due to the recent emergence of multi-drug resistant strains, the development of novel antimicrobial agents has become a critical issue. The use of micronutrient transition metals is a promising approach to overcome this problem since these compounds exhibit significant toxicity at low concentrations in prokaryotic cells. In this work, we demonstrate that at concentrations lower than their minimal inhibitory concentrations and in combination with different antibiotics, it is possible to mitigate the barriers to employ metallic micronutrients as therapeutic agents. Here, we show that when administered as a combinatorial treatment, Cu2+, Zn2+, Co2+, Cd2+, and Ni2+ increase susceptibility of Escherichia coli and Staphylococcus aureus to ampicillin and kanamycin. Furthermore, ampicillin-resistant E. coli is re-sensitized to ampicillin when the ampicillin is administered in combination with Cu2+, Cd2+, or Ni2. Similarly, Cu2+, Zn2+, or Cd2+ re-sensitize kanamycin-resistant E. coli and S. aureus to kanamycin when administered in a combinatorial treatment with those transition metals. Here, we demonstrate that for both susceptible and resistant bacteria, transition-metal micronutrients, and antibiotics interact synergistically in combinatorial treatments and exhibit increased effects when compared to the treatment with the antibiotic alone. Moreover, in vitro and in vivo assays, using a murine topical infection model, showed no toxicological effects of either treatment at the administered concentrations. Lastly, we show that combinatorial treatments can clear a murine topical infection caused by an antibiotic-resistant strain. Altogether, these results suggest that antibiotic-metallic micronutrient combinatorial treatments will play an important role in future developments of antimicrobial agents and treatments against infections caused by both susceptible and resistant strains.
Collapse
Affiliation(s)
- Javier Alberto Garza-Cervantes
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, UANL, San Nicolás de los Garza, Mexico.,Centro de Investigación en Biotecnologíay Nanotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, Apodaca, Mexico
| | - Jesus F Meza-Bustillos
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, UANL, San Nicolás de los Garza, Mexico
| | - Haziel Resendiz-Hernández
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, UANL, San Nicolás de los Garza, Mexico
| | - Ivan A Suárez-Cantú
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, UANL, San Nicolás de los Garza, Mexico
| | - Oscar Antonio Ortega-Rivera
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Eva Salinas
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Carlos Enrique Escárcega-González
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, UANL, San Nicolás de los Garza, Mexico.,Centro de Investigación en Biotecnologíay Nanotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, Apodaca, Mexico
| | - Jose Ruben Morones-Ramírez
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, UANL, San Nicolás de los Garza, Mexico.,Centro de Investigación en Biotecnologíay Nanotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, Apodaca, Mexico
| |
Collapse
|
11
|
Tsegka KG, Voulgaris GL, Kyriakidou M, Falagas ME. Intravenous fosfomycin for the treatment of patients with central nervous system infections: evaluation of the published evidence. Expert Rev Anti Infect Ther 2020; 18:657-668. [DOI: 10.1080/14787210.2020.1754193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Katerina G Tsegka
- Alfa Institute of Biomedical Sciences, Athens, Greece
- Department of Medicine, Henry Dunant Hospital Center, Athens, Greece
| | - Georgios L Voulgaris
- Alfa Institute of Biomedical Sciences, Athens, Greece
- Laboratory of Pharmacokinetics and Toxicology, Department of Pharmacy, 401 General Military Hospital, Athens, Greece
| | - Margarita Kyriakidou
- Alfa Institute of Biomedical Sciences, Athens, Greece
- School of Applied Mathematical and Physical Sciences, National Technical University, Athens, Greece
| | - Matthew E Falagas
- Alfa Institute of Biomedical Sciences, Athens, Greece
- Department of Medicine, Henry Dunant Hospital Center, Athens, Greece
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
12
|
Antibacterial Activity of combinatorial treatments composed of transition-metal/antibiotics against Mycobacterium tuberculosis. Sci Rep 2019; 9:5471. [PMID: 30940878 PMCID: PMC6445279 DOI: 10.1038/s41598-019-42049-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Notwithstanding evidence that tuberculosis (TB) is declining, one of the greatest concerns to public health is the emergence and spread of multi-drug resistant strains of Mycobacterium tuberculosis (MDR-TB). MDR-TB are defined as strains which are resistant to at least isoniazid (INH) and rifampicin, the two most potent TB drugs, and their increasing incidence is a serious concern. Recently, notable efforts have been spent on research to pursue novel treatments against MDR-TB, especially on synergistic drug combinations as they have the potential to improve TB treatment. Our research group has previously reported promising synergistic antimicrobial effects between transition-metal compounds and antibiotics in Gram-negative and Gram-positive bacteria. In this work, we evaluated antimycobacterial activity of transition-metals/antibiotics combinatorial treatments against first-line drug resistant strains of Mycobacterium tuberculosis. Our data showed that INH/AgNO3 combinatorial treatment had an additive effect (bactericidal activity) in an isoniazid-resistant clinical strain of Mycobacterium tuberculosis. Moreover, in vitro evaluation of cytotoxicity induced by both, the individual tratments of AgNO3 and INH and the combinatorial treatment of INH/AgNO3 in murine RAW 264.7 macrophages and human A549 lung cells; showed no toxic effects. Together, this data suggests that the INH/AgNO3 combinatorial treatment could be used in the development of new strategies to treat resistant strains of Mycobacterium tuberculosis.
Collapse
|
13
|
Gómara M, Ramón-García S. The FICI paradigm: Correcting flaws in antimicrobial in vitro synergy screens at their inception. Biochem Pharmacol 2019; 163:299-307. [PMID: 30836058 DOI: 10.1016/j.bcp.2019.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/01/2019] [Indexed: 10/27/2022]
Abstract
Antibiotics have become the corner stone of modern medicine. However, our society is currently facing one of the greatest challenges of its time: the emergence of antimicrobial resistance. It is estimated that if no new therapies are implemented by 2050, 10 million people will die worldwide every year as a result of infections caused by bacteria resistant to current antibiotics; new antimicrobials are thus urgently needed. However, drug development is a tedious and very costly endeavor of hundreds of millions that can take up to 15-20 years from the bench discovery to the bedside. Under this scenario, drug repurposing, which consists in identifying new uses for old, clinically approved drugs, has gathered momentum within the pharmaceutical industry. Because most of these drugs have safety and toxicity information packages available, clinical evaluation could be done in a much shorter period than standard timelines. Synergistic combinations of these clinically approved drugs could also be a promising approach to identify novel antimicrobial therapies that might provide rational choices of available drugs to shorten treatment, increase efficacy, reduce toxicity, prevent resistance and treat infections caused by drug-resistant strains. However, although simple in its conception, translating results from in vitro synergy screens into in vivo efficacy or the clinical practice has proven to be a paramount challenge. In this Commentary, we will discuss common flaws at the inception of synergy research programs, with a special focus on the use of the Fractional Inhibitory Concentration Index (FICI), and evaluate potential interventions that can be made at different developmental pre-clinical stages in order to improve the odds of translation from in vitro studies.
Collapse
Affiliation(s)
- Marta Gómara
- Mycobacterial Genetics Group, Department of Microbiology, Preventive Medicine and Public Health. Faculty of Medicine, University of Zaragoza, Spain
| | - Santiago Ramón-García
- Mycobacterial Genetics Group, Department of Microbiology, Preventive Medicine and Public Health. Faculty of Medicine, University of Zaragoza, Spain; Research & Development Agency of Aragon (ARAID) Foundation, Spain; CIBER Respiratory Diseases, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
14
|
Laishram S, Pragasam AK, Bakthavatchalam YD, Veeraraghavan B. An update on technical, interpretative and clinical relevance of antimicrobial synergy testing methodologies. Indian J Med Microbiol 2018; 35:445-468. [PMID: 29405135 DOI: 10.4103/ijmm.ijmm_17_189] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Testing for antimicrobial interactions has gained popularity in the last decade due to the increasing prevalence of drug-resistant organisms and limited options for the treatment of these infections. In vitro combination testing provides information, on which two or more antimicrobials can be combined for a good clinical outcome. Amongst the various in vitro methods of drug interactions, time-kill assay (TKA), checkerboard (CB) assay and E-test-based methods are most commonly used. Comparative performance of these methods reveals the TKA as the most promising method to detect synergistic combinations followed by CB assay and E-test. Various combinations of antimicrobials have been tested to demonstrate synergistic activity. Promising results were obtained for the combinations of meropenem plus colistin and rifampicin plus colistin against Acinetobacter baumannii, colistin plus carbapenem and carbapenem plus fluoroquinolones against Pseudomonas aeruginosa and colistin/polymyxin B plus rifampicin/meropenem against Klebsiella pneumoniae. Antagonism was detected in only few instances. The presence of synergy or antagonism with a combination seems to correlate with minimum inhibitory concentration of the agent and molecular mechanism involved in the resistance. Further studies need to be conducted to assess the utility of in vitro testing to predict clinical outcome and direct therapy for drug-resistant organisms.
Collapse
Affiliation(s)
- Shakti Laishram
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu,, India
| | - Agila Kumari Pragasam
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu,, India
| | | | - Balaji Veeraraghavan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu,, India
| |
Collapse
|
15
|
Optimization and Evaluation of Piperacillin-Tobramycin Combination Dosage Regimens against Pseudomonas aeruginosa for Patients with Altered Pharmacokinetics via the Hollow-Fiber Infection Model and Mechanism-Based Modeling. Antimicrob Agents Chemother 2018; 62:AAC.00078-18. [PMID: 29463528 DOI: 10.1128/aac.00078-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/12/2018] [Indexed: 02/07/2023] Open
Abstract
Augmented renal clearance (ARC) in critically ill patients can result in suboptimal drug exposures and treatment failure. Combination dosage regimens accounting for ARC have never been optimized and evaluated against Pseudomonas aeruginosa by use of the hollow-fiber infection model (HFIM). Using a P. aeruginosa isolate from a critically ill patient and static-concentration time-kill experiments (SCTKs), we studied clinically relevant piperacillin and tobramycin concentrations, alone and in combinations, against two inocula (105.8 and 107.6 CFU/ml) over 72 h. We subsequently evaluated the effects of optimized piperacillin (4 g every 4 h [q4h], given as 0.5-h infusions) plus tobramycin (5 mg/kg of body weight q24h, 7 mg/kg q24h, or 10 mg/kg q48h, given as 0.5-h infusions) regimens on killing and regrowth in the HFIM, simulating a creatinine clearance of 250 ml/min. Mechanism-based modeling was performed in S-ADAPT. In SCTKs, piperacillin plus tobramycin (except combinations with 8 mg/liter tobramycin and against the low inoculum) achieved synergistic killing (≥2 log10 versus the most active monotherapy at 48 h and 72 h) and prevented regrowth. Piperacillin monotherapy (4 g q4h) in the HFIM provided 2.4-log10 initial killing followed by regrowth at 24 h and resistance emergence. Tobramycin monotherapies displayed rapid initial killing (≥5 log10 at 13 h) followed by extensive regrowth. As predicted by mechanism-based modeling, the piperacillin plus tobramycin dosage regimens were synergistic and provided ≥5-log10 killing with resistance suppression over 8 days in the HFIM. Optimized piperacillin-tobramycin regimens provided significant bacterial killing and suppressed resistance emergence. These regimens appear to be highly promising for effective and early treatment, even in the near-worst-case scenario of ARC.
Collapse
|
16
|
Garza-Cervantes JA, Chávez-Reyes A, Castillo EC, García-Rivas G, Antonio Ortega-Rivera O, Salinas E, Ortiz-Martínez M, Gómez-Flores SL, Peña-Martínez JA, Pepi-Molina A, Treviño-González MT, Zarate X, Elena Cantú-Cárdenas M, Enrique Escarcega-Gonzalez C, Morones-Ramírez JR. Synergistic Antimicrobial Effects of Silver/Transition-metal Combinatorial Treatments. Sci Rep 2017; 7:903. [PMID: 28420878 PMCID: PMC5429853 DOI: 10.1038/s41598-017-01017-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/23/2017] [Indexed: 12/17/2022] Open
Abstract
Due to the emergence of multi-drug resistant strains, development of novel antibiotics has become a critical issue. One promising approach is the use of transition metals, since they exhibit rapid and significant toxicity, at low concentrations, in prokaryotic cells. Nevertheless, one main drawback of transition metals is their toxicity in eukaryotic cells. Here, we show that the barriers to use them as therapeutic agents could be mitigated by combining them with silver. We demonstrate that synergism of combinatorial treatments (Silver/transition metals, including Zn, Co, Cd, Ni, and Cu) increases up to 8-fold their antimicrobial effect, when compared to their individual effects, against E. coli and B. subtilis. We find that most combinatorial treatments exhibit synergistic antimicrobial effects at low/non-toxic concentrations to human keratinocyte cells, blast and melanoma rat cell lines. Moreover, we show that silver/(Cu, Ni, and Zn) increase prokaryotic cell permeability at sub-inhibitory concentrations, demonstrating this to be a possible mechanism of the synergistic behavior. Together, these results suggest that these combinatorial treatments will play an important role in the future development of antimicrobial agents and treatments against infections. In specific, the cytotoxicity experiments show that the combinations have great potential in the treatment of topical infections.
Collapse
Affiliation(s)
- Javier A Garza-Cervantes
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
- Centro de Investigacion en Biotecnologia y Nanotoxicologia, Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Parque de Investigacion e Innovacion Tecnologica, Km. 10 autopista al Aeropuerto Internacional Mariano Escobedo, Apodaca, Nuevo Leon, 66629, Mexico
| | - Arturo Chávez-Reyes
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Monterrey, Parque PIIT, 66600, Apodaca, Nuevo León, Mexico
| | - Elena C Castillo
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
- Centro de Investigación Biomédica, Hospital Zambrano-Hellion, Tecnologico de Monterrey, San Pedro Garza-García, Nuevo León, Mexico
| | - Gerardo García-Rivas
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
- Centro de Investigación Biomédica, Hospital Zambrano-Hellion, Tecnologico de Monterrey, San Pedro Garza-García, Nuevo León, Mexico
| | - Oscar Antonio Ortega-Rivera
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Colonia Ciudad Universitaria C.P., 20131, Aguascalientes, Aguascalientes, Mexico
| | - Eva Salinas
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Colonia Ciudad Universitaria C.P., 20131, Aguascalientes, Aguascalientes, Mexico
| | - Margarita Ortiz-Martínez
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
| | - Sara Leticia Gómez-Flores
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
| | - Jorge A Peña-Martínez
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
| | - Alan Pepi-Molina
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Biológicas, Av. Universidad s/n, Cd. Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
| | - Mario T Treviño-González
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ingeniería Mecánica y Eléctrica, Av. Universidad s/n, Cd. Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
| | - Xristo Zarate
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
- Centro de Investigacion en Biotecnologia y Nanotoxicologia, Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Parque de Investigacion e Innovacion Tecnologica, Km. 10 autopista al Aeropuerto Internacional Mariano Escobedo, Apodaca, Nuevo Leon, 66629, Mexico
| | - María Elena Cantú-Cárdenas
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
- Centro de Investigacion en Biotecnologia y Nanotoxicologia, Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Parque de Investigacion e Innovacion Tecnologica, Km. 10 autopista al Aeropuerto Internacional Mariano Escobedo, Apodaca, Nuevo Leon, 66629, Mexico
| | - Carlos Enrique Escarcega-Gonzalez
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico
- Centro de Investigacion en Biotecnologia y Nanotoxicologia, Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Parque de Investigacion e Innovacion Tecnologica, Km. 10 autopista al Aeropuerto Internacional Mariano Escobedo, Apodaca, Nuevo Leon, 66629, Mexico
| | - J Rubén Morones-Ramírez
- Universidad Autónoma de Nuevo León, UANL, Facultad de Ciencias Químicas, Av. Universidad s/n, Cd, Universitaria, 66451, San Nicolás de los Garza, NL, Mexico.
- Centro de Investigacion en Biotecnologia y Nanotoxicologia, Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Parque de Investigacion e Innovacion Tecnologica, Km. 10 autopista al Aeropuerto Internacional Mariano Escobedo, Apodaca, Nuevo Leon, 66629, Mexico.
| |
Collapse
|
17
|
Abstract
The treatment of bacterial infections suffers from two major problems: spread of multidrug-resistant (MDR) or extensively drug-resistant (XDR) pathogens and lack of development of new antibiotics active against such MDR and XDR bacteria. As a result, physicians have turned to older antibiotics, such as polymyxins, tetracyclines, and aminoglycosides. Lately, due to development of resistance to these agents, fosfomycin has gained attention, as it has remained active against both Gram-positive and Gram-negative MDR and XDR bacteria. New data of higher quality have become available, and several issues were clarified further. In this review, we summarize the available fosfomycin data regarding pharmacokinetic and pharmacodynamic properties, the in vitro activity against susceptible and antibiotic-resistant bacteria, mechanisms of resistance and development of resistance during treatment, synergy and antagonism with other antibiotics, clinical effectiveness, and adverse events. Issues that need to be studied further are also discussed.
Collapse
|
18
|
Abstract
Fosfomycin was discovered over four decades ago, yet has drawn renewed interest as an agent active against a range of multidrug-resistant (MDR) and extensively drug-resistant (XDR) pathogens. Its unique mechanism of action and broad spectrum of activity makes it a promising candidate in the treatment of various MDR/XDR infections. There has been a surge of in vitro data on its activity against MDR/XDR organisms, both when used as a single agent and in combination with other agents. In the United States, fosfomycin is only approved in an oral formulation for the treatment of acute uncomplicated urinary tract infections (UTIs), whereas in some countries both oral and intravenous formulations are available for various indications. Fosfomycin has minimal interactions with other medications and has a relatively favorable safety profile, with diarrhea being the most common adverse reaction. Fosfomycin has low protein binding and is excreted primarily unchanged in the urine. The clinical outcomes of patients treated with fosfomycin are favorable for uncomplicated UTIs, but data are limited for use in other conditions. Fosfomycin maintains activity against most Enterobacteriaceae including Escherichia coli, but plasmid-mediated resistance due to inactivation have appeared in recent years, which has the potential to compromise its use in the future. In this review, we summarize the current knowledge of this resurgent agent and its role in our antimicrobial armamentarium.
Collapse
|
19
|
Viganor L, Galdino ACM, Nunes APF, Santos KRN, Branquinha MH, Devereux M, Kellett A, McCann M, Santos ALS. Anti-Pseudomonas aeruginosa activity of 1,10-phenanthroline-based drugs against both planktonic- and biofilm-growing cells. J Antimicrob Chemother 2015; 71:128-34. [PMID: 26416778 DOI: 10.1093/jac/dkv292] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/21/2015] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES The beneficial antimicrobial properties of 1,10-phenanthroline (phen)-based drugs, together with the imperative need to develop new chemotherapeutic options for prevention/treatment of infections caused by MDR Gram-negative bacteria, led us to evaluate the effects of phen, 1,10-phenanthroline-5,6-dione (phendione), [Ag(phendione)2]ClO4 and [Cu(phendione)3](ClO4)2·4H2O on planktonic- and biofilm-growing Pseudomonas aeruginosa. METHODS Thirty-two non-duplicated Brazilian clinical isolates of P. aeruginosa with distinct genetic backgrounds were used in all experiments. The effect of test compounds on planktonic bacterial proliferation was determined as recommended by CLSI protocol. The effect on biofilm formation was evaluated by crystal violet incorporation (biomass determination) and XTT (viability assay). Mature biofilm disorganization was evidenced by staining with crystal violet. RESULTS Phen-based compounds presented anti-P. aeruginosa activity, but with different potencies concerning the geometric mean MIC: [Cu(phendione)3](2+) (7.76 μM) > [Ag(phendione)2](+) (14.05 μM) > phendione (31.15 μM) > phen (579.28 μM). MICs of each compound were similar irrespective of whether the P. aeruginosa isolates were susceptible or resistant to classical antimicrobials (ceftazidime, meropenem and imipenem). The pretreatment of bacteria with phen, phendione and phendione's metal derivatives at 0.5 × MIC value inhibited biofilm formation, particularly the use of [Cu(phendione)3](2+) and [Ag(phendione)2](+), which significantly reduced both biomass (48% and 44%, respectively) and viability (78% and 77%, respectively). The compounds studied also disrupted mature biofilm in a dose-dependent manner, especially [Ag(phendione)2](+) and [Cu(phendione)3](2+) (IC50, 9.39 and 10.16 μM, respectively). CONCLUSIONS Coordination of phendione to Ag(+) and Cu(2+) represents a new promising group of anti-infective agents, which revealed a potent anti-P. aeruginosa action against both planktonic- and biofilm-growing cells.
Collapse
Affiliation(s)
- Livia Viganor
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil The Inorganic Pharmaceutical and Biomimetic Research Centre, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
| | - Anna Clara M Galdino
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil Biochemistry Post-Graduation Programme, Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula F Nunes
- Pathology Department and Infection Diseases Post-Graduation Programme, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Kátia R N Santos
- Medical Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marta H Branquinha
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michael Devereux
- The Inorganic Pharmaceutical and Biomimetic Research Centre, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
| | - Andrew Kellett
- School of Chemical Sciences and the National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Malachy McCann
- Chemistry Department, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - André L S Santos
- General Microbiology Department, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil The Inorganic Pharmaceutical and Biomimetic Research Centre, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
| |
Collapse
|
20
|
Antimicrobial Activity of Fosfomycin-Tobramycin Combination against Pseudomonas aeruginosa Isolates Assessed by Time-Kill Assays and Mutant Prevention Concentrations. Antimicrob Agents Chemother 2015. [PMID: 26195514 DOI: 10.1128/aac.00822-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The antibacterial activity of fosfomycin-tobramycin combination was studied by time-kill assay in eight Pseudomonas aeruginosa clinical isolates belonging to the fosfomycin wild-type population (MIC = 64 μg/ml) but with different tobramycin susceptibilities (MIC range, 1 to 64 μg/ml). The mutant prevention concentration (MPC) and mutant selection window (MSW) were determined in five of these strains (tobramycin MIC range, 1 to 64 μg/ml) in aerobic and anaerobic conditions simulating environments that are present in biofilm-mediated infections. Fosfomycin-tobramycin was synergistic and bactericidal for the isolates with mutations in the mexZ repressor gene, with a tobramycin MIC of 4 μg/ml. This effect was not observed in strains displaying tobramycin MICs of 1 to 2 μg/ml due to the strong bactericidal effect of tobramycin alone. Fosfomycin presented higher MPC values (range, 2,048 to >2,048 μg/ml) in aerobic and anaerobic conditions than did tobramycin (range, 16 to 256 μg/ml). Interestingly, the association rendered narrow or even null MSWs in the two conditions. However, for isolates with high-level tobramycin resistance that harbored aminoglycoside nucleotidyltransferases, time-kill assays showed no synergy, with wide MSWs in the two environments. glpT gene mutations responsible for fosfomycin resistance in P. aeruginosa were determined in fosfomycin-susceptible wild-type strains and mutant derivatives recovered from MPC studies. All mutant derivatives had changes in the GlpT amino acid sequence, which resulted in a truncated permease responsible for fosfomycin resistance. These results suggest that fosfomycin-tobramycin can be an alternative for infections due to P. aeruginosa since it has demonstrated synergistic and bactericidal activity in susceptible isolates and those with low-level tobramycin resistance. It also prevents the emergence of resistant mutants in either aerobic or anaerobic environments.
Collapse
|
21
|
Reffert JL, Smith WJ. Fosfomycin for the treatment of resistant gram-negative bacterial infections. Insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy 2014; 34:845-57. [PMID: 24782335 DOI: 10.1002/phar.1434] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The antimicrobial agent fosfomycin was discovered in 1969, at a time when bacteria had not yet developed extended-spectrum β-lactamases or carbapenemases. Decades later, it is not uncommon for gram-negative organisms to be multidrug-resistant and even pan-resistant to available antibiotic regimens, leaving clinicians with few therapeutic alternatives. Because fosfomycin has been shown to retain activity against these virulent pathogens, there is renewed interest in its use as a therapeutic agent. Fosfomycin formulations including fosfomycin disodium and the newer tromethamine salt are less toxic than other alternatives and are attractive options for resistant gram-negative and gram-positive infections. Oral fosfomycin tromethamine is approved for urinary tract infections in the United States, and an intravenous formulation is also available outside of the United States for systemic disease. The bactericidal action of fosfomycin occurs at an earlier step in cell wall synthesis than that of β-lactam antibiotics. From an in vitro standpoint, fosfomycin generally has high activity against ESBL- and carbapenemase-producing Enterobacteriaceae; multidrug-resistant Pseudomonas aeruginosa susceptibility appears to be more dependent on the local antibiogram. Fosfomycin formulations have a large volume of distribution, penetrate biofilms, and concentrate in the urine. Both oral and intravenous fosfomycin formulations are effective for a wide range of gram-negative infections and disease severities; however, clinical studies are limited. Fosfomycin formulations are well-tolerated, and mild gastrointestinal distress is the most common adverse effect. The primary limitations of fosfomycin are the lack of established regimens for complicated infections and the lack of availability of the intravenous formulation in the United States. Further study of this promising agent seems warranted in the current climate of antibiotic resistance.
Collapse
|