1
|
Kavanaugh LG, Dey D, Shafer WM, Conn GL. Structural and functional diversity of Resistance-Nodulation-Division (RND) efflux pump transporters with implications for antimicrobial resistance. Microbiol Mol Biol Rev 2024; 88:e0008923. [PMID: 39235227 PMCID: PMC11426026 DOI: 10.1128/mmbr.00089-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
SUMMARYThe discovery of bacterial efflux pumps significantly advanced our understanding of how bacteria can resist cytotoxic compounds that they encounter. Within the structurally and functionally distinct families of efflux pumps, those of the Resistance-Nodulation-Division (RND) superfamily are noteworthy for their ability to reduce the intracellular concentration of structurally diverse antimicrobials. RND systems are possessed by many Gram-negative bacteria, including those causing serious human disease, and frequently contribute to resistance to multiple antibiotics. Herein, we review the current literature on the structure-function relationships of representative transporter proteins of tripartite RND efflux pumps of clinically important pathogens. We emphasize their contribution to bacterial resistance to clinically used antibiotics, host defense antimicrobials and other biocides, as well as highlighting structural similarities and differences among efflux transporters that help bacteria survive in the face of antimicrobials. Furthermore, we discuss technical advances that have facilitated and advanced efflux pump research and suggest future areas of investigation that will advance antimicrobial development efforts.
Collapse
Affiliation(s)
- Logan G Kavanaugh
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
- Graduate Program in Microbiology and Molecular Genetics, Emory University, Atlanta, Georgia, USA
| | - Debayan Dey
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William M Shafer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Laboratories of Microbial Pathogenesis, VA Medical Research Service, Veterans Affairs Medical Center, Decatur, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Graeme L Conn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Scoffone VC, Barbieri G, Irudal S, Trespidi G, Buroni S. New Antimicrobial Strategies to Treat Multi-Drug Resistant Infections Caused by Gram-Negatives in Cystic Fibrosis. Antibiotics (Basel) 2024; 13:71. [PMID: 38247630 PMCID: PMC10812592 DOI: 10.3390/antibiotics13010071] [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: 12/20/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
People with cystic fibrosis (CF) suffer from recurrent bacterial infections which induce inflammation, lung tissue damage and failure of the respiratory system. Prolonged exposure to combinatorial antibiotic therapies triggers the appearance of multi-drug resistant (MDR) bacteria. The development of alternative antimicrobial strategies may provide a way to mitigate antimicrobial resistance. Here we discuss different alternative approaches to the use of classic antibiotics: anti-virulence and anti-biofilm compounds which exert a low selective pressure; phage therapies that represent an alternative strategy with a high therapeutic potential; new methods helping antibiotics activity such as adjuvants; and antimicrobial peptides and nanoparticle formulations. Their mechanisms and in vitro and in vivo efficacy are described, in order to figure out a complete landscape of new alternative approaches to fight MDR Gram-negative CF pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Silvia Buroni
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.B.); (S.I.); (G.T.)
| |
Collapse
|
3
|
Pipitone G, Di Bella S, Maraolo AE, Granata G, Gatti M, Principe L, Russo A, Gizzi A, Pallone R, Cascio A, Iaria C. Intravenous Fosfomycin for Systemic Multidrug-Resistant Pseudomonas aeruginosa Infections. Antibiotics (Basel) 2023; 12:1653. [PMID: 38136687 PMCID: PMC10741068 DOI: 10.3390/antibiotics12121653] [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: 10/03/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Human Pseudomonas infections have high morbidity and mortality rates. Pseudomonas bacteria can cause sepsis or septic shock; they produce biofilm and commonly exhibit a multidrug-resistant phenotype. The choice of antimicrobial therapy in many cases is challenging, and deep knowledge of clinical, microbiological, and pharmacological issues is required. Intravenous fosfomycin is being repurposed in a combination given its favorable pharmacokinetic/pharmacodynamic properties (a small molecule with favorable kinetic both in bloodstream infection and in deep-seated infections), antibiofilm activity, and its interesting synergistic effects with other antimicrobials. Recent literature on epidemiological, microbiological, pharmacological, and clinical data on intravenous fosfomycin therapy against Pseudomonas is herein reviewed and discussed.
Collapse
Affiliation(s)
- Giuseppe Pipitone
- Infectious Diseases Unit, ARNAS Civico-Di Cristina Hospital, 90127 Palermo, Italy; (G.P.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy
| | - Alberto Enrico Maraolo
- First Division of Infectious Diseases, Cotugno Hospital, Azienda Ospedaliera dei Colli, 80131 Naples, Italy
| | - Guido Granata
- Clinical and Research Department for Infectious Diseases, National Institute for Infectious Diseases L. Spallanzani, IRCCS, 00149 Rome, Italy
| | - Milo Gatti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Clinical Pharmacology Unit, IRCCS University Hospital of Bologna, 40138 Bologna, Italy
| | - Luigi Principe
- Microbiology and Virology Unit, Great Metropolitan Hospital “Bianchi-Melacrino-Morelli”, 89133 Reggio Calabria, Italy
| | - Alessandro Russo
- Infectious and Tropical Diseases Unit, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy
| | - Andrea Gizzi
- Infectious Diseases Unit, ARNAS Civico-Di Cristina Hospital, 90127 Palermo, Italy; (G.P.)
- Infectious Diseases Unit, University Hospital P. Giaccone, 90127 Palermo, Italy
| | - Rita Pallone
- Infectious and Tropical Diseases Unit, University Hospital “Renato Dulbecco”, 88100 Catanzaro, Italy
| | - Antonio Cascio
- Infectious Diseases Unit, University Hospital P. Giaccone, 90127 Palermo, Italy
| | - Chiara Iaria
- Infectious Diseases Unit, ARNAS Civico-Di Cristina Hospital, 90127 Palermo, Italy; (G.P.)
| |
Collapse
|
4
|
Gandhi K, Dhiman S, Arora R, Ramirez DM, Ramirez D, Arthur G, Schweizer F. Exploring Antibiotic-Potentiating Effects of Tobramycin-Deferiprone Conjugates in Pseudomonas aeruginosa. Antibiotics (Basel) 2023; 12:1261. [PMID: 37627681 PMCID: PMC10451322 DOI: 10.3390/antibiotics12081261] [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: 07/12/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Metal ions, including Fe3+, affect the target site binding of some antibiotics and control the porin- and siderophore-mediated uptake of antibiotics. Amphiphilic tobramycins are an emerging class of antibiotic potentiators capable of synergizing with multiple classes of antibiotics against Gram-negative bacteria, including Pseudomonas aeruginosa. To study how the antibiotic-potentiating effect of amphiphilic tobramycins is affected by the presence of intermolecular iron chelators, we conjugated the FDA-approved iron chelator deferiprone (DEF) to tobramycin (TOB). Three TOB-DEF conjugates differing in the length of the carbon tether were prepared and tested for antibacterial activity and synergistic relationships with a panel of antibiotics against clinical isolates of P. aeruginosa. While all TOB-DEF conjugates were inactive against P. aeruginosa, the TOB-DEF conjugates strongly synergized with outer-membrane-impermeable antibiotics, such as novobiocin and rifampicin. Among the three TOB-DEF conjugates, 1c containing a C12 tether showed a remarkable and selective potentiating effect to improve the susceptibility of multidrug-resistant P. aeruginosa isolates to tetracyclines when compared with other antibiotics. However, the antibacterial activity and antibiotic-potentiating effect of the optimized conjugate was not enhanced under iron-depleted conditions, indicating that the function of the antibiotic potentiator is not affected by the Fe3+ concentration.
Collapse
Affiliation(s)
- Karan Gandhi
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (K.G.); (S.D.); (R.A.); (D.M.R.); (D.R.)
| | - Shiv Dhiman
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (K.G.); (S.D.); (R.A.); (D.M.R.); (D.R.)
| | - Rajat Arora
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (K.G.); (S.D.); (R.A.); (D.M.R.); (D.R.)
| | - Danzel Marie Ramirez
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (K.G.); (S.D.); (R.A.); (D.M.R.); (D.R.)
| | - Danyel Ramirez
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (K.G.); (S.D.); (R.A.); (D.M.R.); (D.R.)
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Frank Schweizer
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (K.G.); (S.D.); (R.A.); (D.M.R.); (D.R.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3R 0J9, Canada
| |
Collapse
|
5
|
Lyu J, Chen H, Bao J, Liu S, Chen Y, Cui X, Guo C, Gu B, Li L. Clinical Distribution and Drug Resistance of Pseudomonas aeruginosa in Guangzhou, China from 2017 to 2021. J Clin Med 2023; 12:jcm12031189. [PMID: 36769837 PMCID: PMC9917919 DOI: 10.3390/jcm12031189] [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: 11/08/2022] [Revised: 01/05/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023] Open
Abstract
The aim of the current study was to analyse the distribution of antimicrobial drug resistance (AMR) among Pseudomonas aeruginosa (P. aeruginosa, PA) isolates from Guangdong Provincial People's Hospital (GDPH) from 2017 to 2021, and the impact of the COVID-19 outbreak on changes in the clinical distribution and drug resistance rate of P. aeruginosa to establish guidelines for empiric therapy. Electronic clinical data registry records from 2017 to 2021 were retrospectively analysed to study the AMR among P. aeruginosa strains from GDPH. The strains were identified by VITEK 2 Compact and MALDI-TOF MS, MIC method or Kirby-Bauer method for antibiotic susceptibility testing. The results were interpreted according to the CLSI 2020 standard, and the data were analysed using WHONET 5.6 and SPSS 23.0 software. A total of 3036 P. aeruginosa strains were detected in the hospital from 2017 to 2021, and they were primarily distributed in the ICU (n = 1207, 39.8%). The most frequent specimens were respiratory tract samples (59.6%). The detection rate for P. aeruginosa in 5 years was highest in September, and the population distribution was primarily male(68.2%). For the trend in the drug resistance rate, the 5-year drug resistance rate of imipenem (22.4%), aztreonam (21.5%) and meropenem (19.3%) remained at high levels. The resistance rate of cefepime decreased from 9.4% to 4.8%, showing a decreasing trend year by year (p < 0.001). The antibiotics with low resistance rates were aminoglycoside antibiotics, which were gentamicin (4.4%), tobramycin (4.3%), and amikacin (1.4%), but amikacin showed an increasing trend year by year (p = 0.008). Our analysis indicated that the detection rate of clinically resistant P. aeruginosa strains showed an upwards trend, and the number of multidrug-resistant (MDR) strains increased year by year, which will lead to stronger pathogenicity and mortality. However, after the outbreak of COVID-19 in 2020, the growth trend in the number of MDR bacteria slowed, presumably due to the strict epidemic prevention and control measures in China. This observation suggests that we should reasonably use antibiotics and treatment programs in the prevention and control of P. aeruginosa infection. Additionally, health prevention and control after the outbreak of the COVID-19 epidemic (such as wearing masks, washing hands with disinfectant, etc., which reduced the prevalence of drug resistance) led to a slowdown in the growth of the drug resistance rate of P. aeruginosa in hospitals, effectively reducing the occurrence and development of drug resistance, and saving patient's treatment costs and time.
Collapse
Affiliation(s)
- Jingwen Lyu
- Department of Clinical Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Huimin Chen
- Department of Clinical Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Jinwei Bao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Suling Liu
- Department of Clinical Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Yiling Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xuxia Cui
- Department of Clinical Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Caixia Guo
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou 511316, China
- Correspondence: (C.G.); (B.G.); (L.L.)
| | - Bing Gu
- Department of Clinical Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
- Correspondence: (C.G.); (B.G.); (L.L.)
| | - Lu Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Correspondence: (C.G.); (B.G.); (L.L.)
| |
Collapse
|
6
|
Chetri S. The culmination of multidrug-resistant efflux pumps vs. meager antibiotic arsenal era: Urgent need for an improved new generation of EPIs. Front Microbiol 2023; 14:1149418. [PMID: 37138605 PMCID: PMC10149990 DOI: 10.3389/fmicb.2023.1149418] [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: 01/21/2023] [Accepted: 03/13/2023] [Indexed: 05/05/2023] Open
Abstract
Efflux pumps function as an advanced defense system against antimicrobials by reducing the concentration of drugs inside the bacteria and extruding the substances outside. Various extraneous substances, including antimicrobials, toxic heavy metals, dyes, and detergents, have been removed by this protective barrier composed of diverse transporter proteins found in between the cell membrane and the periplasm within the bacterial cell. In this review, multiple efflux pump families have been analytically and widely outlined, and their potential applications have been discussed in detail. Additionally, this review also discusses a variety of biological functions of efflux pumps, including their role in the formation of biofilms, quorum sensing, their survivability, and the virulence in bacteria, and the genes/proteins associated with efflux pumps have also been explored for their potential relevance to antimicrobial resistance and antibiotic residue detection. A final discussion centers around efflux pump inhibitors, particularly those derived from plants.
Collapse
|
7
|
Wesseling CJ, Martin NI. Synergy by Perturbing the Gram-Negative Outer Membrane: Opening the Door for Gram-Positive Specific Antibiotics. ACS Infect Dis 2022; 8:1731-1757. [PMID: 35946799 PMCID: PMC9469101 DOI: 10.1021/acsinfecdis.2c00193] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
New approaches to target antibacterial agents toward Gram-negative bacteria are key, given the rise of antibiotic resistance. Since the discovery of polymyxin B nonapeptide as a potent Gram-negative outer membrane (OM)-permeabilizing synergist in the early 1980s, a vast amount of literature on such synergists has been published. This Review addresses a range of peptide-based and small organic compounds that disrupt the OM to elicit a synergistic effect with antibiotics that are otherwise inactive toward Gram-negative bacteria, with synergy defined as a fractional inhibitory concentration index (FICI) of <0.5. Another requirement for the inclusion of the synergists here covered is their potentiation of a specific set of clinically used antibiotics: erythromycin, rifampicin, novobiocin, or vancomycin. In addition, we have focused on those synergists with reported activity against Gram-negative members of the ESKAPE family of pathogens namely, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, and/or Acinetobacter baumannii. In cases where the FICI values were not directly reported in the primary literature but could be calculated from the published data, we have done so, allowing for more direct comparison of potency with other synergists. We also address the hemolytic activity of the various OM-disrupting synergists reported in the literature, an effect that is often downplayed but is of key importance in assessing the selectivity of such compounds for Gram-negative bacteria.
Collapse
|
8
|
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
|
9
|
Huang L, Wu C, Gao H, Xu C, Dai M, Huang L, Hao H, Wang X, Cheng G. Bacterial Multidrug Efflux Pumps at the Frontline of Antimicrobial Resistance: An Overview. Antibiotics (Basel) 2022; 11:antibiotics11040520. [PMID: 35453271 PMCID: PMC9032748 DOI: 10.3390/antibiotics11040520] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/01/2022] [Accepted: 04/08/2022] [Indexed: 02/06/2023] Open
Abstract
Multidrug efflux pumps function at the frontline to protect bacteria against antimicrobials by decreasing the intracellular concentration of drugs. This protective barrier consists of a series of transporter proteins, which are located in the bacterial cell membrane and periplasm and remove diverse extraneous substrates, including antimicrobials, organic solvents, toxic heavy metals, etc., from bacterial cells. This review systematically and comprehensively summarizes the functions of multiple efflux pumps families and discusses their potential applications. The biological functions of efflux pumps including their promotion of multidrug resistance, biofilm formation, quorum sensing, and survival and pathogenicity of bacteria are elucidated. The potential applications of efflux pump-related genes/proteins for the detection of antibiotic residues and antimicrobial resistance are also analyzed. Last but not least, efflux pump inhibitors, especially those of plant origin, are discussed.
Collapse
|
10
|
Sharma S, Rao R, Reeve SM, Phelps GA, Bharatham N, Katagihallimath N, Ramachandran V, Raveendran S, Sarma M, Nath A, Thomas T, Manickam D, Nagaraj S, Balasubramanian V, Lee RE, Hameed P S, Datta S. Azaindole Based Potentiator of Antibiotics against Gram-Negative Bacteria. ACS Infect Dis 2021; 7:3009-3024. [PMID: 34699190 DOI: 10.1021/acsinfecdis.1c00171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We discovered azaindole-based compounds with weak innate activity that exhibit substantial potentiation of antibacterial activities of different antibiotics, viz., rifampicin, erythromycin, solithromycin, and novobiocin in Gram-negative bacteria. In the presence of the azaindole derivatives, these antibiotics exhibited submicromolar minimum inhibitory concentrations (MICs) against Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii. The fold improvements in MIC of these antibiotics that were otherwise weak or inactive on their own against these bacteria were also observed against drug-resistant clinical isolates. Our studies indicate that this selective potentiation is probably through destabilization of the outer membrane's integrity, known to be regulated by the lipopolysaccharides (LPS). Thus, the azaindole based compounds described here open opportunities for those antibiotics that are otherwise ineffective due to LPS mediated entry barriers in Gram-negative bacteria.
Collapse
Affiliation(s)
- Sreevalli Sharma
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka-560064, India
| | - Ranga Rao
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka-560064, India
| | - Stephanie M. Reeve
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Gregory A. Phelps
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Nagakumar Bharatham
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka-560064, India
| | - Nainesh Katagihallimath
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka-560064, India
| | - Vasanthi Ramachandran
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka-560064, India
| | - Savitha Raveendran
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
| | - Maitrayee Sarma
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
| | - Anubha Nath
- St. John’s Research Institute, Bengaluru, Karnataka-560034, India
| | - Teby Thomas
- St. John’s Research Institute, Bengaluru, Karnataka-560034, India
| | - Dhanasekaran Manickam
- Syngene International Ltd., Plot 2 & 3, Bommasandra Industrial Estate - Phase-IV, Bommasandra-Jigani Link Road, Bengaluru, Karnataka 560099, India
| | - Savitha Nagaraj
- St. John’s Medical Hospital, Bengaluru, Karnataka-560034, India
| | - V. Balasubramanian
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
| | - Richard E. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Shahul Hameed P
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka-560064, India
| | - Santanu Datta
- BUGWORKS Research India Pvt. Ltd., Centre for Cellular & Molecular Platforms, GKVK, Bellary Rd, Bangalore, Karnataka 560065, India
| |
Collapse
|
11
|
Dezanet C, Kempf J, Mingeot-Leclercq MP, Décout JL. Amphiphilic Aminoglycosides as Medicinal Agents. Int J Mol Sci 2020; 21:E7411. [PMID: 33049963 PMCID: PMC7583001 DOI: 10.3390/ijms21197411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/27/2020] [Accepted: 10/02/2020] [Indexed: 12/25/2022] Open
Abstract
The conjugation of hydrophobic group(s) to the polycationic hydrophilic core of the antibiotic drugs aminoglycosides (AGs), targeting ribosomal RNA, has led to the development of amphiphilic aminoglycosides (AAGs). These drugs exhibit numerous biological effects, including good antibacterial effects against susceptible and multidrug-resistant bacteria due to the targeting of bacterial membranes. In the first part of this review, we summarize our work in identifying and developing broad-spectrum antibacterial AAGs that constitute a new class of antibiotic agents acting on bacterial membranes. The target-shift strongly improves antibiotic activity against bacterial strains that are resistant to the parent AG drugs and to antibiotic drugs of other classes, and renders the emergence of resistant Pseudomonas aeruginosa strains highly difficult. Structure-activity and structure-eukaryotic cytotoxicity relationships, specificity and barriers that need to be crossed in their development as antibacterial agents are delineated, with a focus on their targets in membranes, lipopolysaccharides (LPS) and cardiolipin (CL), and the corresponding mode of action against Gram-negative bacteria. At the end of the first part, we summarize the other recent advances in the field of antibacterial AAGs, mainly published since 2016, with an emphasis on the emerging AAGs which are made of an AG core conjugated to an adjuvant or an antibiotic drug of another class (antibiotic hybrids). In the second part, we briefly illustrate other biological and biochemical effects of AAGs, i.e., their antifungal activity, their use as delivery vehicles of nucleic acids, of short peptide (polyamide) nucleic acids (PNAs) and of drugs, as well as their ability to cleave DNA at abasic sites and to inhibit the functioning of connexin hemichannels. Finally, we discuss some aspects of structure-activity relationships in order to explain and improve the target selectivity of AAGs.
Collapse
Affiliation(s)
- Clément Dezanet
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| | - Julie Kempf
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| | - Marie-Paule Mingeot-Leclercq
- Cellular and Molecular Pharmacology Unit, Louvain Drug Research Institute, Catholic University of Louvain, Avenue E. Mounier 73, UCL B1.73.05, 1200 Brussels, Belgium
| | - Jean-Luc Décout
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| |
Collapse
|
12
|
Silva Júnior VV, Raposo BL, Lopes ACS, Araújo PSR, Fontes A, Cabral Filho PE, Maciel MAV. Activity of carbonyl cyanide-3-chlorophenylhydrazone on biofilm formation and antimicrobial resistance in Pseudomonas aeruginosa using quantum dots-meropenem conjugates as nanotools. Methods Appl Fluoresc 2020; 8:045005. [PMID: 33021210 DOI: 10.1088/2050-6120/aba7a2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hospital infections associated with multidrug-resistant (MDR) Pseudomonas aeruginosa are a worldwide public health problem. Efflux systems and biofilm formation are mechanisms related to resistance to carbapenemics. In this study, quantum dots (QDs) were used to evaluate the effect of carbonyl cyanide-3-chlorophenylhydrazone (CCCP), an efflux pump system inhibitor, on biofilm formation and antimicrobial resistance profile of P. aeruginosa strains. For this, QDs were covalently conjugated to meropenem (MPM) and incubated with a P. aeruginosa resistant isolate (P118) or a control sensitive strain (ATCC Pa27853). P118 was also analyzed with conjugates after previous CCCP efflux inhibitor incubation. Fluorescence microscopy images showed that both sensitive and resistant bacteria were efficiently labeled. Nevertheless, P118 isolates presented fluorescent cell agglomerates, suggesting biofilm formation. The addition of the CCCP changed the labeling profile of the resistant isolate, and the absence of agglomerates was observed, indicating no biofilm formation. Genetic assays revealed the presence of MexA and MexE genes encoding channel proteins from efflux pump systems in both resistant and sensitive strains. Disk-diffusion and broth microdilution tests determined drug susceptibility profiles in the presence and absence of CCCP for P118 isolates. We verified that the CCCP efflux system inhibitor may contribute to P. aeruginosa resistant phenotype reduction for some antimicrobials. This study verified the efficiency of QD-MPM conjugates to trigger and study biofilm formation, or its inhibition, before and after CCCP addition. QDs conjugated to antimicrobials can be used as nanotools to investigate multidrug-resistant bacterial strains on biofilm formation.
Collapse
Affiliation(s)
- Valdemir V Silva Júnior
- Coordenação de Área Medicina Tropical, Centro de Ciências Médicas, Universidade Federal de Pernambuco, 50670-901, Recife, Pernambuco, Brasil
| | | | | | | | | | | | | |
Collapse
|
13
|
AlMatar M, Albarri O, Makky EA, Köksal F. Efflux pump inhibitors: new updates. Pharmacol Rep 2020; 73:1-16. [PMID: 32946075 DOI: 10.1007/s43440-020-00160-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/17/2022]
Abstract
The discovery of antibiotics ought to have ended the issue of bacterial infections, but this was not the case as it has led to the evolution of various mechanisms of bacterial resistance against various antibiotics. The efflux pump remains one of the mechanisms through which organisms develop resistance against antibiotics; this is because organisms can extrude most of the clinically relevant antibiotics from the interior cell environment to the exterior environment via the efflux pumps. Efflux pumps are thought to contribute significantly to biofilm formation as highlighted by various studies. Therefore, the inhibition of these efflux pumps can be a potential way of improving the activity of antibiotics, particularly now that the discovery of novel antibiotics is becoming tedious. Efflux pump inhibitors (EPIs) are molecules that can inhibit efflux pumps; they have been considered potential therapeutic agents for rejuvenating the activity of antibiotics that have already lost their activity against bacteria. However, studies are yet to determine the specific substrates for such pumps; the effect of altered efflux activity of these pumps on biofilm formation is still being investigated. A clear knowledge of the involvement of efflux pumps in biofilm development could aid in developing new agents that can interfere with their function and help to prevent biofilms formation; thereby, improving the outcome of treatment strategies. This review focuses on the novel update of EPIs and discusses the evidence of the roles of efflux pumps in biofilm formation; the potential approaches towards overcoming the increasing problem of biofilm-based infections are also discussed.
Collapse
Affiliation(s)
- Manaf AlMatar
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), 26300, Gambang, Kuantan, Malaysia.
| | - Osman Albarri
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitüsü), Çukurova University, Adana, Turkey
| | - Essam A Makky
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), 26300, Gambang, Kuantan, Malaysia.
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
14
|
Ramirez D, Berry L, Domalaon R, Brizuela M, Schweizer F. Dilipid Ultrashort Tetrabasic Peptidomimetics Potentiate Novobiocin and Rifampicin Against Multidrug-Resistant Gram-Negative Bacteria. ACS Infect Dis 2020; 6:1413-1426. [PMID: 32357292 DOI: 10.1021/acsinfecdis.0c00017] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of new antibacterial agents and therapeutic approaches is of high importance to address the global problem of antibiotic resistance. Although antimicrobial peptides are known to synergize with certain antibiotics, their clinical application is limited by their systemic toxicity, protease instability, and high production cost. To overcome these problems, nine dilipid ultrashort tetrabasic peptidomimetics (dUSTBPs) were prepared consisting of three basic amino acids separated by a molecular scaffold, bis(3-aminopropyl)glycine, and were ligated to two fatty acids. Several nonhemolytic dUSTBPs were shown to enhance the activity of several antibiotics against pathogenic Gram-negative bacteria. More importantly, dUSTBP 8, consisting of three l-arginine units and a dilipid of 8 carbons long, potentiated novobiocin and rifampicin consistently against multidrug-resistant (MDR) clinical isolates of Pseudomonas aeruginosa, Acinetobacter baumannii, and Enterobacteriaceae. Preliminary studies suggested that dUSTBPs were likely to potentiate antibiotics through outer membrane permeabilization and/or disruption of active efflux and that dUSTBP 8 exhibited enhanced resistance to trypsin in comparison to the previously described di-C9-KKKK-NH2 antibiotic potentiator. The antibacterial activity of rifampicin and novobiocin was enhanced by dUSTBP 8 comparable to other known outer membrane permeabilizing potentiators including the gold standard polymyxin B nonapeptide. Our results indicate that ultrashort tetrabasic peptidomimetics are potent adjuvants that repurpose novobiocin and rifampicin as potent agents against priority MDR Gram-negative pathogens.
Collapse
Affiliation(s)
- Danyel Ramirez
- Department of Chemistry, Faculty of Science, University of Manitoba, 144 Dysart Road, Winnipeg, Manitoba R3T 2N2, Canada
| | - Liam Berry
- Department of Chemistry, Faculty of Science, University of Manitoba, 144 Dysart Road, Winnipeg, Manitoba R3T 2N2, Canada
| | - Ronald Domalaon
- Department of Chemistry, Faculty of Science, University of Manitoba, 144 Dysart Road, Winnipeg, Manitoba R3T 2N2, Canada
| | - Marc Brizuela
- Department of Chemistry, Faculty of Science, University of Manitoba, 144 Dysart Road, Winnipeg, Manitoba R3T 2N2, Canada
| | - Frank Schweizer
- Department of Chemistry, Faculty of Science, University of Manitoba, 144 Dysart Road, Winnipeg, Manitoba R3T 2N2, Canada
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, 727 McDermot Avenue, Winnipeg, Manitoba R3T 1R9, Canada
| |
Collapse
|
15
|
Amphiphilic nebramine-based hybrids Rescue legacy antibiotics from intrinsic resistance in multidrug-resistant Gram-negative bacilli. Eur J Med Chem 2019; 175:187-200. [DOI: 10.1016/j.ejmech.2019.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/24/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
|
16
|
Baker KR, Jana B, Hansen AM, Nielsen HM, Franzyk H, Guardabassi L. Repurposing Azithromycin and Rifampicin Against Gram-Negative Pathogens by Combination With Peptidomimetics. Front Cell Infect Microbiol 2019; 9:236. [PMID: 31334131 PMCID: PMC6615261 DOI: 10.3389/fcimb.2019.00236] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022] Open
Abstract
Synthetic peptidomimetics may be designed to mimic functions of antimicrobial peptides, including potentiation of antibiotics, yet possessing improved pharmacological properties. Pairwise screening of 42 synthetic peptidomimetics combined with the antibiotics azithromycin and rifampicin in multidrug-resistant (MDR) Escherichia coli ST131 and Klebsiella pneumoniae ST258 led to identification of two subclasses of α-peptide/β-peptoid hybrids that display synergy with azithromycin and rifampicin (fractional inhibitory concentration indexes of 0.03–0.38). Further screening of the best three peptidomimetics in combination with a panel of 21 additional antibiotics led to identification of peptidomimetics that potentiated ticarcillin/clavulanate and erythromycin against E. coli, and clindamycin against K. pneumoniae. The study of six peptidomimetics was extended to Pseudomonas aeruginosa, confirming synergy with antibiotics for five of them. The most promising compound, H-(Lys-βNPhe)8-NH2, exerted only a minor effect on the viability of mammalian cells (EC50 ≥ 124–210 μM), and thus exhibited the highest selectivity toward bacteria. This compound also synergized with rifampicin and azithromycin at sub-micromolar concentrations (0.25–0.5 μM), thereby inducing susceptibility to these antibiotics at clinically relevant concentrations in clinical MDR isolates. This peptidomimetic lead and its analogs constitute promising candidates for efficient repurposing of rifampicin and azithromycin against Gram-negative pathogens.
Collapse
Affiliation(s)
- Kristin R Baker
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Bimal Jana
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Anna Mette Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Luca Guardabassi
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, United Kingdom
| |
Collapse
|
17
|
Berry L, Domalaon R, Brizuela M, Zhanel GG, Schweizer F. Polybasic peptide-levofloxacin conjugates potentiate fluoroquinolones and other classes of antibiotics against multidrug-resistant Gram-negative bacteria. MEDCHEMCOMM 2019; 10:517-527. [PMID: 31057731 PMCID: PMC6482413 DOI: 10.1039/c9md00051h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/04/2019] [Indexed: 01/30/2023]
Abstract
To address the rising threat of multidrug-resistant (MDR) bacteria, new therapeutic strategies must be developed. Efficacious drug combinations consisting of existing antibiotics and enhancer biomolecules called adjuvants offers a viable strategy. We have previously reported antibiotic hybrids consisting of tobramycin appended to different fluoroquinolones that possess potential as stand-alone antimicrobials as well as adjuvants. Herein, we report the synthesis of polybasic peptide-levofloxacin conjugates based on these tobramycin-fluoroquinolone hybrids. It was found that conjugating polybasic peptides to the fluoroquinolone levofloxacin, along with the addition of an aliphatic hydrocarbon tether, resulted in the ability of these compounds to potentiate fluoroquinolones and other antibiotics against MDR Gram-negative bacteria. The conjugates were able to potentiate ciprofloxacin, levofloxacin and moxifloxacin against MDR clinical isolates of Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae and to a lesser extent, Acinetobacter baumannii. Preliminary data revealed that the conjugates interfered with active efflux of fluoroquinolones in P. aeruginosa. In addition, synergy was observed with a wide array of other antibiotics against P. aeruginosa, including those that suffered from restricted outer membrane penetration, suggesting that in addition to blocking active efflux, the polybasic peptide-levofloxacin conjugates possessed the ability to disrupt and permeabilize the outer membrane of Gram-negative bacteria.
Collapse
Affiliation(s)
- Liam Berry
- Department of Chemistry , University of Manitoba , Winnipeg , MB , Canada .
| | - Ronald Domalaon
- Department of Chemistry , University of Manitoba , Winnipeg , MB , Canada .
| | - Marc Brizuela
- Department of Chemistry , University of Manitoba , Winnipeg , MB , Canada .
| | - George G Zhanel
- Department of Medical Microbiology and Infectious Diseases , University of Manitoba , Winnipeg , MB , Canada
| | - Frank Schweizer
- Department of Chemistry , University of Manitoba , Winnipeg , MB , Canada .
- Department of Medical Microbiology and Infectious Diseases , University of Manitoba , Winnipeg , MB , Canada
| |
Collapse
|
18
|
Dimopoulos G, Koulenti D, Parker SL, Roberts JA, Arvaniti K, Poulakou G. Intravenous fosfomycin for the treatment of multidrug-resistant pathogens: what is the evidence on dosing regimens? Expert Rev Anti Infect Ther 2019; 17:201-210. [PMID: 30668931 DOI: 10.1080/14787210.2019.1573669] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The intravenous (IV) formulation of fosfomycin has been re-introduced in clinical practice mainly to overcome treatment failures against multidrug-resistant (MDR) bacteria. Appropriate dosing schedules of the IV formulation have not yet been established. Areas covered: The mechanism of action and resistance development, commercial IV formulations, pharmacokinetic/pharmacodynamic (PK/PD) properties, IV dosing regimens for the treatment of MDR infections along with efficacy and safety issues were reviewed. Data regarding specific MDR pathogens, daily doses and patients' outcomes, gaps in the current literature, and in progress research agenda are presented. Expert opinion: The doses of fosfomycin IV range between 12 and 24 grams/day depending on the severity of infection. The efficacy and safety of the commonly administered doses have been shown mainly in observational non-comparative trials. The optimal dose ensuring maximal efficacy with minimal toxicity along with the most appropriate co-administered antibiotic(s) need further evaluation. The pharmacokinetic/pharmacodynamic parameter associated with maximum efficacy has not yet been established, although, the ratio of the area under the concentration-time curve (AUC) for the free unbound fraction of fosfomycin versus the MIC (fAUC/MIC) may be linked to optimal treatment. RCTs and other comparative studies are underway to address gaps of knowledge in adult patients and neonates.
Collapse
Affiliation(s)
- George Dimopoulos
- a Department of Critical Care , University Hospital ATTIKON, National and Kapodistrian University of Athens , Athens , Greece
| | - Despoina Koulenti
- a Department of Critical Care , University Hospital ATTIKON, National and Kapodistrian University of Athens , Athens , Greece.,b UQ Centre for Clinical Research, Faculty of Medicine , The University of Queensland , Brisbane , Australia
| | - Suzanne L Parker
- b UQ Centre for Clinical Research, Faculty of Medicine , The University of Queensland , Brisbane , Australia
| | - Jason A Roberts
- b UQ Centre for Clinical Research, Faculty of Medicine , The University of Queensland , Brisbane , Australia.,c School of Pharmacy, Centre for Translational Anti-infective Pharmacodynamics , The University of Queensland , Brisbane , Australia.,d Department of Intensive Care Medicine , Royal Brisbane and Women's Hospital , Brisbane , Australia.,e Pharmacy Department , Royal Brisbane and Women's Hospital , Brisbane , Australia
| | - Kostoula Arvaniti
- f Intensive Care Unit , Papageorgiou University Affiliated Hospital , Thessaloniki , Greece
| | - Garyphalia Poulakou
- g 3rd Department of Internal Medicine, SOTIRIA Hospital , National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
19
|
Blanco P, Sanz-García F, Hernando-Amado S, Martínez JL, Alcalde-Rico M. The development of efflux pump inhibitors to treat Gram-negative infections. Expert Opin Drug Discov 2018; 13:919-931. [PMID: 30198793 DOI: 10.1080/17460441.2018.1514386] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION One of the possibilities for reducing the emergence and spread of antibiotic resistance is the use of anti-resistance compounds capable of resensitizing resistant microorganisms to current antimicrobials. For this purpose, multidrug efflux pumps, whose inhibition may increase bacterial susceptibility to several antibiotics, including macrolides to which Gram-negatives are considered intrinsically resistant, have emerged as suitable targets. Areas covered: In the current review, the authors discuss different mechanisms that can be exploited for inhibiting multidrug efflux pumps and describe the properties and the potential therapeutic value of already studied efflux pumps inhibitors. Although efforts have already been made to develop these inhibitors, there are currently no good candidates for treating infectious diseases. Consequently, the authors also discuss potential approaches for their development. Expert opinion: Classical anti-resistance drugs such as beta-lactamases inhibitors, while useful, are only purposeful for treating infections caused by beta-lactamase producers. However, inhibitors of multidrug efflux pumps, which are present on all organisms, can sensitize both susceptible and resistant bacteria to antibiotics belonging to several different structural families. Since some efflux pumps are involved in bacterial infections, their inhibition may also reduce the infectivity of Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Paula Blanco
- a Department of Microbial Biotechnology , Centro Nacional de Biotecnología. CSIC , Madrid , Spain
| | - Fernando Sanz-García
- a Department of Microbial Biotechnology , Centro Nacional de Biotecnología. CSIC , Madrid , Spain
| | - Sara Hernando-Amado
- a Department of Microbial Biotechnology , Centro Nacional de Biotecnología. CSIC , Madrid , Spain
| | - José Luis Martínez
- a Department of Microbial Biotechnology , Centro Nacional de Biotecnología. CSIC , Madrid , Spain
| | - Manuel Alcalde-Rico
- a Department of Microbial Biotechnology , Centro Nacional de Biotecnología. CSIC , Madrid , Spain
| |
Collapse
|