1
|
Vishwakarma K, Bisht A, Kumar P, Kumar S, Akhter J, Payasi A, Chaudhary S, Aggarwal A. Toxicokinetic profiling of VRP-034: Evaluating its potential in mitigating polymyxin-B-associated nephrotoxicity. Int J Antimicrob Agents 2025; 65:107393. [PMID: 39612992 DOI: 10.1016/j.ijantimicag.2024.107393] [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: 07/10/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
This study assessed the nephrotoxicity and toxicokinetic profile of VRP-034 [a novel formulation of polymyxin B (PMB)] compared with marketed PMB over a 7-day repeat-dose regimen. Three objectives were pursued: evaluating PMB pharmacokinetics in both groups, alongside assessing the impact of VRP-034 on mitigating PMB-associated kidney injury; analysing the reversibility of kidney injury; and validating novel kidney injury biomarkers against traditional markers using histopathological scoring. Sixty-eight Sprague-Dawley rats were divided into three groups: 30 in each of the marketed PMB and VRP-034 groups, and eight in the control group. Rats received drugs at 6 mg/kg subcutaneously every 8 h (human equivalent dose ∼3 mg/kg/day). Toxicokinetic evaluations were conducted on selected animals on days 1, 2, 4, and 7 (after 3rd, 6th, 12th and 21st dose), while the remaining animals were observed for an additional 7 days without treatment. Samples were collected up to 12 h post-administration, followed by necropsy and histopathological examination. Plasma PMB concentrations were quantified; and kidney injury biomarkers, oxidative stress and anti-inflammatory markers were evaluated. Receiver operating characteristic curve analysis was performed to validate kidney injury biomarkers against histopathological grading. Similar plasma PMB concentrations and pharmacokinetic parameters were found in the two treatment groups. However, the VRP-034 group exhibited significantly lower nephrotoxicity, with reduced levels of kidney injury biomarkers, and diminished oxidative stress and inflammation levels compared with the marketed PMB group. Histopathological examination confirmed reduced renal damage in the VRP-034 group. Novel kidney injury biomarkers demonstrated superior sensitivity, specificity and early detection capability over traditional markers. In conclusion, VRP-034 demonstrated reduced nephrotoxicity compared with marketed PMB, suggesting its potential as a safer alternative.
Collapse
Affiliation(s)
| | - Anmol Bisht
- Venus Medicine Research Centre, Bhatoli Kalan, Baddi, India
| | - Parveen Kumar
- Venus Medicine Research Centre, Bhatoli Kalan, Baddi, India
| | - Satish Kumar
- Venus Medicine Research Centre, Bhatoli Kalan, Baddi, India
| | - Jawed Akhter
- Venus Medicine Research Centre, Bhatoli Kalan, Baddi, India
| | - Anurag Payasi
- Venus Medicine Research Centre, Bhatoli Kalan, Baddi, India
| | | | - Anmol Aggarwal
- Venus Medicine Research Centre, Bhatoli Kalan, Baddi, India.
| |
Collapse
|
2
|
Payasi A, Yadav MK, Chaudhary S, Aggarwal A. Evaluating nephrotoxicity reduction in a novel polymyxin B formulation: insights from a 3D kidney-on-a-chip model. Antimicrob Agents Chemother 2024; 68:e0021924. [PMID: 39225483 PMCID: PMC11459911 DOI: 10.1128/aac.00219-24] [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] [Received: 02/09/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to assess the nephrotoxicity associated with VRP-034 (novel formulation of polymyxin B [PMB]) compared to marketed PMB in a three-dimensional (3D) kidney-on-a-chip model. To model the human kidney proximal tubule for analysis, tubular structures were established using 23 triple-channel chips seeded with RPTEC/hTERT1 cells. These cells were exposed to VRP-034 or PMB at seven concentrations (1-200 µM) over 12, 24, and 48 h. A suite of novel kidney injury biomarkers, cell health, and inflammatory markers were quantitatively assessed in the effluent. Additionally, caspase and cytochrome C levels were measured, and cell viability was evaluated using calcein AM and ethidium homodimer-1 (EthD-1). Exposure to marketed PMB resulted in significantly elevated levels (P < 0.05) of four key biomarkers (KIM-1, cystatin C, clusterin, and OPN) compared to VRP-034, particularly at clinically relevant concentrations of ≥10 µM. At 25 µM, all biomarkers demonstrated a significant increase (P < 0.05) with marketed PMB exposure compared to VRP-034. Inflammatory markers (interleukin-6 and interleukin-8) increased significantly (P < 0.05) with marketed PMB at concentrations of ≥5 µM, relative to VRP-034. VRP-034 displayed superior cell health outcomes, exhibiting lower lactate dehydrogenase release, while ATP levels remained comparable. Morphological analysis revealed that marketed PMB induced more severe damage, disrupting tubular integrity. Both treatments activated cytochrome C, caspase-3, caspase-8, caspase-9, and caspase-12 in a concentration-dependent manner; however, caspase activation was significantly reduced (P < 0.05) with VRP-034. This study demonstrates that VRP-034 significantly reduces nephrotoxicity compared to marketed PMB within a 3D microphysiological system, suggesting its potential to enable the use of full therapeutic doses of PMB with an improved safety profile, addressing the need for less nephrotoxic polymyxin antibiotics.
Collapse
Affiliation(s)
- Anurag Payasi
- Department of Cell Culture, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| | - Manoj Kumar Yadav
- Department of Cell Culture, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| | | | - Anmol Aggarwal
- Department of Pipeline Strategy, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| |
Collapse
|
3
|
Song MH, Xiang BX, Yang CY, Lee CH, Yan YX, Yang QJ, Yin WJ, Zhou Y, Zuo XC, Xie YL. A pilot clinical risk model to predict polymyxin-induced nephrotoxicity: a real-world, retrospective cohort study. J Antimicrob Chemother 2024; 79:1919-1928. [PMID: 38946304 DOI: 10.1093/jac/dkae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024] Open
Abstract
OBJECTIVES Polymyxin-induced nephrotoxicity (PIN) is a major safety concern and challenge in clinical practice, which limits the clinical use of polymyxins. This study aims to investigate the risk factors and to develop a scoring tool for the early prediction of PIN. METHODS Data on critically ill patients who received intravenous polymyxin B or colistin sulfate for over 24 h were collected. Logistic regression with the least absolute shrinkage and selection operator (LASSO) was used to identify variables that are associated with outcomes. The eXtreme Gradient Boosting (XGB) classifier algorithm was used to further visualize factors with significant differences. A prediction model for PIN was developed through binary logistic regression analysis and the model was assessed by temporal validation and external validation. Finally, a risk-scoring system was developed based on the prediction model. RESULTS Of 508 patients, 161 (31.6%) patients developed PIN. Polymyxin type, loading dose, septic shock, concomitant vasopressors and baseline blood urea nitrogen (BUN) level were identified as significant predictors of PIN. All validation exhibited great discrimination, with the AUC of 0.742 (95% CI: 0.696-0.787) for internal validation, of 0.708 (95% CI: 0.605-0.810) for temporal validation and of 0.874 (95% CI: 0.759-0.989) for external validation, respectively. A simple risk-scoring tool was developed with a total risk score ranging from -3 to 4, corresponding to a risk of PIN from 0.79% to 81.24%. CONCLUSIONS This study established a prediction model for PIN. Before using polymyxins, the simple risk-scoring tool can effectively identify patients at risk of developing PIN within a range of 7% to 65%.
Collapse
Affiliation(s)
- Mong-Hsiu Song
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Bi-Xiao Xiang
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
- College of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Chien-Yi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Chou-Hsi Lee
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Yu-Xuan Yan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Qin-Jie Yang
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Wen-Jun Yin
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Yangang Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiao-Cong Zuo
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Yue-Liang Xie
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
4
|
Padhy I, Dwibedy SK, Mohapatra SS. A molecular overview of the polymyxin-LPS interaction in the context of its mode of action and resistance development. Microbiol Res 2024; 283:127679. [PMID: 38508087 DOI: 10.1016/j.micres.2024.127679] [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: 07/31/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
With the rising incidences of antimicrobial resistance (AMR) and the diminishing options of novel antimicrobial agents, it is paramount to decipher the molecular mechanisms of action and the emergence of resistance to the existing drugs. Polymyxin, a cationic antimicrobial lipopeptide, is used to treat infections by Gram-negative bacterial pathogens as a last option. Though polymyxins were identified almost seventy years back, their use has been restricted owing to toxicity issues in humans. However, their clinical use has been increasing in recent times resulting in the rise of polymyxin resistance. Moreover, the detection of "mobile colistin resistance (mcr)" genes in the environment and their spread across the globe have complicated the scenario. The mechanism of polymyxin action and the development of resistance is not thoroughly understood. Specifically, the polymyxin-bacterial lipopolysaccharide (LPS) interaction is a challenging area of investigation. The use of advanced biophysical techniques and improvement in molecular dynamics simulation approaches have furthered our understanding of this interaction, which will help develop polymyxin analogs with better bactericidal effects and lesser toxicity in the future. In this review, we have delved deeper into the mechanisms of polymyxin-LPS interactions, highlighting several models proposed, and the mechanisms of polymyxin resistance development in some of the most critical Gram-negative pathogens.
Collapse
Affiliation(s)
- Indira Padhy
- Molecular Microbiology Lab, Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur 760007, Odisha, India
| | - Sambit K Dwibedy
- Molecular Microbiology Lab, Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur 760007, Odisha, India
| | - Saswat S Mohapatra
- Molecular Microbiology Lab, Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur 760007, Odisha, India.
| |
Collapse
|
5
|
Varache M, Rizzo S, Sayers EJ, Newbury L, Mason A, Liao CT, Chiron E, Bourdiec N, Jones A, Fraser DJ, Taylor PR, Jones AT, Thomas DW, Ferguson EL. Dextrin conjugation to colistin inhibits its toxicity, cellular uptake and acute kidney injury in vivo. RSC PHARMACEUTICS 2024; 1:68-79. [PMID: 38646595 PMCID: PMC11024668 DOI: 10.1039/d3pm00014a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/04/2024] [Indexed: 04/23/2024]
Abstract
The acute kidney injury (AKI) and dose-limiting nephrotoxicity, which occurs in 20-60% of patients following systemic administration of colistin, represents a challenge in the effective treatment of multi-drug resistant Gram-negative infections. To reduce clinical toxicity of colistin and improve targeting to infected/inflamed tissues, we previously developed dextrin-colistin conjugates, whereby colistin is designed to be released by amylase-triggered degradation of dextrin in infected and inflamed tissues, after passive targeting by the enhanced permeability and retention effect. Whilst it was evident in vitro that polymer conjugation can reduce toxicity and prolong plasma half-life, without significant reduction in antimicrobial activity of colistin, it was unclear how dextrin conjugation would alter cellular uptake and localisation of colistin in renal tubular cells in vivo. We discovered that dextrin conjugation effectively reduced colistin's toxicity towards human kidney proximal tubular epithelial cells (HK-2) in vitro, which was mirrored by significantly less cellular uptake of Oregon Green (OG)-labelled dextrin-colistin conjugate, when compared to colistin. Using live-cell confocal imaging, we revealed localisation of both, free and dextrin-bound colistin in endolysosome compartments of HK-2 and NRK-52E cells. Using a murine AKI model, we demonstrated dextrin-colistin conjugation dramatically diminishes both proximal tubular injury and renal accumulation of colistin. These findings reveal new insight into the mechanism by which dextrin conjugation can overcome colistin's renal toxicity and show the potential of polymer conjugation to improve the side effect profile of nephrotoxic drugs.
Collapse
Affiliation(s)
- Mathieu Varache
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Siân Rizzo
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Edward J Sayers
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University CF10 3NB UK
| | - Lucy Newbury
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University Cardiff CF14 4XN UK
| | - Anna Mason
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University Cardiff CF14 4XN UK
| | - Chia-Te Liao
- Systems Immunity Research Institute, Division of Infection and Immunity, School of Medicine, Cardiff University Cardiff CF14 4XN UK
| | - Emilie Chiron
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Nathan Bourdiec
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Adam Jones
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
- Cellular Pathology Department, University Dental Hospital, Cardiff and Vale University Health Board Cardiff CF14 4XY UK
| | - Donald J Fraser
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University Cardiff CF14 4XN UK
| | - Philip R Taylor
- Systems Immunity Research Institute, Division of Infection and Immunity, School of Medicine, Cardiff University Cardiff CF14 4XN UK
- UK Dementia Research Institute at Cardiff Hadyn Ellis Building Maindy Road Cardiff CF24 4HQ UK
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University CF10 3NB UK
| | - David W Thomas
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
- Systems Immunity Research Institute, Division of Infection and Immunity, School of Medicine, Cardiff University Cardiff CF14 4XN UK
| | - Elaine L Ferguson
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| |
Collapse
|
6
|
Slingerland C, Martin NI. Recent Advances in the Development of Polymyxin Antibiotics: 2010-2023. ACS Infect Dis 2024; 10:1056-1079. [PMID: 38470446 PMCID: PMC11019560 DOI: 10.1021/acsinfecdis.3c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 03/13/2024]
Abstract
The polymyxins are nonribosomal lipopeptides produced by Paenibacillus polymyxa and are potent antibiotics with activity specifically directed against Gram-negative bacteria. While the clinical use of polymyxins has historically been limited due to their toxicity, their use is on the rise given the lack of alternative treatment options for infections due to multidrug resistant Gram-negative pathogens. The Gram-negative specificity of the polymyxins is due to their ability to target lipid A, the membrane embedded LPS anchor that decorates the cell surface of Gram-negative bacteria. Notably, the mechanisms responsible for polymyxin toxicity, and in particular their nephrotoxicity, are only partially understood with most insights coming from studies carried out in the past decade. In parallel, many synthetic and semisynthetic polymyxin analogues have been developed in recent years in an attempt to mitigate the nephrotoxicity of the natural products. Despite these efforts, to date, no polymyxin analogues have gained clinical approval. This may soon change, however, as at the moment there are three novel polymyxin analogues in clinical trials. In this context, this review provides an update of the most recent insights with regard to the structure-activity relationships and nephrotoxicity of new polymyxin variants reported since 2010. We also discuss advances in the synthetic methods used to generate new polymyxin analogues, both via total synthesis and semisynthesis.
Collapse
Affiliation(s)
- Cornelis
J. Slingerland
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Nathaniel I. Martin
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| |
Collapse
|
7
|
Kim SJ, Jo J, Kim J, Ko KS, Lee W. Polymyxin B nonapeptide potentiates the eradication of Gram-negative bacterial persisters. Microbiol Spectr 2024; 12:e0368723. [PMID: 38391225 PMCID: PMC10986493 DOI: 10.1128/spectrum.03687-23] [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] [Received: 10/16/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Antibiotic-resistant Gram-negative bacteria remain a globally leading cause of bacterial infection-associated mortality, and it is imperative to identify novel therapeutic strategies. Recently, the advantage of using antibacterials selective against Gram-negative bacteria has been demonstrated with polymyxins that specifically target the lipopolysaccharides of Gram-negative bacteria. However, the severe cytotoxicity of polymyxins limits their clinical use. Here, we demonstrate that polymyxin B nonapeptide (PMBN), a polymyxin B derivative without the terminal amino acyl residue, can significantly enhance the effectiveness of commonly used antibiotics against only Gram-negative bacteria and their persister cells. We show that although PMBN itself does not exhibit antibacterial activity or cytotoxicity well above the 100-fold minimum inhibitory concentration of polymyxin B, PMBN can increase the potency of co-treated antibiotics. We also demonstrate that using PMBN in combination with other antibiotics significantly reduces the frequency of resistant mutant formation. Together, this work provides evidence of the utilities of PMBN as a novel potentiator for antibiotics against Gram-negative bacteria and insights for the eradication of bacterial persister cells during antibiotic treatment. IMPORTANCE The significance of our study lies in addressing the problem of antibiotic-resistant Gram-negative bacteria, which continue to be a global cause of mortality associated with bacterial infections. Therefore, identifying innovative therapeutic approaches is an urgent need. Recent research has highlighted the potential of selective antibacterials like polymyxins, which specifically target the lipopolysaccharides of Gram-negative bacteria. However, the clinical use of polymyxins is limited by their severe cytotoxicity. This study unveils the effectiveness of polymyxin B nonapeptide (PMBN) in significantly enhancing the eradication of persister cells in Gram-negative bacteria. Although PMBN itself does not exhibit antibacterial activity or cytotoxicity, it remarkably reduces persister cells during the treatment of antibiotics. Moreover, combining PMBN with other antibiotics reduces the emergence of resistant mutants. Our research emphasizes the utility of PMBN as a novel potentiator to decrease persister cells during antibiotic treatments for Gram-negative bacteria.
Collapse
Affiliation(s)
- Sun Ju Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeongwoo Jo
- Department of Microbiology, School of Medicine, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jihyeon Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwan Soo Ko
- Department of Microbiology, School of Medicine, Sungkyunkwan University, Suwon, Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
8
|
Wang X, Patil N, Li F, Wang Z, Zhan H, Schmidt D, Thompson P, Guo Y, Landersdorfer CB, Shen HH, Peleg AY, Li J, Song J. PmxPred: A data-driven approach for the identification of active polymyxin analogues against gram-negative bacteria. Comput Biol Med 2024; 168:107681. [PMID: 37992470 DOI: 10.1016/j.compbiomed.2023.107681] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/07/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023]
Abstract
The multidrug-resistant Gram-negative bacteria has evolved into a worldwide threat to human health; over recent decades, polymyxins have re-emerged in clinical practice due to their high activity against multidrug-resistant bacteria. Nevertheless, the nephrotoxicity and neurotoxicity of polymyxins seriously hinder their practical use in the clinic. Based on the quantitative structure-activity relationship (QSAR), analogue design is an efficient strategy for discovering biologically active compounds with fewer adverse effects. To accelerate the polymyxin analogues discovery process and find the polymyxin analogues with high antimicrobial activity against Gram-negative bacteria, here we developed PmxPred, a GCN and catBoost-based machine learning framework. The RDKit descriptors were used for the molecule and residues representation, and the ensemble learning model was utilized for the antimicrobial activity prediction. This framework was trained and evaluated on multiple Gram-negative bacteria datasets, including Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and a general Gram-negative bacteria dataset achieving an AUROC of 0.857, 0.880, 0.756, 0.895 and 0.865 on the independent test, respectively. PmxPred outperformed the transfer learning method that trained on 10 million molecules. We interpreted our model well-trained model by analysing the importance of global and residue features. Overall, PmxPred provides a powerful additional tool for predicting active polymyxin analogues, and holds the potential elucidate the mechanisms underlying the antimicrobial activity of polymyxins. The source code is publicly available on GitHub (https://github.com/yanwu20/PmxPred).
Collapse
Affiliation(s)
- Xiaoyu Wang
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia; Monash Data Futures Institute, Monash University, Melbourne, VIC, 3800, Australia; Centre to Impact AMR, Monash University, Melbourne, VIC, 3800, Australia
| | - Nitin Patil
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Fuyi Li
- College of Information Engineering, Northwest A&F University, Yangling, 712100, Shaanxi, China; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Zhikang Wang
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia; Monash Data Futures Institute, Monash University, Melbourne, VIC, 3800, Australia; Centre to Impact AMR, Monash University, Melbourne, VIC, 3800, Australia
| | - Haolan Zhan
- Faculty of Information Technology, Monash University, Melbourne, VIC, 3800, Australia
| | - Daniel Schmidt
- Monash Data Futures Institute, Monash University, Melbourne, VIC, 3800, Australia; Faculty of Information Technology, Monash University, Melbourne, VIC, 3800, Australia
| | - Philip Thompson
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Yuming Guo
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Cornelia B Landersdorfer
- Centre to Impact AMR, Monash University, Melbourne, VIC, 3800, Australia; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Hsin-Hui Shen
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia; Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Anton Y Peleg
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia; Centre to Impact AMR, Monash University, Melbourne, VIC, 3800, Australia; Department of Infectious Diseases, Alfred Hospital, Alfred Health, Melbourne, Victoria, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia; Monash Data Futures Institute, Monash University, Melbourne, VIC, 3800, Australia; Centre to Impact AMR, Monash University, Melbourne, VIC, 3800, Australia.
| | - Jiangning Song
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia; Monash Data Futures Institute, Monash University, Melbourne, VIC, 3800, Australia; Centre to Impact AMR, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
9
|
Slingerland CJ, Lysenko V, Chaudhuri S, Wesseling CMJ, Barnes D, Masereeuw R, Martin NI. Semisynthetic polymyxins with potent antibacterial activity and reduced kidney cell toxicity. RSC Med Chem 2023; 14:2417-2425. [PMID: 37974968 PMCID: PMC10650952 DOI: 10.1039/d3md00456b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/06/2023] [Indexed: 11/19/2023] Open
Abstract
The growing incidence of infections caused by multi-drug resistant Gram-negative bacteria has led to an increased use of last-resort antibiotics such as the polymyxins. Polymyxin therapy is limited by toxicity concerns, most notably nephrotoxicity. Recently we reported the development of a novel class of semisynthetic polymyxins with reduced toxicity wherein the N-terminal lipid and diaminobutyric acid residue are replaced by a cysteine-linked lipid featuring a reductively labile disulfide bond. In the present study we further explored the potential of this approach by also varying the amino acid residue directly adjacent to the polymyxin macrocycle. This led to the identification of new semisynthetic polymyxins that maintain the potent antibacterial activity of the clinically used polymyxin B while exhibiting a further reduction in toxicity toward human proximal tubule epithelial cells. Furthermore, these new polymyxins were found to effectively synergize with novobiocin, rifampicin, and erythromycin against mcr-positive, polymyxin resistant E. coli.
Collapse
Affiliation(s)
- Cornelis J Slingerland
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Vladyslav Lysenko
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Samhita Chaudhuri
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Charlotte M J Wesseling
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| | - Devon Barnes
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht University 3584 CG Utrecht The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht University 3584 CG Utrecht The Netherlands
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University Sylviusweg 72 2333 BE Leiden The Netherlands
| |
Collapse
|
10
|
Campbell RE, Chen CH, Edelstein CL. Overview of Antibiotic-Induced Nephrotoxicity. Kidney Int Rep 2023; 8:2211-2225. [PMID: 38025228 PMCID: PMC10658282 DOI: 10.1016/j.ekir.2023.08.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 12/01/2023] Open
Abstract
Drug-induced nephrotoxicity accounts for up to 60% of cases of acute kidney injury (AKI) in hospitalized patients and is associated with increased morbidity and mortality in both adults and children. Antibiotics are one of the most common causes of drug-induced nephrotoxicity. Mechanisms of antibiotic-induced nephrotoxicity include glomerular injury, tubular injury or dysfunction, distal tubular obstruction from casts, and acute interstitial nephritis (AIN) mediated by a type IV (delayed-type) hypersensitivity response. Clinical manifestations of antibiotic-induced nephrotoxicity include acute tubular necrosis (ATN), AIN, and Fanconi syndrome. Given the potential nephrotoxic effects of antibiotics on critically ill patients, the use of novel biomarkers can provide information to optimize dosing and duration of treatment and can help prevent nephrotoxicity when traditional markers, such as creatinine, are unreliable. Use of novel kidney specific biomarkers, such as cystatin C and urinary kidney injury molecule-1 (KIM-1), may result in earlier detection of AKI, dose adjustment, or discontinuation of antibiotic and development of nonnephrotoxic antibiotics.
Collapse
Affiliation(s)
- Ruth E. Campbell
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chang Huei Chen
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Charles L. Edelstein
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
11
|
Bintang MAKM, Nopparat J, Srichana T. In vivo evaluation of nephrotoxicity and neurotoxicity of colistin formulated with sodium deoxycholate sulfate in a mice model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3243-3252. [PMID: 37249614 DOI: 10.1007/s00210-023-02531-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023]
Abstract
Neurotoxicity and nephrotoxicity are the major dose-limiting factors for the clinical use of colistin against multidrug-resistant (MDR) Gram-negative bacteria. This study aimed to investigate the neurotoxic and nephrotoxic effects of colistin formulated with in-house synthesized sodium deoxycholate sulfate (SDCS) in a mouse model. Male mice C57BL/6 were randomly divided into four groups: control (saline solution), colistin (15 mg/kg/day), colistin:SDCS 1:1, and colistin:SDCS 1:2. In the colistin:SDCS treatment groups, the dosage was 15 mg/kg/day colistin equivalent; all mice were treated for 7 successive days. The thermal tolerance, body weight gain and organ weights were measured. The levels of serum blood urea nitrogen (BUN), creatinine (Cr), superoxide dismutase (SOD), and catalase (CAT) were assessed. Histopathological damages were assessed on mice organ. The colistin:SDCS formulations significantly improved thermal pain response of the mice comparable to the control group. The administration did not impair kidney function as evidence from BUN and Cr results; however, the oxidative stress biomarkers decreased in the colistin and colistin-SDCS treated mice. Several abnormalities were observed in the kidney, liver, spleen, and sciatic nerve tissues following colistin treatment, which indicated evidence of toxicity. The colistin-SDCS formulations were associated with less acute toxicity and fewer nephrotoxic and neurotoxic changes compared with the colistin alone group which indicated that SDCS attenuated colistin nephrotoxicity and neurotoxicity. This study highlights the potential application of colistin formulated with SDCS for safer clinical use against MDR Gram-negative bacteria.
Collapse
Affiliation(s)
- Muhammad Ali Khumaini Mudhar Bintang
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
| | - Jongdee Nopparat
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Teerapol Srichana
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand.
| |
Collapse
|
12
|
Slingerland C, Wesseling CMJ, Innocenti P, Westphal KGC, Masereeuw R, Martin NI. Synthesis and Evaluation of Polymyxins Bearing Reductively Labile Disulfide-Linked Lipids. J Med Chem 2022; 65:15878-15892. [PMID: 36399613 PMCID: PMC9743094 DOI: 10.1021/acs.jmedchem.2c01528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Polymyxins are a class of lipopeptide anti-infective agents with potent and specific activity against Gram-negative bacteria. While toxicity concerns associated with polymyxin B and E (colistin) have historically limited their clinical application, today they are increasingly used as last-resort antibiotics given the rise of multidrug-resistant Gram-negative pathogens. The adverse side effects of polymyxins are well known, particularly as related to their nephrotoxicity. Here, we describe the synthesis and evaluation of a novel series of polymyxin analogues, aimed at reducing their nephrotoxic effects. Using a semisynthetic approach, we explored modifications of the exocyclic part of the polymyxin scaffold, namely, the terminal amino acid and lipophilic tail. By incorporating a reductively labile disulfide linkage in the lipid tail, we obtained novel polymyxins that exhibit potent antibacterial activity on par with polymyxin B but with reduced toxicity toward human renal proximal tubular epithelial cells.
Collapse
Affiliation(s)
- Cornelis
J. Slingerland
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Charlotte M. J. Wesseling
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Paolo Innocenti
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Koen G. C. Westphal
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Nathaniel I. Martin
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands,
| |
Collapse
|
13
|
Mally A, Jarzina S. Mapping Adverse Outcome Pathways for Kidney Injury as a Basis for the Development of Mechanism-Based Animal-Sparing Approaches to Assessment of Nephrotoxicity. FRONTIERS IN TOXICOLOGY 2022; 4:863643. [PMID: 35785263 PMCID: PMC9242087 DOI: 10.3389/ftox.2022.863643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023] Open
Abstract
In line with recent OECD activities on the use of AOPs in developing Integrated Approaches to Testing and Assessment (IATAs), it is expected that systematic mapping of AOPs leading to systemic toxicity may provide a mechanistic framework for the development and implementation of mechanism-based in vitro endpoints. These may form part of an integrated testing strategy to reduce the need for repeated dose toxicity studies. Focusing on kidney and in particular the proximal tubule epithelium as a key target site of chemical-induced injury, the overall aim of this work is to contribute to building a network of AOPs leading to nephrotoxicity. Current mechanistic understanding of kidney injury initiated by 1) inhibition of mitochondrial DNA polymerase γ (mtDNA Polγ), 2) receptor mediated endocytosis and lysosomal overload, and 3) covalent protein binding, which all present fairly well established, common mechanisms by which certain chemicals or drugs may cause nephrotoxicity, is presented and systematically captured in a formal description of AOPs in line with the OECD AOP development programme and in accordance with the harmonized terminology provided by the Collaborative Adverse Outcome Pathway Wiki. The relative level of confidence in the established AOPs is assessed based on evolved Bradford-Hill weight of evidence considerations of biological plausibility, essentiality and empirical support (temporal and dose-response concordance).
Collapse
|
14
|
Wu XL, Long WM, Lu Q, Teng XQ, Qi TT, Qu Q, He GF, Qu J. Polymyxin B-Associated Nephrotoxicity and Its Predictors: A Retrospective Study in Carbapenem-Resistant Gram-Negative Bacterial Infections. Front Pharmacol 2022; 13:672543. [PMID: 35571125 PMCID: PMC9096016 DOI: 10.3389/fphar.2022.672543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Polymyxin B (PMB), a kind of polymyxin, was widely used in carbapenem-resistant Gram-negative bacterial (CR-GNB) infections. However, adverse reactions such as nephrotoxicity and neurotoxicity limit its use in clinical practice. The aim of this study was to explore PMB associated with nephrotoxicity and its predictors. Patients who received PMB intravenous drip for more than 72 h were eligible for the study. Characteristics of patients, concomitant nephrotoxic agents, underlying disease, and antimicrobial susceptibility were submitted for descriptive analysis. Univariate analysis and binary logistic regression were used to assess the factors leading to acute kidney injury (AKI). AKI was assessed with serum creatinine variations according to the classification of risk (stage R), injury (stage I), failure (stage F), loss, and end-stage of kidney disease. Among 234 patients with CR-GNB infections who used PMB in our study, 67 (28.63%) patients developed AKI, including 31 (14.25%) patients in stage R, 15 (6.41%) patients in stage I, and 21 (8.97%) patients in stage F. The incident rate of PMB-related nephrotoxicity in patients with normal renal function was 32.82% (43/131). The higher risk factors of AKI include males [odds ratio (OR) = 3.237; 95% confidence interval (95%CI) = 1.426–7.350], digestive system diseases [OR = 2.481 (1.127–5.463)], using furosemide (>20 mg/day) [OR = 2.473 (1.102–5.551)], and baseline serum creatinine [OR = 0.994 (0.990–0.999)]. Nonparametric tests of K-independent samples showed that baseline serum creatinine and the PMB maintenance dose were associated with the severity of nephrotoxicity (both p < 0.05). Male, digestive system diseases, using furosemide (>20 mg/day), and high baseline serum creatinine were the independent risk factors of PMB-associated AKI development. The maintenance dose of PMB may be related to the severity of AKI. These risk factors should be taken into consideration when initiating PMB-based therapy. The serum creatinine value should be closely monitored when using PMB.
Collapse
Affiliation(s)
- Xiao-Li Wu
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wen-Ming Long
- Department of Pharmacy, Second People’s Hospital of Huaihua City, Huaihua, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xin-Qi Teng
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Ting-Ting Qi
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Ge-Fei He
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
- *Correspondence: Jian Qu,
| |
Collapse
|
15
|
van Groesen E, Slingerland CJ, Innocenti P, Mihajlovic M, Masereeuw R, Martin NI. Vancomyxins: Vancomycin-Polymyxin Nonapeptide Conjugates That Retain Anti-Gram-Positive Activity with Enhanced Potency against Gram-Negative Strains. ACS Infect Dis 2021; 7:2746-2754. [PMID: 34387988 PMCID: PMC8438664 DOI: 10.1021/acsinfecdis.1c00318] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
![]()
Vancomycin functions
by binding to lipid II, the penultimate bacterial
cell wall building block used by both Gram-positive and Gram-negative
species. However, vancomycin is generally only able to exert its antimicrobial
effect against Gram-positive strains as it cannot pass the outer membrane
(OM) of Gram-negative bacteria. To address this challenge, we here
describe efforts to conjugate vancomycin to the OM disrupting polymyxin
E nonapeptide (PMEN) to yield the hybrid “vancomyxins”.
In designing these hybrid antibiotics, different spacers and conjugation
sites were explored for connecting vancomycin and PMEN. The vancomyxins
show improved activity against Gram-negative strains compared with
the activity of vancomycin or vancomycin supplemented with PMEN separately.
In addition, the vancomyxins maintain the antimicrobial effect of
vancomycin against Gram-positive strains and, in some cases, show
enhanced activity against vancomycin-resistant strains. The hybrid
antibiotics described here have reduced nephrotoxicity when compared
with clinically used polymyxin antibiotics. This study demonstrates
that covalent conjugation to an OM disruptor contributes to sensitizing
Gram-negative strains to vancomycin while retaining anti-Gram-positive
activity.
Collapse
Affiliation(s)
- Emma van Groesen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Cornelis J. Slingerland
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Paolo Innocenti
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| |
Collapse
|
16
|
Artim CM, Kunala M, O'Leary MK, Alabi CA. PEGylated Oligothioetheramide Prodrugs Activated by Host Serum Proteases. Chembiochem 2021; 22:2697-2702. [PMID: 34227209 PMCID: PMC8497000 DOI: 10.1002/cbic.202100146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/03/2021] [Indexed: 11/08/2022]
Abstract
Due to the increasing prominence of antibiotic resistance, novel drug discovery and delivery approaches targeting bacteria are essential. In this work we evaluate a prodrug design to improve the cytotoxic profile of polycationic oligothioetheramides (oligoTEAs), which are promising antimicrobials. Herein we chemically modify the oligoTEA, PDT-4G, with a polyethylene glycol (PEG) and show that 1, 2, and 5 kDa PEGs mitigate cytotoxicity. As PEGylation reduces antibacterial activity, we evaluate two peptide linkers which, unlike oligoTEAs, are susceptible to proteolytic cleavage in serum. To gain insight into the prodrug reactivation, two linkers were tested, the 5-residue peptide sequence LMPTG, and the dipeptide sequence VC-PABC. In the presence of 20 % serum, prodrugs made with the VC-PABC linker successfully inhibited bacterial growth. Overall, we observed reactivation of oligoTEAs facilitated by serum protease cleavage of the peptide linkers. This work opens the door to the future design of antimicrobial prodrugs with tunable release profiles.
Collapse
Affiliation(s)
- Christine M Artim
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY 14853, USA
| | - Manisha Kunala
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY 14853, USA
| | - Meghan K O'Leary
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY 14853, USA
| | - Christopher A Alabi
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY 14853, USA
| |
Collapse
|
17
|
Kato Y, Tonomura Y, Hanafusa H, Nishimura K, Fukushima T, Ueno M. Adult Zebrafish Model for Screening Drug-Induced Kidney Injury. Toxicol Sci 2021; 174:241-253. [PMID: 32040193 DOI: 10.1093/toxsci/kfaa009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drug-induced kidney injury is a serious safety issue in drug development. In this study, we evaluated the usefulness of adult zebrafish as a small in vivo system for detecting drug-induced kidney injury. We first investigated the effects of typical nephrotoxicants, gentamicin and doxorubicin, on adult zebrafish. We found that gentamicin induced renal tubular necrosis with increased lysosome and myeloid bodies, and doxorubicin caused foot process fusion of glomerular podocytes. These findings were similar to those seen in mammals, suggesting a common pathogenesis. Second, to further evaluate the performance of the model in detecting drug-induced kidney injury, adult zebrafish were treated with 28 nephrotoxicants or 14 nonnephrotoxicants for up to 4 days, euthanized 24 h after the final treatment, and examined histopathologically. Sixteen of the 28 nephrotoxicants and none of the 14 nonnephrotoxicants caused drug-induced kidney injury in zebrafish (sensitivity, 57%; specificity, 100%; positive predictive value, 100%; negative predictive value, 54%). Finally, we explored genomic biomarker candidates using kidneys isolated from gentamicin- and cisplatin-treated zebrafish using microarray analysis and identified 3 candidate genes, egr1, atf3, and fos based on increased expression levels and biological implications. The expression of these genes was upregulated dose dependently in cisplatin-treated groups and was > 25-fold higher in gentamicin-treated than in the control group. In conclusion, these results suggest that the adult zebrafish has (1) similar nephrotoxic response to those of mammals, (2) considerable feasibility as an experimental model for toxicity studies, and (3) applicability to pathological examination and genomic biomarker evaluation in drug-induced kidney injury.
Collapse
Affiliation(s)
- Yuki Kato
- Drug Safety Evaluation, Research Laboratory for Development, Shionogi and Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Yutaka Tonomura
- Drug Safety Evaluation, Research Laboratory for Development, Shionogi and Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Hiroyuki Hanafusa
- Drug Safety Evaluation, Research Laboratory for Development, Shionogi and Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Kyohei Nishimura
- Drug Safety Evaluation, Research Laboratory for Development, Shionogi and Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Tamio Fukushima
- Drug Safety Evaluation, Research Laboratory for Development, Shionogi and Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Motonobu Ueno
- Drug Safety Evaluation, Research Laboratory for Development, Shionogi and Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| |
Collapse
|
18
|
Abstract
Antibiotic resistance is a major global health challenge and, worryingly, several key Gram negative pathogens can become resistant to most currently available antibiotics. Polymyxins have been revived as a last-line therapeutic option for the treatment of infections caused by multidrug-resistant Gram negative bacteria, in particular Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacterales. Polymyxins were first discovered in the late 1940s but were abandoned soon after their approval in the late 1950s as a result of toxicities (e.g., nephrotoxicity) and the availability of "safer" antibiotics approved at that time. Therefore, knowledge on polymyxins had been scarce until recently, when enormous efforts have been made by several research teams around the world to elucidate the chemical, microbiological, pharmacokinetic/pharmacodynamic, and toxicological properties of polymyxins. One of the major achievements is the development of the first scientifically based dosage regimens for colistin that are crucial to ensure its safe and effective use in patients. Although the guideline has not been developed for polymyxin B, a large clinical trial is currently being conducted to optimize its clinical use. Importantly, several novel, safer polymyxin-like lipopeptides are developed to overcome the nephrotoxicity, poor efficacy against pulmonary infections, and narrow therapeutic windows of the currently used polymyxin B and colistin. This review discusses the latest achievements on polymyxins and highlights the major challenges ahead in optimizing their clinical use and discovering new-generation polymyxins. To save lives from the deadly infections caused by Gram negative "superbugs," every effort must be made to improve the clinical utility of the last-line polymyxins. SIGNIFICANCE STATEMENT: Antimicrobial resistance poses a significant threat to global health. The increasing prevalence of multidrug-resistant (MDR) bacterial infections has been highlighted by leading global health organizations and authorities. Polymyxins are a last-line defense against difficult-to-treat MDR Gram negative pathogens. Unfortunately, the pharmacological information on polymyxins was very limited until recently. This review provides a comprehensive overview on the major achievements and challenges in polymyxin pharmacology and clinical use and how the recent findings have been employed to improve clinical practice worldwide.
Collapse
Affiliation(s)
- Sue C Nang
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Mohammad A K Azad
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Tony Velkov
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Qi Tony Zhou
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| |
Collapse
|
19
|
Abstract
Despite efforts to develop new antibiotics, antibacterial resistance still develops too fast for drug discovery to keep pace. Often, resistance against a new drug develops even before it reaches the market. This continued resistance crisis has demonstrated that resistance to antibiotics with single protein targets develops too rapidly to be sustainable. Most successful long-established antibiotics target more than one molecule or possess targets, which are encoded by multiple genes. This realization has motivated a change in antibiotic development toward drug candidates with multiple targets. Some mechanisms of action presuppose multiple targets or at least multiple effects, such as targeting the cytoplasmic membrane or the carrier molecule bactoprenol phosphate and are therefore particularly promising. Moreover, combination therapy approaches are being developed to break antibiotic resistance or to sensitize bacteria to antibiotic action. In this Review, we provide an overview of antibacterial multitarget approaches and the mechanisms behind them.
Collapse
Affiliation(s)
- Declan Alan Gray
- Newcastle University
Biosciences Institute, Newcastle University, NE2 4HH Newcastle
upon Tyne, United Kingdom
| | - Michaela Wenzel
- Division of Chemical
Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| |
Collapse
|
20
|
Dai C, Wang Y, Sharma G, Shen J, Velkov T, Xiao X. Polymyxins-Curcumin Combination Antimicrobial Therapy: Safety Implications and Efficacy for Infection Treatment. Antioxidants (Basel) 2020; 9:antiox9060506. [PMID: 32526966 PMCID: PMC7346118 DOI: 10.3390/antiox9060506] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
The emergence of antimicrobial resistance in Gram-negative bacteria poses a huge health challenge. The therapeutic use of polymyxins (i.e., colistin and polymyxin B) is commonplace due to high efficacy and limiting treatment options for multidrug-resistant Gram-negative bacterial infections. Nephrotoxicity and neurotoxicity are the major dose-limiting factors that limit the therapeutic window of polymyxins; nephrotoxicity is a complication in up to ~60% of patients. The emergence of polymyxin-resistant strains or polymyxin heteroresistance is also a limiting factor. These caveats have catalyzed the search for polymyxin combinations that synergistically kill polymyxin-susceptible and resistant organisms and/or minimize the unwanted side effects. Curcumin—an FDA-approved natural product—exerts many pharmacological activities. Recent studies showed that polymyxins–curcumin combinations showed a synergistically inhibitory effect on the growth of bacteria (e.g., Gram-positive and Gram-negative bacteria) in vitro. Moreover, curcumin co-administration ameliorated colistin-induced nephrotoxicity and neurotoxicity by inhibiting oxidative stress, mitochondrial dysfunction, inflammation and apoptosis. In this review, we summarize the current knowledge-base of polymyxins–curcumin combination therapy and discuss the underlying mechanisms. For the clinical translation of this combination to become a reality, further research is required to develop novel polymyxins–curcumin formulations with optimized pharmacokinetics and dosage regimens.
Collapse
Affiliation(s)
- Chongshan Dai
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing 100193, China; (Y.W.); (J.S.)
- Correspondence: (C.D.); (X.X.); Tel.: +86-156-5282-6026 (C.D.); +86-010-6273-3377 (X.X.)
| | - Yang Wang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing 100193, China; (Y.W.); (J.S.)
| | - Gaurav Sharma
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Jianzhong Shen
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing 100193, China; (Y.W.); (J.S.)
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, Faculty of Medicine, School of Biomedical Sciences, Dentistry and Health Sciences, the University of Melbourne, Parkville 3052, Australia;
| | - Xilong Xiao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing 100193, China; (Y.W.); (J.S.)
- Correspondence: (C.D.); (X.X.); Tel.: +86-156-5282-6026 (C.D.); +86-010-6273-3377 (X.X.)
| |
Collapse
|
21
|
Fowler S, Chen WLK, Duignan DB, Gupta A, Hariparsad N, Kenny JR, Lai WG, Liras J, Phillips JA, Gan J. Microphysiological systems for ADME-related applications: current status and recommendations for system development and characterization. LAB ON A CHIP 2020; 20:446-467. [PMID: 31932816 DOI: 10.1039/c9lc00857h] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over the last decade, progress has been made on the development of microphysiological systems (MPS) for absorption, distribution, metabolism, and excretion (ADME) applications. Central to this progress has been proof of concept data generated by academic and industrial institutions followed by broader characterization studies, which provide evidence for scalability and applicability to drug discovery and development. In this review, we describe some of the advances made for specific tissue MPS and outline the desired functionality for such systems, which are likely to make them applicable for practical use in the pharmaceutical industry. Single organ MPS platforms will be valuable for modelling tissue-specific functions. However, dynamic organ crosstalk, especially in the context of disease or toxicity, can only be obtained with the use of inter-linked MPS models which will enable scientists to address questions at the intersection of pharmacokinetics (PK) and efficacy, or PK and toxicity. In the future, successful application of MPS platforms that closely mimic human physiology may ultimately reduce the need for animal models to predict ADME outcomes and decrease the overall risk and cost associated with drug development.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharma Research and Early Development, F.Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | | | - David B Duignan
- Department of Drug Metabolism, Pharmacokinetics & Bioanalysis, AbbVie Bioresearch Center, Worcester, Massachusetts 01605, USA
| | - Anshul Gupta
- Amgen Research, 360 Binney St, Cambridge, MA 02141, USA
| | - Niresh Hariparsad
- Department of Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, USA
| | - Jane R Kenny
- DMPK, Genentech, 1 DNA Way, South San Francisco 94080, USA
| | | | - Jennifer Liras
- Medicine Design, Pfizer Inc, 1 Portland Ave, Cambridge, MA 02139, USA
| | | | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb R&D, PO Box 4000, Princeton, NJ 08543-4000, USA.
| |
Collapse
|
22
|
Troth SP, Vlasakova K, Amur S, Amin RP, Glaab WE. Translational Safety Biomarkers of Kidney Injury. Semin Nephrol 2019; 39:202-214. [PMID: 30827342 DOI: 10.1016/j.semnephrol.2018.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Acute kidney injury continues to be a common problem and there continues to be a medical need for sensitive translational biomarkers for clinical monitoring. The past decade has yielded unprecedented progress in fundamental research into novel kidney biomarker evaluation and the mechanistic understanding of kidney injury; as such, these novel biomarkers increasingly are being used in preclinical drug development and in early clinical trials of drug candidates on a case-by-case basis, as well as in medical and veterinary practice. With the recent successful clinical qualification of a subset of novel accessible biomarker candidates for use in early phase clinical trials, continued clinical evaluation may enable expanded regulatory qualification for more generalized clinical use. This review provides a comprehensive overview about the discovery and development of kidney safety biomarkers with a focus on current progress in nonclinical research, progress toward translation to the clinic, and perspectives on future opportunities.
Collapse
Affiliation(s)
- Sean P Troth
- Merck Research Laboratories, Department of Safety Assessment and Laboratory Animal Resources, Merck & Co, Inc, West Point, PA.
| | - Katerina Vlasakova
- Merck Research Laboratories, Department of Safety Assessment and Laboratory Animal Resources, Merck & Co, Inc, West Point, PA
| | - Shashi Amur
- Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Rupesh P Amin
- Merck Research Laboratories, Department of Safety Assessment and Laboratory Animal Resources, Merck & Co, Inc, West Point, PA
| | - Warren E Glaab
- Merck Research Laboratories, Department of Safety Assessment and Laboratory Animal Resources, Merck & Co, Inc, West Point, PA
| |
Collapse
|
23
|
Renal glycosuria as a novel early sign of colistin-induced kidney damage in mice. Antimicrob Agents Chemother 2019:AAC.01650-19. [PMID: 31591120 PMCID: PMC6879251 DOI: 10.1128/aac.01650-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The polymyxin colistin represents a last-resort antibiotic for multidrug-resistant infections, but its use is limited by the frequent onset of acute drug-induced kidney injury (DIKI). It is essential to closely monitor kidney function prior to and during colistin treatment in order to pinpoint early signs of injury and minimize long-term renal dysfunction. To facilitate this, a mouse model of colistin-induced nephrotoxicity was used to uncover novel early markers of colistin-induced DIKI. The polymyxin colistin represents a last-resort antibiotic for multidrug-resistant infections, but its use is limited by the frequent onset of acute drug-induced kidney injury (DIKI). It is essential to closely monitor kidney function prior to and during colistin treatment in order to pinpoint early signs of injury and minimize long-term renal dysfunction. To facilitate this, a mouse model of colistin-induced nephrotoxicity was used to uncover novel early markers of colistin-induced DIKI. Increased urinary levels of kidney injury molecule-1 (Kim-1) as well as glycosuria were observed in colistin-treated mice, where alterations of established clinical markers of acute kidney injury (serum creatinine and albuminuria) and emerging markers such as cystatin C were inaccurate in flagging renal damage as confirmed by histology. A direct interaction of colistin with renal glucose reabsorption was ruled out by a cis-inhibition assay in mouse brush border membrane vesicles (BBMV). Immunohistochemical examination and protein quantification by Western blotting showed a marked reduction in the protein amount of sodium-glucose transporter 2 (Sglt2), the main kidney glucose transporter, in renal tissue from colistin-treated mice in comparison to that in control animals. Consistently, BBMV isolated from treated mouse kidneys also showed a reduction in ex vivo glucose uptake compared to that in BBMV isolated from control kidneys. These findings support pathology observations of colistin-induced proximal tubule damage at the site of the brush border membrane, where Sglt2 is expressed, and open avenues for the study of glycosuria as a sensitive, specific, and accessible marker of DIKI during colistin therapy.
Collapse
|
24
|
Barnett LMA, Cummings BS. Nephrotoxicity and Renal Pathophysiology: A Contemporary Perspective. Toxicol Sci 2019; 164:379-390. [PMID: 29939355 DOI: 10.1093/toxsci/kfy159] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The kidney consists of numerous cell types organized into the nephron, which is the basic functional unit of the kidney. Any stimuli that induce loss of these cells can induce kidney damage and renal failure. The cause of renal failure can be intrinsic or extrinsic. Extrinsic causes include cardiovascular disease, obesity, diabetes, sepsis, and lung and liver failure. Intrinsic causes include glomerular nephritis, polycystic kidney disease, renal fibrosis, tubular cell death, and stones. The kidney plays a prominent role in mediating the toxicity of numerous drugs, environmental pollutants and natural substances. Drugs known to be nephrotoxic include several cancer therapeutics, drugs of abuse, antibiotics, and radiocontrast agents. Environmental pollutants known to target the kidney include cadmium, mercury, arsenic, lead, trichloroethylene, bromate, brominated-flame retardants, diglycolic acid, and ethylene glycol. Natural nephrotoxicants include aristolochic acids and mycotoxins such as ochratoxin, fumonisin B1, and citrinin. There are several common characteristics between mechanisms of renal failure induced by nephrotoxicants and extrinsic causes. This common ground exists primarily due to similarities in the molecular mechanisms mediating renal cell death. This review summarizes the current state of the field of nephrotoxicity. It emphasizes integrating our understanding of nephrotoxicity with pathological-induced renal failure. Such approaches are needed to address major questions in the field, which include the diagnosis, prognosis and treatment of both acute and chronic renal failure, and the progression of acute kidney injury to chronic kidney disease.
Collapse
Affiliation(s)
| | - Brian S Cummings
- Interdisciplinary Toxicology Program.,Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
25
|
Korucu B, Unal I, Pekcan M, Inkaya AC, Yeter H, Cetinkaya MA, Kaymaz FF, Unal S, Akova M, Erdem Y. Ultrastructural evaluation of urine alkalinization versus hydration on colistin-induced nephrotoxicity. Hum Exp Toxicol 2019; 38:1366-1377. [PMID: 31280613 DOI: 10.1177/0960327119862339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Colistin is a vital antibiotic used in multidrug-resistant infections. Its most important side effect is nephrotoxicity. Colistin is a weak acid. This study aims to evaluate whether urine alkalinization is protective in the nephrotoxicity of colistin. METHODS Twenty-eight male Sprague-Dawley rats were divided into groups. Group I (n = 4) was injected with intramuscular distilled water twice a day for 7 days. Group II (n = 8) was injected with 750,000 IU/kg/day colistin for 7 days. Group III (n = 8) was injected with the same dose of colistin after their urinary pH was ≥7 through the addition of bicarbonate in their drinking water. Group IV (n = 8) was injected with the same dose of colistin after their urine density fell below 1010 through the addition of NaCl molds in their food and 12.6 mg/L NaCl in their drinking water. RESULTS According to tubular degenerations (scored 0-5), group I scored 0, group II scored 4.25, group III scored 2, and group IV scored 1.5. In groups III and IV, protection was achieved (p = 0.001). The bicarbonate group was not superior to the NaCl group (p = 0.789). In transmission electron microscopy, group III had more microvilli integrity and autophagic vacuoles compared to group IV. Group IV had mitochondrial swelling and cristae lysis. A lower urine density was related to lower tubular scores (p = 0.001). CONCLUSIONS Colistin was highly nephrotoxic without protection. Light microscopy findings revealed that urinary alkalinization and NaCl hydration were similarly protective. Urine alkalinization further prevents ultrastructural changes as revealed by electron microscopy.
Collapse
Affiliation(s)
- B Korucu
- Department of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - I Unal
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - M Pekcan
- Department of Biochemistry, Ankara University Faculty of Veterinary Medicine, Ankara, Turkey
| | - A C Inkaya
- Department of Infectious Diseases, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - H Yeter
- Department of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - M A Cetinkaya
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - F F Kaymaz
- Department of Infectious Diseases, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - S Unal
- Department of Infectious Diseases, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - M Akova
- Department of Infectious Diseases, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Y Erdem
- Department of Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
26
|
Shifting Gears: The Future of Polymyxin Antibiotics. Antibiotics (Basel) 2019; 8:antibiotics8020042. [PMID: 31013818 PMCID: PMC6628003 DOI: 10.3390/antibiotics8020042] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/05/2019] [Accepted: 04/09/2019] [Indexed: 11/17/2022] Open
Abstract
The manuscripts contained in this special edition of Antibiotics represent a current review of the polymyxins as well as highlights from the 3rd International Polymyxin Conference, which was held in Madrid, Spain, April 25 to 26, 2018. The role of the polymyxin antibiotics has evolved over time based on the availability of alternative agents. After high rates of nephrotoxicity caused the drug class to fall out of favor, polymyxins were once against utilized in the 21st century to combat drug-resistant pathogens. However, the introduction of safer agents with activity against drug-resistant organisms has brought the future utility of polymyxins into question. The present review investigates the future niche of polymyxins by evaluating currently available and future treatment options for difficult-to-treat pathogens. The introduction of ceftazidime-avibactam, meropenem-vaborbactam and plazomicin are likely to decrease polymyxin utilization for infections caused by Enterobacteriaceae. Similarly, the availability of ceftolozane-tazobactam will reduce the use of polymyxins to counter multidrug-resistant Pseudomonas aeruginosa. In contrast, polymyxins will likely continue be an important option for combatting carbapenem-resistant Acinetobacter baumannii until better options become commercially available. Measuring polymyxin concentrations in patients and individualizing therapy may be a future strategy to optimize clinical outcomes while minimizing nephrotoxicity. Inhaled polymyxins will continue to be an adjunctive option for pulmonary infections but further clinical trials are needed to clarify the efficacy of inhaled polymyxins. Lastly, safer polymyxin analogs will potentially be an important addition to the antimicrobial armamentarium.
Collapse
|
27
|
Rat models of colistin nephrotoxicity: previous experimental researches and future perspectives. Eur J Clin Microbiol Infect Dis 2019; 38:1387-1393. [PMID: 30949899 DOI: 10.1007/s10096-019-03546-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/25/2019] [Indexed: 01/12/2023]
Abstract
Colistin is an old antibiotic, which is abandoned decades ago because of high nephrotoxicity rates. However, it is reintroduced to clinical medicine due to lack of newly discovered antibiotics and is still widely used for the treatment of resistant gram-negative infections. Discovering mechanisms to reduce nephrotoxicity risk is of significant importance since exposed patients may have many other factors that alter kidney functions. Several agents were evaluated in animal models of colistin nephrotoxicity as a means to prevent kidney injury. Considerable heterogeneity exists in terms of reporting colistin dosing and experimental designs. This issue leads clinicians to face difficulties in designing studies and sometimes may lead to report dosing strategies inadequately. Here, we present a review according to animal models of colistin nephrotoxicity using data gathered from previous experiments to draw attention on possible complexities that researchers may encounter.
Collapse
|
28
|
Diekjürgen D, Grainger DW. A murine ex vivo 3D kidney proximal tubule model predicts clinical drug-induced nephrotoxicity. Arch Toxicol 2019; 93:1349-1364. [PMID: 30863989 DOI: 10.1007/s00204-019-02430-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/05/2019] [Indexed: 12/18/2022]
Abstract
Drug attrition and clinical product withdrawals due to nephrotoxicity remain major challenges for pharmaceutical drug development pipelines. Currently, no reliable high-throughput in vitro screening models are available that provide reliable, predictive toxicology data for clinical nephrotoxicity. Drug screens to predict toxicity and pharmacology assessments are compromised by standard two-dimensional (2D) cell monoculture models. Here we extend a previously reported murine three-dimensional (3D) kidney-derived intact proximal tubule model to provide ex vivo drug toxicity data that reliably compare to clinical experiences and improve nephrotoxicity predictions over current 2D cell assays. Proximal tubule cytotoxicity was monitored by ATP depletion for 12 compounds (acarbose, acetylsalicylic acid, captopril, cimetidine, cidofovir, cisplatin, doxorubicin, gentamicin, polymyxin B, polymyxin B nonapeptide, probenecid and vancomycin) in 3D proximal tubule ex vivo assays. Drug concentration-response curves (1-1000 µM) and IC50, lowest effective concentration (LEC) and AUC values were compared to clinical therapeutic exposure levels (Cmax). The 100-fold Cmax threshold demonstrated best sensitivity (96.9%) and specificity (87.5%) for this assay, with high positive (93.9%) and negative (93.3%) predictive values for nephrotoxicity. IC50 values were superior to LEC. Results also support the model's capability to predict substrate-inhibitor/competitor interactions, yielding toxicity results similar to those reported in vivo. These 3D proximal tubule-based drug screens provide more reliable nephrotoxicity predictions, and more insight into complex mechanisms implicated in nephrotoxicity than current standard 2D cell assays. This new approach for rapid drug toxicity testing produces more reliable clinical comparisons than current 2D cell culture screening techniques.
Collapse
Affiliation(s)
- Dorina Diekjürgen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA. .,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112-5820, USA.
| |
Collapse
|
29
|
A permeability-increasing drug synergizes with bacterial efflux pump inhibitors and restores susceptibility to antibiotics in multi-drug resistant Pseudomonas aeruginosa strains. Sci Rep 2019; 9:3452. [PMID: 30837499 PMCID: PMC6401119 DOI: 10.1038/s41598-019-39659-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 01/28/2019] [Indexed: 01/06/2023] Open
Abstract
Resistance to antibiotics poses a major global threat according to the World Health Organization. Restoring the activity of existing drugs is an attractive alternative to address this challenge. One of the most efficient mechanisms of bacterial resistance involves the expression of efflux pump systems capable of expelling antibiotics from the cell. Although there are efflux pump inhibitors (EPIs) available, these molecules are toxic for humans. We hypothesized that permeability-increasing antimicrobial peptides (AMPs) could lower the amount of EPI necessary to sensitize bacteria to antibiotics that are efflux substrates. To test this hypothesis, we measured the ability of polymyxin B nonapeptide (PMBN), to synergize with antibiotics in the presence of EPIs. Assays were performed using planktonic and biofilm-forming cells of Pseudomonas aeruginosa strains overexpressing the MexAB-OprM efflux system. Synergy between PMBN and EPIs boosted azithromycin activity by a factor of 2,133 and sensitized P. aeruginosa to all tested antibiotics. This reduced several orders of magnitude the amount of inhibitor needed for antibiotic sensitization. The selected antibiotic-EPI-PMBN combination caused a 10 million-fold reduction in the viability of biofilm forming cells. We proved that AMPs can synergize with EPIs and that this phenomenon can be exploited to sensitize bacteria to antibiotics.
Collapse
|
30
|
Molecular Mechanisms of Colistin-Induced Nephrotoxicity. Molecules 2019; 24:molecules24030653. [PMID: 30759858 PMCID: PMC6384669 DOI: 10.3390/molecules24030653] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
The emergence of multidrug resistant (MDR) infections and the shortage of new therapeutic options have made colistin, a polymyxin antibiotic, the main option for the treatment of MDR Gram-negative bacterial infections in the last decade. However, the rapid onset of renal damage often prevents the achievement of optimal therapeutic doses and/or forces the physicians to interrupt the therapy, increasing the risk of drug resistance. The proper management of colistin-induced nephrotoxicity remains challenging, mostly because the investigation of the cellular and molecular pharmacology of this drug, off the market for decades, has been largely neglected. For years, the renal damage induced by colistin was considered a mere consequence of the detergent activity of this drug on the cell membrane of proximal tubule cells. Lately, it has been proposed that the intracellular accumulation is a precondition for colistin-mediated renal damage, and that mitochondria might be a primary site of damage. Antioxidant approaches (e.g., ascorbic acid) have shown promising results in protecting the kidney of rodents exposed to colistin, yet none of these strategies have yet reached the bedside. Here we provide a critical overview of the possible mechanisms that may contribute to colistin-induced renal damage and the potential protective strategies under investigation.
Collapse
|
31
|
Polymyxin Derivatives that Sensitize Gram-Negative Bacteria to Other Antibiotics. Molecules 2019; 24:molecules24020249. [PMID: 30641878 PMCID: PMC6359160 DOI: 10.3390/molecules24020249] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/27/2018] [Accepted: 01/07/2019] [Indexed: 01/11/2023] Open
Abstract
Polymyxins (polymyxin B (PMB) and polymyxin E (colistin)) are cyclic lipodecapeptide antibiotics, highly basic due to five free amino groups, and rapidly bactericidal against Gram-negative bacteria, such as the majority of Enterobacteriaceae as well as Acinetobacter baumannii and Pseudomonas aeruginosa. Their clinical use was abandoned in the 1960s because of nephrotoxicity and because better-tolerated drugs belonging to other antibiotic classes were introduced. Now, due to the global dissemination of extremely-drug resistant Gram-negative bacterial strains, polymyxins have resurged as the last-line drugs against those strains. Novel derivatives that are less toxic and/or more effective at tolerable doses are currently under preclinical development and their properties have recently been described in several extensive reviews. Other derivatives lack any direct bactericidal activity but damage the outermost permeability barrier, the outer membrane, of the target bacteria and make it more permeable to many other antibiotics. This review describes the properties of three thus far best-characterized “permeabilizer” derivatives, i.e., the classic permeabilizer polymyxin B nonapeptide (PMBN), NAB7061, and SPR741/NAB741, a compound that recently successfully passed the clinical phase 1. Also, a few other permeabilizer compounds are brought up.
Collapse
|
32
|
Assadi F. The Growing Epidemic of Chronic Kidney Disease: Preventive Strategies to Delay the Risk for Progression to ESRD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1121:57-59. [PMID: 31392652 DOI: 10.1007/978-3-030-10616-4_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hypertension, obesity and metabolic syndromes are leading risk factors for the development of chronic kidney disease (CKD). Considering the high prevalence of hypertension and obesity in children and adolescents and it's risk of progression to cardiovascular disease, CKD should be considered a serious long-term health issue in children with metabolic syndrome. Prevention of CKD requires a professional teamwork consisting of primary care physicians, nephrologists, nutritionist, pharmacist, and social work to identify and manage children at risk of developing CKD in order to provide a highly valuable management strategies. This review focuses on the principles underlying the importance of a team approach for CKD prevention.
Collapse
Affiliation(s)
- Farahnak Assadi
- Department of Pediatrics, Division of Nephrology, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
33
|
Weber EJ, Lidberg KA, Wang L, Bammler TK, MacDonald JW, Li MJ, Redhair M, Atkins WM, Tran C, Hines KM, Herron J, Xu L, Monteiro MB, Ramm S, Vaidya V, Vaara M, Vaara T, Himmelfarb J, Kelly EJ. Human kidney on a chip assessment of polymyxin antibiotic nephrotoxicity. JCI Insight 2018; 3:123673. [PMID: 30568031 DOI: 10.1172/jci.insight.123673] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/06/2018] [Indexed: 12/22/2022] Open
Abstract
Drug-induced kidney injury, largely caused by proximal tubular intoxicants, limits development and clinical use of new and approved drugs. Assessing preclinical nephrotoxicity relies on animal models that are frequently insensitive; thus, potentially novel techniques - including human microphysiological systems, or "organs on chips" - are proposed to accelerate drug development and predict safety. Polymyxins are potent antibiotics against multidrug-resistant microorganisms; however, clinical use remains restricted because of high risk of nephrotoxicity and limited understanding of toxicological mechanisms. To mitigate risks, structural analogs of polymyxins (NAB739 and NAB741) are currently in clinical development. Using a microphysiological system to model human kidney proximal tubule, we exposed cells to polymyxin B (PMB) and observed significant increases of injury signals, including kidney injury molecule-1 KIM-1and a panel of injury-associated miRNAs (each P < 0.001). Surprisingly, transcriptional profiling identified cholesterol biosynthesis as the primary cellular pathway induced by PMB (P = 1.22 ×10-16), and effluent cholesterol concentrations were significantly increased after exposure (P < 0.01). Additionally, we observed no upregulation of the nuclear factor (erythroid derived-2)-like 2 pathway, despite this being a common pathway upregulated in response to proximal tubule toxicants. In contrast with PMB exposure, minimal changes in gene expression, injury biomarkers, and cholesterol concentrations were observed in response to NAB739 and NAB741. Our findings demonstrate the preclinical safety of NAB739 and NAB741 and reveal cholesterol biosynthesis as a potentially novel pathway for PMB-induced injury. To our knowledge, this is the first demonstration of a human-on-chip platform used for simultaneous safety testing of new chemical entities and defining unique toxicological pathway responses of an FDA-approved molecule.
Collapse
Affiliation(s)
| | | | - Lu Wang
- Department of Environmental and Occupational Health Sciences, and
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, and
| | | | - Mavis J Li
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Michelle Redhair
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Cecilia Tran
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Kelly M Hines
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Josi Herron
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Maria Beatriz Monteiro
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, USA
| | - Susanne Ramm
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, USA
| | - Vishal Vaidya
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, USA
| | - Martti Vaara
- Northern Antibiotics Ltd., Espoo, Finland.,Division of Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland.,Department of Bacteriology and Immunology, Helsinki University Medical School, Helsinki, Finland
| | - Timo Vaara
- Northern Antibiotics Ltd., Espoo, Finland
| | - Jonathan Himmelfarb
- Department of Medicine, Division of Nephrology, Kidney Research Institute, Seattle, Washington, USA
| | | |
Collapse
|
34
|
Gameiro J, Agapito Fonseca J, Jorge S, Lopes JA. Acute kidney injury in HIV-infected patients: a critical review. HIV Med 2018; 20:77-87. [PMID: 30411475 DOI: 10.1111/hiv.12685] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2018] [Indexed: 12/20/2022]
Abstract
Acute kidney injury (AKI) is characterized by a rapid decline of renal function associated with worse outcomes. The purpose of the authors is to perform a critical review of the incidence, risk factors, pathogenesis and outcome of AKI in HIV-infected patients. Human immunodeficiency virus (HIV)-infected patients have an increased risk of developing AKI, to which contribute both HIV-dependent and HIV-independent factors as well as the nephrotoxicity of drugs used. The increased risk of AKI in HIV-infected patients and its negative impact on prognosis highlights the need for identification of patients at risk, creation of prevention strategies and management. HIV-infected patients have an increased risk of developing AKI, to which both HIV-dependent and HIV-independent factors contribute, as well as the nephrotoxicity of drugs used. The increased risk of AKI in HIV-infected patients and its negative impact on prognosis highlight the need for identification of patients at risk, creation of prevention strategies and management.
Collapse
Affiliation(s)
- J Gameiro
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Lisboa Norte, EPE, Lisboa, Portugal
| | - J Agapito Fonseca
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Lisboa Norte, EPE, Lisboa, Portugal
| | - S Jorge
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Lisboa Norte, EPE, Lisboa, Portugal
| | - J A Lopes
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Lisboa Norte, EPE, Lisboa, Portugal
| |
Collapse
|
35
|
Hoff EA, Artim CM, Brown JS, Alabi CA. Sensitivity of Antibacterial Activity to Backbone Sequence in Constitutionally Isomeric OligoTEAs. Macromol Biosci 2018; 18:e1800241. [PMID: 30238615 DOI: 10.1002/mabi.201800241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/03/2018] [Indexed: 01/28/2023]
Abstract
Antimicrobial peptides are promising alternatives to traditional antibiotics but their translational potential is limited due to rapid degradation by serum proteases. Recently, a number of peptidomimetics with backbones resistant to proteolysis have been synthesized and their antimicrobial potential evaluated as a function of their hydrophobic to cationic ratio. However, these mimetics also have a fixed backbone thus making it difficult to isolate the effect of backbone hydrophobic composition and sequence. In this work, advantage is taken of the oligothioetheramide (oligoTEA) synthetic strategy that allows for precise control over backbone and pendant group placement to systematically study the effect of backbone hydrophobic sequence while keeping pendant group constant. Biophysical data acquired with a set of constitutional oligoTEA isomers show that backbone hydrophobic sequence, that is, local hydrophobicity, affects the mode of oligoTEA interaction with lipid bilayers. This differential interaction among the constitutionally isomeric oligoTEAs is manifested in their antibacterial activities and points to the possibility of using backbone hydrophobic sequence to tune antibacterial potency and selectivity.
Collapse
Affiliation(s)
- Emily A Hoff
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY, 14853, USA
| | - Christine M Artim
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY, 14853, USA
| | - Joseph S Brown
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY, 14853, USA
| | - Christopher A Alabi
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, NY, 14853, USA
| |
Collapse
|
36
|
Qiu X, Zhou X, Miao Y, Li B. An in vitro method for nephrotoxicity evaluation using HK-2 human kidney epithelial cells combined with biomarkers of nephrotoxicity. Toxicol Res (Camb) 2018; 7:1205-1213. [PMID: 30510689 DOI: 10.1039/c8tx00095f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/06/2018] [Indexed: 02/02/2023] Open
Abstract
The kidney is one of the major target organs for drug-induced toxicity. During drug development, the traditional markers of nephrotoxicity indicate only severe and late damage, which leads to high costs. The new biomarkers are needed for a more sensitive and reliable evaluation of nephrotoxicity, especially for the regulatory accepted and validated in vitro model. We developed an in vitro model based on the HK-2 cell using the biomarkers of nephrotoxicity as endpoints for the evaluation of nephrotoxicity. The predictive performance of the biomarkers including LDH, GGT, KIM-1, clusterin, CysC, NGAL, TIMP-1, GSTπ and osteopontin was evaluated with 22 well characterized compounds. The area under the curve (AUC) values of KIM-1, clusterin, CysC and osteopontin ranged between 0.79 and 0.84. The combination of clusterin, KIM-1 and/or osteopontin improved the AUC value (ranging between 0.88 and 0.95) compared to one biomarker. Taken together, these results suggest that the model based on the HK-2 cell using clusterin, osteopontin, CysC and KIM-1 as endpoints would allow the prediction of nephrotoxicity at early preclinical stages.
Collapse
Affiliation(s)
- Xuan Qiu
- Chinese Academy of Medical Sciences & Peking Union Medical College , No. 9 , Dongdan Santiao , Dongcheng District , Beijing 100730 , China . .,National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| | - Xiaobing Zhou
- National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| | - Yufa Miao
- National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College , No. 9 , Dongdan Santiao , Dongcheng District , Beijing 100730 , China . .,National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| |
Collapse
|
37
|
Artim CM, Phan NN, Alabi CA. Effect of Composition on Antibacterial Activity of Sequence-Defined Cationic Oligothioetheramides. ACS Infect Dis 2018; 4:1257-1263. [PMID: 29750860 DOI: 10.1021/acsinfecdis.8b00079] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In response to the urgent need for new antibiotic development strategies, antimicrobial peptides and their synthetic mimetics are being investigated as promising alternatives to traditional antibiotics. To facilitate their development into clinically viable candidates, we need to understand what molecular features and physicochemical properties are needed to induce cell death. Within the context of sequence-defined oligothioetheramides (oligoTEAs), we explore the impact of the cationic pendant group and backbone hydrophobicity on the potency and selectivity of antibacterial oligoTEAs. Through antibacterial, cytotoxicity, membrane destabilization, and membrane depolarization assays, we find a strong dependency on the nature of the cationic group and improved selectivity toward bacteria by tuning backbone hydrophobicity. In particular, compounds with the guanidinium headgroup are more potent than those with amines. Finally, we identify a promising oligoTEA, PDT-4G, with enhanced activity in vitro (minimum inhibitory concentration (MIC) ∼ 0.78 μM) and moderate activity in a mouse thigh infection model of methicillin-resistant Staphylococcus aureus. The studies outlined in this work provide insights into the effect of macromolecular physicochemical properties on antibacterial potency. This knowledge base will be vital for researchers engaged in the ongoing development of clinically viable antibacterial agents.
Collapse
Affiliation(s)
- Christine M. Artim
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
| | - Ngoc N. Phan
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
| | - Christopher A. Alabi
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
| |
Collapse
|
38
|
Talih G, Esmaoğlu A, Bayram A, Yazici C, Deniz K, Talih T. Dexmedetomidina impede a nefrotoxicidade da colistina? Braz J Anesthesiol 2018; 68:383-387. [DOI: 10.1016/j.bjan.2018.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 01/11/2018] [Accepted: 01/25/2018] [Indexed: 12/31/2022] Open
|
39
|
Talih G, Esmaoğlu A, Bayram A, Yazici C, Deniz K, Talih T. Does dexmedetomidine prevent colistin nephrotoxicity? Braz J Anesthesiol 2018. [PMID: 29885938 PMCID: PMC9391814 DOI: 10.1016/j.bjane.2018.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background In this study, we aimed to investigate the effect of dexmedetomidine on colistin nephrotoxicity in rats. Methods Thirty-two Wistar albino rats were allocated into four groups. Intraperitoneal (ip) saline at 1 mL.kg−1 was administered to the control group and 10 mg.kg−1 ip colistin was given to the colistin group. In the DEX10 group 10 mcg.kg−1 dexmedetomidine ip was given 20 min before the injection of 10 mg.kg−1 ip colistin. In the DEX20 group ip 20 mcg.kg−1 dexmedetomidine was injected 20 min before the administration of 10 mg.kg−1 ip colistin. These treatments were continued twice a day for seven days. Samples were taken on the eighth day. BUN, Cr, KIM-1, TAS, and TOS were examined in blood samples and caspase-3 was examined in kidney tissue samples. Results The values for BUN, Cr and TOS were significantly higher in the colistin group than in the control group. BUN, Cr and TOS changes in the DEX10 and DEX20 groups were not significant compared with the control group but they were significantly lower compared with the colistin group. TAS values in the DEX10 group were significantly lower than in the control group. Apoptotic activity was significantly higher in the colistin group compared with the control group, but there was no significant difference in terms of caspase-3 staining activity when DEX10 and DEX20 groups were compared with the control group. Conclusion Oxidative damage and apoptosis played roles in colistin nephrotoxicity, and colistin nephrotoxicity could be prevented by treatment with dexmedetomidine.
Collapse
Affiliation(s)
- Gamze Talih
- Akcakale State Hospital, Department of Anesthesiology and Reanimation, Şanlıurfa, Turquia
| | - Aliye Esmaoğlu
- Erciyes University, Medical Faculty, Department of Anesthesiology and Reanimation, Kayseri, Turquia.
| | - Adnan Bayram
- Erciyes University, Medical Faculty, Department of Anesthesiology and Reanimation, Kayseri, Turquia
| | - Cevat Yazici
- Erciyes University, Medical Faculty, Department of Biochemistry, Kayseri, Turquia
| | - Kemal Deniz
- Erciyes University, Medical Faculty, Department of Pathology, Kayseri, Turquia
| | - Tutkun Talih
- Akcakale State Hospital, Department of General Surgery, Şanlıurfa, Turquia
| |
Collapse
|
40
|
Blaskovich MAT, Pitt ME, Elliott AG, Cooper MA. Can octapeptin antibiotics combat extensively drug-resistant (XDR) bacteria? Expert Rev Anti Infect Ther 2018; 16:485-499. [PMID: 29848132 DOI: 10.1080/14787210.2018.1483240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
INTRODUCTION The octapeptins are a family of cyclic lipopeptides first reported in the 1970s then largely ignored. At the time, their reported antibiotic activity against polymyxin-resistant bacteria was a curiosity. Today, the advent of widespread drug resistance in Gram-negative bacteria has prompted their 'rediscovery.' The paucity of new antibiotics in the clinical pipeline is coupled with a global spread of increasing antibiotic resistance, particularly to meropenem and polymyxins B and E (colistin). Areas covered: We review the original discovery of octapeptins, their recent first chemical syntheses, and their mode of action, then discuss their potential as a new class of antibiotics to treat extensively drug-resistant (XDR) Gram-negative infections, with direct comparisons to the closely related polymyxins. Expert commentary: Cyclic lipopeptides in clinical use (polymyxin antibiotics) have significant dose-limiting nephrotoxicity inherent to their chemotype. This toxicity has prevented improved polymyxin analogs from progressing to the clinic, and tainted the perception of lipopeptide antibiotics in general. We argue that the octapeptins are fundamentally different from the polymyxins, with a disparate mode of action, spectra of action against MDR and XDR bacteria and a superior preclinical safety profile. They represent early-stage candidates that can help prime the antibiotic discovery pipeline.
Collapse
Affiliation(s)
- Mark A T Blaskovich
- a Institute for Molecular Bioscience , The University of Queensland , Brisbane , Queensland , Australia
| | - Miranda E Pitt
- a Institute for Molecular Bioscience , The University of Queensland , Brisbane , Queensland , Australia
| | - Alysha G Elliott
- a Institute for Molecular Bioscience , The University of Queensland , Brisbane , Queensland , Australia
| | - Matthew A Cooper
- a Institute for Molecular Bioscience , The University of Queensland , Brisbane , Queensland , Australia
| |
Collapse
|
41
|
Polymyxin B causes DNA damage in HK-2 cells and mice. Arch Toxicol 2018; 92:2259-2271. [PMID: 29556720 DOI: 10.1007/s00204-018-2192-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
Abstract
Increasing incidence of multidrug-resistant bacteria presents an imminent risk to global health. Polymyxins are 'last-resort' antibiotics against Gram-negative 'superbugs'; however, nephrotoxicity remains a key impediment in their clinical use. Molecular mechanisms underlying this nephrotoxicity remain poorly defined. Here, we examined the pathways which led to polymyxin B induced cell death in vitro and in vivo. Human proximal tubular cells were treated with polymyxin B (12.5-100 μM) for up to 24 h and showed a significant increase in micronuclei frequency, as well as abnormal mitotic events (over 40% in treated cells, p < 0.05). Time-course studies were performed using a mouse nephrotoxicity model (cumulative 72 mg/kg). Kidneys were collected over 48 h and investigated for histopathology and DNA damage. Notable increases in γH2AX foci (indicative of double-stranded breaks) were observed in both cell culture (up to ~ 44% cells with 5+ foci at 24 h, p < 0.05) and mice treated with polymyxin B (up to ~ 25%, p < 0.05). Consistent with these results, in vitro assays showed high binding affinity of polymyxin B to DNA. Together, our results indicate that polymyxin B nephrotoxicity is associated with DNA damage, leading to chromosome missegregation and genome instability. This novel mechanistic information may lead to new strategies to overcome the nephrotoxicity of this important last-line class of antibiotics.
Collapse
|
42
|
Nagarajan D, Nagarajan T, Roy N, Kulkarni O, Ravichandran S, Mishra M, Chakravortty D, Chandra N. Computational antimicrobial peptide design and evaluation against multidrug-resistant clinical isolates of bacteria. J Biol Chem 2018; 293:3492-3509. [PMID: 29259134 PMCID: PMC5846155 DOI: 10.1074/jbc.m117.805499] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/04/2017] [Indexed: 12/19/2022] Open
Abstract
There is a pressing need for new therapeutics to combat multidrug- and carbapenem-resistant bacterial pathogens. This challenge prompted us to use a long short-term memory (LSTM) language model to understand the underlying grammar, i.e. the arrangement and frequencies of amino acid residues, in known antimicrobial peptide sequences. According to the output of our LSTM network, we synthesized 10 peptides and tested them against known bacterial pathogens. All of these peptides displayed broad-spectrum antimicrobial activity, validating our LSTM-based peptide design approach. Our two most effective antimicrobial peptides displayed activity against multidrug-resistant clinical isolates of Escherichia coli, Acinetobacter baumannii, Klebsiella pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus, and coagulase-negative staphylococci strains. High activity against extended-spectrum β-lactamase, methicillin-resistant S. aureus, and carbapenem-resistant strains was also observed. Our peptides selectively interacted with and disrupted bacterial cell membranes and caused secondary gene-regulatory effects. Initial structural characterization revealed that our most effective peptide appeared to be well folded. We conclude that our LSTM-based peptide design approach appears to have correctly deciphered the underlying grammar of antimicrobial peptide sequences, as demonstrated by the experimentally observed efficacy of our designed peptides.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, and
- Centre for Biosystems Science and Engineering, Indian Institute of Science (IISc), Bangalore 560012, India
| | - Nagasuma Chandra
- From the Departments of Biochemistry and
- Centre for Biosystems Science and Engineering, Indian Institute of Science (IISc), Bangalore 560012, India
| |
Collapse
|
43
|
Vaara M. New polymyxin derivatives that display improved efficacy in animal infection models as compared to polymyxin B and colistin. Med Res Rev 2018; 38:1661-1673. [PMID: 29485690 DOI: 10.1002/med.21494] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/24/2018] [Accepted: 02/01/2018] [Indexed: 01/01/2023]
Abstract
Polymyxin B and colistin (polymyxin E) are bactericidal pentacationic lipopeptides that act specifically on Gram-negative bacteria, first by disrupting their outermost permeability barrier, the outer membrane (OM), and then damaging the cytoplasmic membrane. The discovery of both polymyxin B and colistin was published independently by three laboratories as early as in 1947. They were subsequently used in intravenous therapy. Unfortunately, they also exhibit significant and dose-limiting nephrotoxicity. Therefore, polymyxins were reserved as agents of last-line defense. The emergence of extremely multiresistant strains has now forced clinicians to reinstate polymyxins in the therapy of severe infections. However, the current dosage regimens lead to insufficient drug concentrations in serum and clinicians have been advised to use larger doses, which further increases the risk of nephrotoxicity. Very recently, the interest in developing better tolerated and more effective polymyxins has grown. This review focuses on describing four development programs that have yielded novel derivatives that are more effective than the old polymyxins in animal infection models. Compounds from three programs are superior to the old polymyxins in the rodent lung infection model with Acinetobacter baumannii and/or Pseudomonas aeruginosa. One of them is also more effective than polymyxin B in A. baumannii mouse thigh infection. The fourth program includes compounds that are approximately tenfold more effective in Escherichia coli murine pyelonephritis than polymyxin B.
Collapse
Affiliation(s)
- Martti Vaara
- Northern Antibiotics Ltd., Espoo, Finland.,Department of Bacteriology and Immunology, Helsinki University Medical School, Helsinki, Finland
| |
Collapse
|
44
|
Larki RA, Jamali B, Meidani M, Mousavi S. Serum Cystatin C for Evaluation of Acute Kidney Injury in Adults Treated with Colistin. J Res Pharm Pract 2018; 7:178-181. [PMID: 30622984 PMCID: PMC6298137 DOI: 10.4103/jrpp.jrpp_18_53] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Objective Recent studies have shown that serum cystatin C (Cys C) is a better marker for measuring the glomerular filtration rate and may rise more quickly with acute kidney injury (AKI). The purpose of this study was to evaluate the clinical application of serum Cys C to predict colistin-induced nephrotoxicity in comparison with serum creatinine (SCr). Methods Thirty-two adult patients with no history of acute or chronic kidney injury having been planned to receive intravenous colistin for an anticipated duration of at least 1 week for any indication were recruited. At baseline and 5 days after colistin treatment, serum Cys C as well as creatinine levels were measured. The incidence of colistin-induced acute renal failure was defined according to the AKIN criteria for SCr. Rise in concentration of Cys C by more than 10% from baseline considered as AKI. Findings Colistin-induced nephrotoxicity (defined as SCr ≥0.3 mg/dl) occurred in 6 patients (18.8%). A Cys C increase concentration ≥10% after 5 days of colistin treatment was detected in 15 patients (46.9%). There was a poor agreement between the presence and absence of any SCr-AKI and Cys C-AKI (κ = 0.28, P = 0.04). Conclusion Serum Cys C is a better marker of renal function in early stages of AKI and predictive of persistent AKI on colistin treatment.
Collapse
Affiliation(s)
- Rozina Abbasi Larki
- Department of Internal Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Bahareh Jamali
- Department of Internal Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohsen Meidani
- Department of Infectious Diseases, Al-Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sarah Mousavi
- Department of Clinical Pharmacy and Pharmacy Practice, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
45
|
Kim EJ, Kim ES. Exploring New Predictors of Colistin-Associated Nephrotoxicity. Infect Chemother 2018; 50:283-285. [PMID: 30270590 PMCID: PMC6167514 DOI: 10.3947/ic.2018.50.3.283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Indexed: 11/24/2022] Open
Affiliation(s)
- Eun Jung Kim
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Eu Suk Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Araujo M, Doi SQ. Editorial: Biomarkers in CKD. Front Med (Lausanne) 2017; 4:168. [PMID: 29085823 PMCID: PMC5649135 DOI: 10.3389/fmed.2017.00168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/25/2017] [Indexed: 11/20/2022] Open
Affiliation(s)
- Magali Araujo
- Department of Medicine, Georgetown University, Washington, DC, United States
| | - Sonia Q Doi
- Department of Medicine, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
47
|
Evaluation of Urinary KIM-1 for Prediction of Polymyxin B-Induced Nephrotoxicity. Antimicrob Agents Chemother 2017; 61:AAC.01735-17. [PMID: 28848003 DOI: 10.1128/aac.01735-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
48
|
Corona-Villalobos CP, Shlipak MG, Tin A, Parikh C, Moore RD, Vittinghoff E, Monroy-Trujillo JM, Atta MG, Estrella MM. Predictors of Acute Renal Injury Study (PARIS) among HIV-positive individuals: design and methods. BMC Nephrol 2017; 18:289. [PMID: 28882126 PMCID: PMC5590192 DOI: 10.1186/s12882-017-0696-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/22/2017] [Indexed: 12/29/2022] Open
Abstract
Background Acute kidney injury (AKI), which is common among HIV-positive individuals, may contribute to the excess burden of chronic kidney disease (CKD) in this patient population; however, conventional clinical methods to detect AKI do not capture kidney injury sufficiently early to prevent irreversible damage. Further, large observational and interventional studies of AKI generally exclude HIV-positive persons in spite of their disproportionate risk. Methods The Predictors of Acute Renal Injury Study (PARIS) is a prospective observational cohort study among HIV-positive individuals established to determine the ability of candidate kidney injury biomarkers to predict future hospitalized clinical AKI, to characterize hospitalized subclinical AKI, and to discern the risk of progressive kidney disease following subclinical and clinical AKI. Among the candidate kidney injury markers, we will select the most promising to translate into a clinically viable, multiplex panel of urinary biomarkers which we will integrate with clinical factors to develop a model prognostic of risks for AKI and subsequent kidney function decline. This study has a targeted enrollment of 2000 participants. The overall follow-up of participants consists of two phases: 1) a 5-year active follow-up phase which involves serial evaluations at enrollment, annual clinic visits, and among participants who are hospitalized during this period, an evaluation at index hospitalization and 3 and 12 months post-hospitalization; and 2) a subsequent passive follow-up phase for the duration that the participant receives medical care at The Johns Hopkins Hospital. Discussions This study will serve as an important resource for future studies of AKI by establishing a repository with both ambulatory and inpatient biospecimens, a resource that is currently lacking in existing HIV clinical cohorts and studies of AKI. Upon completion of this study, the resulting prognostic model which will incorporate results from the multiplex HIV-AKI Risk Pane could serve as a pharmacodynamic endpoint for early phase therapeutic candidates for AKI.
Collapse
Affiliation(s)
- Celia P Corona-Villalobos
- Department of Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, 21287, MD, USA
| | - Michael G Shlipak
- San Francisco and San Francisco VA Health Care System, Kidney Health Research Collaborative, University of California, 1450 Clement St., 111A1, San Francisco, 94121, CA, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Room W6017, Baltimore, 21287, MD, USA
| | - Chirag Parikh
- Department of Medicine, Division of Nephrology, Yale University School of Medicine, 60 Temple St., Suite 6C, New Haven, 06510, CT, USA
| | - Richard D Moore
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Room W6017, Baltimore, 21287, MD, USA.,Department of Medicine, Division of General Internal Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St., Suite 8059, Baltimore, 21287, MD, USA
| | - Eric Vittinghoff
- San Francisco and San Francisco VA Health Care System, Kidney Health Research Collaborative, University of California, 1450 Clement St., 111A1, San Francisco, 94121, CA, USA
| | - Jose Manuel Monroy-Trujillo
- Department of Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, 21287, MD, USA
| | - Mohamed G Atta
- Department of Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, 21287, MD, USA
| | - Michelle M Estrella
- San Francisco and San Francisco VA Health Care System, Kidney Health Research Collaborative, University of California, 1450 Clement St., 111A1, San Francisco, 94121, CA, USA.
| |
Collapse
|
49
|
Rabanal F, Cajal Y. Recent advances and perspectives in the design and development of polymyxins. Nat Prod Rep 2017. [PMID: 28628170 DOI: 10.1039/c7np00023e] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Covering: 1947-early 2017, particularly from 2005-early 2017The rise of bacterial pathogens with acquired resistance to almost all available antibiotics is becoming a serious public health issue. Polymyxins, antibiotics that were mostly abandoned a few decades ago because of toxicity concerns, are ultimately considered as a last-line therapy to treat infections caused by multi-drug resistant Gram-negative bacteria. This review surveys the progress in understanding polymyxin structure, and their chemistry, mechanisms of antibacterial activity and nephrotoxicity, biomarkers, synergy and combination with other antimicrobial agents and antibiofilm properties. An update of recent efforts in the design and development of a new generation of polymyxin drugs is also discussed. A novel approach considering the modification of the scaffold of polymyxins to integrate metabolism and detoxification issues into the drug design process is a promising new line to potentially prevent accumulation in the kidneys and reduce nephrotoxicity.
Collapse
Affiliation(s)
- Francesc Rabanal
- Organic Chemistry Section, Department of Inorganic and Organic Chemistry, Faculty of Chemistry, University of Barcelona, Spain.
| | | |
Collapse
|
50
|
Role of Renal Drug Exposure in Polymyxin B-Induced Nephrotoxicity. Antimicrob Agents Chemother 2017; 61:AAC.02391-16. [PMID: 28096166 DOI: 10.1128/aac.02391-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/12/2017] [Indexed: 12/13/2022] Open
Abstract
Despite dose-limiting nephrotoxic potentials, polymyxin B has reemerged as the last line of therapy against multidrug-resistant Gram-negative bacterial infections. However, the handling of polymyxin B by the kidneys is still not thoroughly understood. The objectives of this study were to evaluate the impact of renal polymyxin B exposure on nephrotoxicity and to explore the role of megalin in renal drug accumulation. Sprague-Dawley rats (225 to 250 g) were divided into three dosing groups, and polymyxin B was administered (5 mg/kg, 10 mg/kg, and 20 mg/kg) subcutaneously once daily. The onset of nephrotoxicity over 7 days and renal drug concentrations 24 h after the first dose were assessed. The effects of sodium maleate (400 mg/kg intraperitoneally) on megalin homeostasis were evaluated by determining the urinary megalin concentration and electron microscopic study of renal tissue. The serum/renal pharmacokinetics of polymyxin B were assessed in megalin-shedding rats. The onset of nephrotoxicity was correlated with the daily dose of polymyxin B. Renal polymyxin B concentrations were found to be 3.6 ± 0.4 μg/g, 9.9 ± 1.5 μg/g, and 21.7 ± 4.8 μg/g in the 5-mg/kg, 10-mg/kg, and 20-mg/kg dosing groups, respectively. In megalin-shedding rats, the serum pharmacokinetics of polymyxin B remained unchanged, but the renal exposure was attenuated by 40% compared to that of control rats. The onset of polymyxin B-induced nephrotoxicity is correlated with the renal drug exposure. In addition, megalin appears to play a pivotal role in the renal accumulation of polymyxin B, which might contribute to nephrotoxicity.
Collapse
|