1
|
Huang Y, Truelson KA, Stewart IA, O'Doherty GA, Kirby JE. Enhanced Activity of Apramycin and Apramycin-Based Combinations Against Mycobacteroides abscessus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.09.648020. [PMID: 40391321 PMCID: PMC12087987 DOI: 10.1101/2025.04.09.648020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Background Mycobacteroides abscessus are rapidly growing non-tuberculous mycobacteria that cause chronic lung and soft tissue infections. Treatment options are often severely limited due to intrinsic resistance to most antimicrobials. Amikacin has historically been a mainstay of combination treatment regimens. However, irreversible hearing loss and vestibular toxicity have led to a search for alternative agents. Apramycin is a novel aminoglycoside currently in phase I clinical trials that may offer lower potential for ototoxic and renal toxic side effects. Objectives The goal of this study was to compare apramycin's in vitro activity with amikacin and other aminoglycosides against a large collection of M. abscessus clinical isolates, both alone and in combination with clofazimine or linezolid. We also tested the activity of apramycin against a more limited collection of other species of rapidly growing mycobacteria. Methods Analysis was performed using reference broth microdilution minimal inhibitory concentration testing, inkjet printer-assisted checkerboard assays, and time-kill assays. Results Against M. abscessus , the MIC 50/90 for apramycin (2 µg/mL) was 8-fold lower than for amikacin (16 µg/mL). Plazomicin was inactive, and organisms were rarely susceptible to tobramycin. Synergy was not detected by checkerboard assay. In time-kill studies, clofazimine modestly potentiated activity of apramycin and. to a lesser extent, amikacin. Apramycin and amikacin showed delayed bacterial killing that either achieved or approached a bactericidal threshold. Apramycin was similarly potent against other rapidly growing mycobacteria tested. Conclusions Apramycin exhibits more potent in vitro activity against a diverse set of M. abscessus and other rapidly growing mycobacteria than approved aminoglycosides.
Collapse
|
2
|
Hernández-Lozano I, Aranzana-Climent V, Cao S, Matias C, Ulf Hansen J, Liepinsh E, Hughes D, Hobbie SN, Vingsbo Lundberg C, Friberg LE. Model-informed drug development for antimicrobials: translational pharmacokinetic-pharmacodynamic modelling of apramycin to facilitate prediction of efficacious dose in complicated urinary tract infections. J Antimicrob Chemother 2025; 80:301-310. [PMID: 39548844 PMCID: PMC11695905 DOI: 10.1093/jac/dkae409] [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: 05/13/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024] Open
Abstract
OBJECTIVES The use of mouse models of complicated urinary tract infection (cUTI) has usually been limited to a single timepoint assessment of bacterial burden. Based on longitudinal in vitro and in vivo data, we developed a pharmacokinetic-pharmacodynamic (PKPD) model to assess the efficacy of apramycin, a broad-spectrum aminoglycoside antibiotic, in mouse models of cUTI. METHODS Two Escherichia coli strains were studied (EN591 and ATCC 700336). Apramycin exposure-effect relationships were established with in vitro time-kill data at pH 6 and pH 7.4 and in mice with cUTI. Immunocompetent mice were treated with apramycin (1.5-30 mg/kg) starting 24 h post-infection. Kidney and bladder tissue were collected 6-96 h post-infection for cfu determination. A PKPD model integrating all data was developed and simulations were performed to predict bacterial burden in humans. RESULTS Treatment with apramycin reduced the bacterial load in kidneys and bladder tissue up to 4.3-log compared with vehicle control. In vitro and in vivo tissue time-course efficacy data were integrated into the PKPD model, showing 76%-98% reduction of bacterial net growth and 3- to 145-fold increase in apramycin potency in vivo compared with in vitro. Simulations suggested that an 11 mg/kg daily dose would be sufficient to achieve bacterial stasis in kidneys and bladder in humans. CONCLUSIONS PKPD modelling with in vitro and in vivo PK and PD data enabled simultaneous evaluation of the different components that influence drug effect, an approach that had not yet been evaluated for antibiotics in the cUTI model and that has potential to enhance model-informed drug development of antibiotics.
Collapse
Affiliation(s)
| | - Vincent Aranzana-Climent
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
- PHAR2, Inserm U1070, Université de Poitiers, Poitiers, France
| | - Sha Cao
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Carina Matias
- Department of Bacteria, Parasites & Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Jon Ulf Hansen
- Department of Bacteria, Parasites & Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Edgars Liepinsh
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Diarmaid Hughes
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sven N Hobbie
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
- Division of Clinical Bacteriology and Mycology, University Hospital Basel, Basel, Switzerland
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
Loffredo MR, Cappiello F, Cappella G, Capuozzo E, Torrini L, Diaco F, Di YP, Mangoni ML, Casciaro B. The pH-Insensitive Antimicrobial and Antibiofilm Activities of the Frog Skin Derived Peptide Esc(1-21): Promising Features for Novel Anti-Infective Drugs. Antibiotics (Basel) 2024; 13:701. [PMID: 39200001 PMCID: PMC11350779 DOI: 10.3390/antibiotics13080701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024] Open
Abstract
The number of antibiotic-resistant microbial infections is dramatically increasing, while the discovery of new antibiotics is significantly declining. Furthermore, the activity of antibiotics is negatively influenced by the ability of bacteria to form sessile communities, called biofilms, and by the microenvironment of the infection, characterized by an acidic pH, especially in the lungs of patients suffering from cystic fibrosis (CF). Antimicrobial peptides represent interesting alternatives to conventional antibiotics, and with expanding properties. Here, we explored the effects of an acidic pH on the antimicrobial and antibiofilm activities of the AMP Esc(1-21) and we found that it slightly lost activity (from 2- to 4-fold) against the planktonic form of a panel of Gram-negative bacteria, with respect to a ≥ 32-fold of traditional antibiotics. Furthermore, it retained its activity against the sessile form of these bacteria grown in media with a neutral pH, and showed similar or higher effectiveness against the biofilm form of bacteria grown in acidic media, simulating a CF-like acidic microenvironment, compared to physiological conditions.
Collapse
Affiliation(s)
- Maria Rosa Loffredo
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.C.); (E.C.); (B.C.)
| | - Floriana Cappiello
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.C.); (E.C.); (B.C.)
| | - Giacomo Cappella
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.C.); (E.C.); (B.C.)
| | - Elisabetta Capuozzo
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.C.); (E.C.); (B.C.)
| | - Luisa Torrini
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (L.T.); (F.D.)
| | - Fabiana Diaco
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (L.T.); (F.D.)
| | - Yuanpu Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Maria Luisa Mangoni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.C.); (E.C.); (B.C.)
| | - Bruno Casciaro
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.C.); (E.C.); (B.C.)
| |
Collapse
|
4
|
Sun Q, Yan J, Long S, Shi Y, Jiang G, Li H, Huang H, Wang G. Apramycin has high in vitro activity against Mycobacterium tuberculosis. J Med Microbiol 2024; 73. [PMID: 38973691 DOI: 10.1099/jmm.0.001854] [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] [Indexed: 07/09/2024] Open
Abstract
Introduction. Aminoglycoside antibiotics such as amikacin and kanamycin are important components in the treatment of Mycobacterium tuberculosis (Mtb) infection. However, more and more clinical strains are found to be aminoglycoside antibiotic-resistant. Apramycin is another kind of aminoglycoside antibiotic that is commonly used to treat infections in animals.Hypothesis. Apramycin may have in vitro activity against Mtb.Aim. This study aims to evaluate the efficacy of apramycin against Mtb in vitro and determine its epidemiological cut-off (ECOFF) value.Methodology. One hundred Mtb isolates, including 17 pansusceptible and 83 drug-resistant tuberculosis (DR-TB) strains, were analysed for apramycin resistance using the MIC assay.Results. Apramycin exhibited significant inhibitory activity against Mtb clinical isolates, with an MIC50 of 0.5 μg ml-1 and an MIC90 of 1 μg ml-1. We determined the tentative ECOFF value as 1 µg ml-1 for apramycin. The resistant rates of multidrug-resistant tuberculosis (MDR-TB), pre-extensively drug-resistant (pre-XDR-TB) and extensively drug-resistant tuberculosis (XDR-TB) strains were 12.12 % (4/33), 20.69 % (6/29) and 66.67 % (14/21), respectively. The rrs gene A1401G is associated with apramycin resistance, as well as the cross-resistance between apramycin and other aminoglycosides.Conclusion. Apramycin shows high in vitro activity against the Mtb clinical isolates, especially the MDR-TB clinical isolates. This encouraging discovery calls for more research on the functions of apramycin in vivo and as a possible antibiotic for the treatment of drug-resistant TB.
Collapse
Affiliation(s)
- Qing Sun
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, PR China
| | - Jun Yan
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| | - Sibo Long
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| | - Yiheng Shi
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| | - Guanglu Jiang
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, PR China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, PR China
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, PR China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, PR China
| | - Guirong Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| |
Collapse
|
5
|
Frimodt-Møller N, Hansen JU, Plattner M, Huseby DL, Radmer Almind S, Haldimann K, Gysin M, Petersson A, Ercan O, Ganz L, Hughes D, Vingsbo Lundberg C, Hobbie SN. Apramycin efficacy against carbapenem- and aminoglycoside-resistant Escherichia coli and Klebsiella pneumoniae in murine bloodstream infection models. Int J Antimicrob Agents 2024; 64:107181. [PMID: 38653351 DOI: 10.1016/j.ijantimicag.2024.107181] [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: 06/02/2023] [Revised: 03/06/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND The aminoglycoside apramycin has been proposed as a drug candidate for the treatment of critical Gram-negative systemic infections. However, the potential of apramycin in the treatment of drug-resistant bloodstream infections (BSIs) has not yet been assessed. METHODS The resistance gene annotations of 40 888 blood-culture isolates were analysed. In vitro profiling of apramycin comprised cell-free translation assays, broth microdilution, and frequency of resistance determination. The efficacy of apramycin was studied in a mouse peritonitis model for a total of nine Escherichia coli and Klebsiella pneumoniae isolates. RESULTS Genotypic aminoglycoside resistance was identified in 87.8% of all 6973 carbapenem-resistant Enterobacterales blood-culture isolates, colistin resistance was shown in 46.4% and apramycin in 2.1%. Apramycin activity against methylated ribosomes was > 100-fold higher than that for other aminoglycosides. Frequencies of resistance were < 10-9 at 8 × minimum inhibitory concentration (MIC). Tentative epidemiological cut-offs (TECOFFs) were determined as 8 µg/mL for E. coli and 4 µg/mL for K. pneumoniae. A single dose of 5 to 13 mg/kg resulted in a 1-log colony-forming unit (CFU) reduction in the blood and peritoneum. Two doses of 80 mg/kg resulted in an exposure that resembles the AUC observed for a single 30 mg/kg dose in humans and led to complete eradication of carbapenem- and aminoglycoside-resistant bacteraemia. CONCLUSION Encouraging coverage and potent in vivo efficacy against a selection of highly drug-resistant Enterobacterales isolates in the mouse peritonitis model warrants the conduct of clinical studies to validate apramycin as a drug candidate for the prophylaxis and treatment of BSI.
Collapse
Affiliation(s)
| | - Jon U Hansen
- Statens Serum Institute, 2300, Copenhagen, Denmark
| | - Michel Plattner
- University of Zurich, Institute of Medical Microbiology, 8006, Zurich, Switzerland
| | - Douglas L Huseby
- Uppsala University, Department of Medical Biochemistry and Microbiology, 75237, Uppsala, Sweden
| | - Stine Radmer Almind
- Rigshospitalet, Department of Clinical Microbiology, 2100 Copenhagen, Denmark
| | - Klara Haldimann
- University of Zurich, Institute of Medical Microbiology, 8006, Zurich, Switzerland
| | - Marina Gysin
- University of Zurich, Institute of Medical Microbiology, 8006, Zurich, Switzerland
| | - Anna Petersson
- Uppsala University, Department of Medical Biochemistry and Microbiology, 75237, Uppsala, Sweden
| | - Onur Ercan
- Uppsala University, Department of Medical Biochemistry and Microbiology, 75237, Uppsala, Sweden
| | - Lea Ganz
- University of Zurich, Institute of Medical Microbiology, 8006, Zurich, Switzerland
| | - Diarmaid Hughes
- Uppsala University, Department of Medical Biochemistry and Microbiology, 75237, Uppsala, Sweden
| | | | - Sven N Hobbie
- University of Zurich, Institute of Medical Microbiology, 8006, Zurich, Switzerland; University Hospital Basel, Division of Clinical Bacteriology, 4031, Basel, Switzerland.
| |
Collapse
|
6
|
Gross S, Herren S, Gysin M, Rominski A, Roditscheff A, Risch M, Imkamp F, Crich D, Hobbie SN. In vitro susceptibility of Neisseria gonorrhoeae to netilmicin and etimicin in comparison to gentamicin and other aminoglycosides. Eur J Clin Microbiol Infect Dis 2024; 43:821-828. [PMID: 38388739 PMCID: PMC11108870 DOI: 10.1007/s10096-024-04782-2] [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: 11/15/2023] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
PURPOSE Single doses of gentamicin have demonstrated clinical efficacy in the treatment of urogenital gonorrhea, but lower cure rates for oropharyngeal and anorectal gonorrhea. Formulations selectively enriched in specific gentamicin C congeners have been proposed as a less toxic alternative to gentamicin, potentially permitting higher dosing to result in increased plasma exposures at the extragenital sites of infection. The purpose of the present study was to compare the antibacterial activity of individual gentamicin C congeners against Neisseria gonorrhoeae to that of other aminoglycoside antibiotics. METHODS Antimicrobial susceptibility of three N. gonorrhoeae reference strains and 152 clinical isolates was assessed using standard disk diffusion, agar dilution, and epsilometer tests. RESULTS Gentamicin C1, C2, C1a, and C2a demonstrated similar activity against N. gonorrhoeae. Interestingly, susceptibility to the 1-N-ethylated aminoglycosides etimicin and netilmicin was significantly higher than the susceptibility to their parent compounds gentamicin C1a and sisomicin, and to any other of the 25 aminoglycosides assessed in this study. Propylamycin, a 4'-propylated paromomycin analogue, was significantly more active against N. gonorrhoeae than its parent compound, too. CONCLUSION Selectively enriched gentamicin formulations hold promise for a less toxic but equally efficacious alternative to gentamicin. Our study warrants additional consideration of the clinically established netilmicin and etimicin for treatment of genital and perhaps extragenital gonorrhea. Additional studies are required to elucidate the mechanism behind the advantage of alkylated aminoglycosides.
Collapse
Affiliation(s)
- Sonja Gross
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006, Zurich, Switzerland
| | - Sebastian Herren
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006, Zurich, Switzerland
| | - Marina Gysin
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006, Zurich, Switzerland
| | - Anna Rominski
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006, Zurich, Switzerland
| | - Anna Roditscheff
- Private University in the Principality of Liechtenstein, Dorfstrasse 24, 9495, Triesen, Liechtenstein
- Dr. Risch Medical Laboratory, Waldeggstrasse 37, 3097, Liebefeld, Switzerland
| | - Martin Risch
- Private University in the Principality of Liechtenstein, Dorfstrasse 24, 9495, Triesen, Liechtenstein
- Dr. Risch Medical Laboratory, Waldeggstrasse 37, 3097, Liebefeld, Switzerland
| | - Frank Imkamp
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006, Zurich, Switzerland
| | - David Crich
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, GA, 30602, USA
| | - Sven N Hobbie
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006, Zurich, Switzerland.
- Division of Clinical Bacteriology and Mycology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland.
| |
Collapse
|
7
|
Lang M, Carvalho A, Baharoglu Z, Mazel D. Aminoglycoside uptake, stress, and potentiation in Gram-negative bacteria: new therapies with old molecules. Microbiol Mol Biol Rev 2023; 87:e0003622. [PMID: 38047635 PMCID: PMC10732077 DOI: 10.1128/mmbr.00036-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023] Open
Abstract
SUMMARYAminoglycosides (AGs) are long-known molecules successfully used against Gram-negative pathogens. While their use declined with the discovery of new antibiotics, they are now classified as critically important molecules because of their effectiveness against multidrug-resistant bacteria. While they can efficiently cross the Gram-negative envelope, the mechanism of AG entry is still incompletely understood, although this comprehension is essential for the development of new therapies in the face of the alarming increase in antibiotic resistance. Increasing antibiotic uptake in bacteria is one strategy to enhance effective treatments. This review aims, first, to consolidate old and recent knowledge about AG uptake; second, to explore the connection between AG-dependent bacterial stress and drug uptake; and finally, to present new strategies of potentiation of AG uptake for more efficient antibiotic therapies. In particular, we emphasize on the connection between sugar transport and AG potentiation.
Collapse
Affiliation(s)
- Manon Lang
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - André Carvalho
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
8
|
Atlas N, Uzair B, Movellan J, Gracia R, Dupin D, Loinaz I, van Nostrum CF, Hays JP. In vitro activity of novel apramycin-dextran nanoparticles and free apramycin against selected Dutch and Pakistani Klebsiella pneumonia isolates. Heliyon 2023; 9:e22821. [PMID: 38125473 PMCID: PMC10730580 DOI: 10.1016/j.heliyon.2023.e22821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Klebsiella pneumoniae are bacteria associated with respiratory tract infections and are increasingly becoming resistant to antibiotics, including carbapenems. Apramycin is a veterinary antibiotic that may have the potential to be re-purposed for use in human health, for example, for the treatment of respiratory tract infections after coupling to inhalable nanoparticles. In the present study, the antibiotic apramycin was formulated with single chain polymeric nanoparticles and tested in free and formulated forms against a set of 13 Klebsiella pneumoniae isolates (from the Netherlands and Pakistan) expressing different aminoglycoside resistance phenotypes. Minimum Inhibitory Concentration, Time Kill Kinetics and biofilm experiments were performed providing evidence for the potential efficacy of apramycin and apramycin-based nanomedicines for the treatment of human Klebsiella pneumonia infections.
Collapse
Affiliation(s)
- Nagina Atlas
- Dept. Biological Science, International Islamic University Islamabad, Pakistan
- Utrecht Institute for Pharmaceutical Sciences, Dept. of Pharmaceutics, Utrecht University, Utrecht, the Netherlands
| | - Bushra Uzair
- Dept. Biological Science, International Islamic University Islamabad, Pakistan
| | - Julie Movellan
- CIDETEC, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Gipuzkoa, Miramon Pasealekua, 196, Donostia-San Sebastián 20014, Spain
| | - Raquel Gracia
- CIDETEC, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Gipuzkoa, Miramon Pasealekua, 196, Donostia-San Sebastián 20014, Spain
| | - Damien Dupin
- CIDETEC, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Gipuzkoa, Miramon Pasealekua, 196, Donostia-San Sebastián 20014, Spain
| | - Iraida Loinaz
- CIDETEC, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Gipuzkoa, Miramon Pasealekua, 196, Donostia-San Sebastián 20014, Spain
| | - Cornelus F. van Nostrum
- Utrecht Institute for Pharmaceutical Sciences, Dept. of Pharmaceutics, Utrecht University, Utrecht, the Netherlands
| | - John P. Hays
- Dept. Medical Microbiology & Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), Rotterdam, the Netherlands
| |
Collapse
|
9
|
Mohamad-Ramshan R, Ande C, Matsushita T, Haldimann K, Vasella A, Hobbie SN, Crich D. Synthesis of 4- O-(4-Amino-4-deoxy-β-D-xylopyranosyl)paromomycin and 4- S-(β-D-Xylopyranosyl)-4-deoxy-4'-thio-paromomycin and Evaluation of their Antiribosomal and Antibacterial Activity. Tetrahedron 2023; 135:133330. [PMID: 37035443 PMCID: PMC10081503 DOI: 10.1016/j.tet.2023.133330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The design, synthesis and antiribosomal and antibacterial activity of two novel glycosides of the aminoglycoside antibiotic paromomycin are described. The first carries of 4-amino-4-deoxy-β-D-xylopyranosyl moiety at the paromomycin 4'-position and is approximately two-fold more active than the corresponding β-D-xylopyranosyl derivative. The second is a 4'-(β-D-xylopyranosylthio) derivative of 4'-deoxyparomomycin that is unexpectedly less active than the simple β-D-xylopyranosyl derivative, perhaps because of the insertion of the conformationally more mobile thioglycosidic linkage.
Collapse
Affiliation(s)
| | - Chennaiah Ande
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, GA 30602, USA
| | - Takahiko Matsushita
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA
| | - Klara Haldimann
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006 Zürich, Switzerland
| | - Andrea Vasella
- Organic Chemistry Laboratory, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Sven N Hobbie
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006 Zürich, Switzerland
| | - David Crich
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, GA 30602, USA
- Department of Chemistry, University of Georgia, 302 East Campus Road, Athens, GA 30602, USA
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| |
Collapse
|
10
|
Otsuka Y, Umemura E, Takamiya Y, Ishibashi T, Hayashi C, Yamada K, Igarashi M, Shibasaki M, Takahashi Y. Aprosamine Derivatives Active against Multidrug-Resistant Gram-Negative Bacteria. ACS Infect Dis 2023; 9:886-898. [PMID: 36893496 DOI: 10.1021/acsinfecdis.2c00557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Novel aprosamine derivatives were synthesized for the development of aminoglycoside antibiotics active against multidrug-resistant Gram-negative bacteria. The synthesis of aprosamine derivatives involved glycosylation at the C-8' position and subsequent modification (epimerization and deoxygenation at the C-5 position and 1-N-acylation) of the 2-deoxystreptamine moiety. All 8'-β-glycosylated aprosamine derivatives (3a-h) showed excellent antibacterial activity against carbapenem-resistant Enterobacteriaceae and 16S ribosomal RNA methyltransferase-producing multidrug-resistant Gram-negative bacteria compared to the clinical drug, arbekacin. The antibacterial activity of 5-epi (6a-d) and 5-deoxy derivatives (8a,b and 8h) of β-glycosylated aprosamine was further enhanced. On the other hand, the derivatives (10a,b and 10h) in which the amino group at the C-1 position was acylated with (S)-4-amino-2-hydroxybutyric acid showed excellent activity (MICs 0.25-0.5 μg/mL) against resistant bacteria that produce the aminoglycoside-modifying enzyme, aminoglycoside 3-N-acetyltransferase IV, which induces high resistance against parent apramycin (MIC > 64 μg/mL). In particular, 8b and 8h showed approximately 2- to 8-fold antibacterial activity against carbapenem-resistant Enterobacteriaceae and 8- to 16-fold antibacterial activity against resistant Gram-positive bacteria, such as methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci, compared to apramycin. Our results showed that aprosamine derivatives have immense potential in the development of therapeutic agents for multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Yasunari Otsuka
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Eijiro Umemura
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Yukimi Takamiya
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Teruhisa Ishibashi
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Chigusa Hayashi
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Keiko Yamada
- Pharmaceutical Analysis Laboratories, Pharmaceutical Research Center, Meiji Seika Pharma Co., Ltd., 788 Kayama, Odawara-shi 250-0852, Kanagawa, Japan
| | - Masayuki Igarashi
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Masakatsu Shibasaki
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Yoshiaki Takahashi
- Institute of Microbial Chemistry (BIKAKEN), Tokyo 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| |
Collapse
|
11
|
Caméléna F, Liberge M, Rezzoug I, Merimèche M, Naas T, Berçot B. In vitro activity of apramycin against 16S-RMTase-producing Gram-negative isolates. J Glob Antimicrob Resist 2023; 33:21-25. [PMID: 36822368 DOI: 10.1016/j.jgar.2023.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/10/2023] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
OBJECTIVES Apramycin is an aminoglycoside (AG) with a unique structure that is little affected by plasmid-mediated mechanisms of AG resistance, including most AG-modifying enzymes and 16S rRNA methyltransferases (16S-RMTases). We evaluate the activity of apramycin against a collection of 16S-RMTase-producing isolates, including Enterobacterales, non-fermenting bacteria, and carbapenemase producers. METHODS In total, 164 non-duplicate 16S-RMTase-producing isolates, including 84 Enterobacterales, 53 Acinetobacter baumannii and 27 Pseudomonas aeruginosa isolates, were included in the study. Whole-genome sequencing (WGS) was performed on all isolates with Illumina technology. The minimum inhibitory concentration (MIC) of apramycin was determined by broth microdilution with customized Sensititre plates (Thermo Fisher Scientific, Dardilly, France). RESULTS We found that 95% (156/164) of the 16S-RMTase-producing isolates were susceptible to apramycin, with a MIC50 of 4 mg/L and a MIC90 of 16 mg/L, respectively. Resistance rates were higher in P. aeruginosa (11%) than in A. baumannii (4%) or Enterobacterales (4%) (P < 0.0001 for each comparison). Eight isolates were resistant to apramycin, including one isolate with an MIC >64 mg/L due to the acquisition of the aac(3)-IV gene. The genetic environment of the aac(3)-IV gene was similar to that in the pAH01-4 plasmid of an Escherichia coli isolate from chicken in China. CONCLUSION Resistance to apramycin remains rare in 16S-RMTase-producing isolates. Apramycin may, therefore, be an interesting alternative treatment for infections caused by 16S-RMTase and carbapenemase producers.
Collapse
Affiliation(s)
- François Caméléna
- Department of Bacteriology, Saint-Louis-Lariboisière Hospital Group, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris Cité, INSERM 1137, IAME, Paris, France
| | - Mathilde Liberge
- Department of Bacteriology, Saint-Louis-Lariboisière Hospital Group, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris Cité, INSERM 1137, IAME, Paris, France
| | - Inès Rezzoug
- Department of Bacteriology, Saint-Louis-Lariboisière Hospital Group, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Manel Merimèche
- Department of Bacteriology, Saint-Louis-Lariboisière Hospital Group, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris Cité, INSERM 1137, IAME, Paris, France
| | - Thierry Naas
- Department of Bacteriology-Hygiene, Bicêtre Hospital, Assistance Publique - Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Université de Paris-Saclay, INSERM 1184, RESIST Unit, Le Kremlin-Bicêtre, France; French National Reference Centre for Antimicrobial Resistance, Le Kremlin-Bicêtre, France
| | - Béatrice Berçot
- Department of Bacteriology, Saint-Louis-Lariboisière Hospital Group, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris Cité, INSERM 1137, IAME, Paris, France.
| |
Collapse
|
12
|
Ammar YA, Micky JA, Aboul-Magd DS, Abd El-Hafez SMA, Hessein SA, Ali AM, Ragab A. Development and radiosterilization of new hydrazono-quinoline hybrids as DNA gyrase and topoisomerase IV inhibitors: Antimicrobial and hemolytic activities against uropathogenic isolates with molecular docking study. Chem Biol Drug Des 2023; 101:245-270. [PMID: 36305722 DOI: 10.1111/cbdd.14154] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 09/23/2022] [Accepted: 10/09/2022] [Indexed: 01/14/2023]
Abstract
This study aimed to synthesize new potent quinoline derivatives based on hydrazone moieties and evaluate their antimicrobial activity. The newly synthesized hydrazono-quinoline derivatives 2, 5a, 9, and 10b showed the highest antimicrobial activity with MIC values ≤1.0 μg/ml against bacteria and ≤8.0 μg/ml against the fungi. Further, these derivatives exhibited bactericidal and fungicidal effects with MBC/MIC and MFC/MIC ratio ≤4. Surprisingly, the most active compounds displayed good inhibition to biofilm formation with MBEC values ranging between (40.0 ± 10.0 - 230.0 ± 31.0) and (67.0 ± 24.0 - 347.0 ± 15.0) μg/ml against Staphylococcus aureus and Pseudomonas aeruginosa, respectively. The hemolytic assays confirmed that the hydrazono-quinoline derivatives are non-toxic with low % lysis values ranging from 4.62% to 14.4% at a 1.0 mg/ml concentration. Besides, compound 5a exhibited the lowest hemolytic activity value of ~4.62%. Furthermore, the study suggests that the hydrazono-quinoline analogs exert their antibacterial activity as dual inhibitors for DNA gyrase and DNA topoisomerase IV enzymes with IC50 values ranging between (4.56 ± 0.3 - 21.67 ± 0.45) and (6.77 ± 0.4 - 20.41 ± 0.32) μM, respectively. Additionally, the recent work advocated that compound 5a showed the reference SAL at the ɣ-radiation dose of 10.0 kGy in the sterilization process without affecting its chemical structure. Finally, the in silico drug-likeness, toxicity properties, and molecular docking simulation were performed. Besides, the result exhibited good oral-bioavailability, lower toxicity prediction, and lower binding energy with good binding mode rather than the positive control.
Collapse
Affiliation(s)
- Yousry A Ammar
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Jehan A Micky
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Dina S Aboul-Magd
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Egypt
| | - Sondos M A Abd El-Hafez
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Sadia A Hessein
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Abeer M Ali
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ahmed Ragab
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
13
|
Lubriks D, Haldimann K, Hobbie SN, Vasella A, Suna E, Crich D. Synthesis, Antibacterial and Antiribosomal Activity of the 3 C-Aminoalkyl Modification in the Ribofuranosyl Ring of Apralogs (5- O-Ribofuranosyl Apramycins). Antibiotics (Basel) 2022; 12:antibiotics12010025. [PMID: 36671225 PMCID: PMC9854789 DOI: 10.3390/antibiotics12010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The synthesis and antiribosomal and antibacterial activity of both anomers of a novel apralog, 5-O-(5-amino-3-C-dimethylaminopropyl-D-ribofuranosyl)apramycin, are reported. Both anomers show excellent activity for the inhibition of bacterial ribosomes and that of MRSA and various wild-type Gram negative pathogens. The new compounds retain activity in the presence of the aminoglycoside phosphoryltransferase aminoglycoside modifying enzymes that act on the primary hydroxy group of typical 4,5-(2-deoxystreptamine)-type aminoglycoside and related apramycin derivatives. Unexpectedly, the two anomers have comparable activity both for the inhibition of bacterial ribosomes and of the various bacterial strains tested.
Collapse
Affiliation(s)
- Dmitrijs Lubriks
- Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia
| | - Klara Haldimann
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006 Zürich, Switzerland
| | - Sven N. Hobbie
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 30, 8006 Zürich, Switzerland
| | - Andrea Vasella
- Organic Chemistry Laboratory, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Edgars Suna
- Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia
- Correspondence: (E.S.); (D.C.); Tel.: +37-16-701-4895 (E.S.); Tel.: +1-706-542-5605 (D.C.)
| | - David Crich
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, GA 30602, USA
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
- Department of Chemistry, University of Georgia, 302 East Campus Road, Athens, GA 30602, USA
- Correspondence: (E.S.); (D.C.); Tel.: +37-16-701-4895 (E.S.); Tel.: +1-706-542-5605 (D.C.)
| |
Collapse
|
14
|
Gysin M, Hon PY, Tan P, Sengduangphachanh A, Simmalavong M, Hinfonthong P, Kaewphanderm N, Pham TD, Nguyen TH, Haldimann K, Becker K, van Doorn HR, Hopkins J, Simpson AJH, Ashley EA, Kesteman T, Tran HH, Vasoo S, Ling CL, Roberts T, Turner P, Hobbie SN. Apramycin susceptibility of multidrug-resistant Gram-negative blood culture isolates in five countries in Southeast Asia. Int J Antimicrob Agents 2022; 60:106659. [PMID: 35988665 DOI: 10.1016/j.ijantimicag.2022.106659] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/21/2022] [Accepted: 08/11/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Bloodstream infections (BSIs) are a leading cause of sepsis, which is a life-threatening condition that significantly contributes to the mortality of bacterial infections. Aminoglycoside antibiotics such as gentamicin or amikacin are essential medicines in the treatment of BSIs, but their clinical efficacy is increasingly being compromised by antimicrobial resistance. The aminoglycoside apramycin has demonstrated preclinical efficacy against aminoglycoside-resistant and multidrug-resistant (MDR) Gram-negative bacilli (GNB) and is currently in clinical development for the treatment of critical systemic infections. METHODS This study collected a panel of 470 MDR GNB isolates from healthcare facilities in Cambodia, Laos, Singapore, Thailand and Vietnam for a multicentre assessment of their antimicrobial susceptibility to apramycin in comparison with other aminoglycosides and colistin by broth microdilution assays. RESULTS Apramycin and amikacin MICs ≤ 16 µg/mL were found for 462 (98.3%) and 408 (86.8%) GNB isolates, respectively. Susceptibility to gentamicin and tobramycin (MIC ≤ 4 µg/mL) was significantly lower at 122 (26.0%) and 101 (21.5%) susceptible isolates, respectively. Of note, all carbapenem and third-generation cephalosporin-resistant Enterobacterales, all Acinetobacter baumannii and all Pseudomonas aeruginosa isolates tested in this study appeared to be susceptible to apramycin. Of the 65 colistin-resistant isolates tested, four (6.2%) had an apramycin MIC > 16 µg/mL. CONCLUSION Apramycin demonstrated best-in-class activity against a panel of GNB isolates with resistances to other aminoglycosides, carbapenems, third-generation cephalosporins and colistin, warranting continued consideration of apramycin as a drug candidate for the treatment of MDR BSIs.
Collapse
Affiliation(s)
- Marina Gysin
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Pei Yun Hon
- National Centre for Infectious Diseases and Tan Tock Seng Hospital, Singapore
| | - Pisey Tan
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, Cambodia
| | - Amphonesavanh Sengduangphachanh
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People's Democratic Republic
| | - Manivone Simmalavong
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People's Democratic Republic
| | - Pattaraporn Hinfonthong
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Napaporn Kaewphanderm
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Thai Duy Pham
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Thanh Ha Nguyen
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Klara Haldimann
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Katja Becker
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - H Rogier van Doorn
- Oxford University Clinical Research Unit, Hanoi, Vietnam; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jill Hopkins
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, Cambodia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew J H Simpson
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People's Democratic Republic; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Elizabeth A Ashley
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People's Democratic Republic; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas Kesteman
- Oxford University Clinical Research Unit, Hanoi, Vietnam; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Hoang Huy Tran
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Shawn Vasoo
- National Centre for Infectious Diseases and Tan Tock Seng Hospital, Singapore
| | - Clare L Ling
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tamalee Roberts
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People's Democratic Republic; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Paul Turner
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, Cambodia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| | - Sven N Hobbie
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
Zhao C, Chirkova A, Rosenborg S, Palma Villar R, Lindberg J, Hobbie SN, Friberg LE. Population pharmacokinetics of apramycin from first-in-human plasma and urine data to support prediction of efficacious dose. J Antimicrob Chemother 2022; 77:2718-2728. [PMID: 35849148 PMCID: PMC9525081 DOI: 10.1093/jac/dkac225] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/15/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Apramycin is under development for human use as EBL-1003, a crystalline free base of apramycin, in face of increasing incidence of multidrug-resistant bacteria. Both toxicity and cross-resistance, commonly seen for other aminoglycosides, appear relatively low owing to its distinct chemical structure. OBJECTIVES To perform a population pharmacokinetic (PPK) analysis and predict an efficacious dose based on data from a first-in-human Phase I trial. METHODS The drug was administered intravenously over 30 min in five ascending-dose groups ranging from 0.3 to 30 mg/kg. Plasma and urine samples were collected from 30 healthy volunteers. PPK model development was performed stepwise and the final model was used for PTA analysis. RESULTS A mammillary four-compartment PPK model, with linear elimination and a renal fractional excretion of 90%, described the data. Apramycin clearance was proportional to the absolute estimated glomerular filtration rate (eGFR). All fixed effect parameters were allometrically scaled to total body weight (TBW). Clearance and steady-state volume of distribution were estimated to 5.5 L/h and 16 L, respectively, for a typical individual with absolute eGFR of 124 mL/min and TBW of 70 kg. PTA analyses demonstrated that the anticipated efficacious dose (30 mg/kg daily, 30 min intravenous infusion) reaches a probability of 96.4% for a free AUC/MIC target of 40, given an MIC of 8 mg/L, in a virtual Phase II patient population with an absolute eGFR extrapolated to 80 mL/min. CONCLUSIONS The results support further Phase II clinical trials with apramycin at an anticipated efficacious dose of 30 mg/kg once daily.
Collapse
Affiliation(s)
- Chenyan Zhao
- Department of Pharmacy, Uppsala University, SE-75123, Uppsala, Sweden
| | | | - Staffan Rosenborg
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska Institutet, Karolinska University Hospital, Huddinge, SE-14186, Stockholm, Sweden
| | - Rodrigo Palma Villar
- Department Chemical and Pharmaceutical Safety, RISE Research Institutes of Sweden, Sweden
| | - Johan Lindberg
- Department Chemical and Pharmaceutical Safety, RISE Research Institutes of Sweden, Sweden
| | - Sven N Hobbie
- Institute of Medical Microbiology, University of Zurich, CH-8006, Zurich, Switzerland
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, SE-75123, Uppsala, Sweden
| |
Collapse
|
16
|
Translational in vitro and in vivo PKPD modelling for apramycin against Gram-negative lung pathogens to facilitate prediction of human efficacious dose in pneumonia. Clin Microbiol Infect 2022; 28:1367-1374. [PMID: 35598857 DOI: 10.1016/j.cmi.2022.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/23/2022] [Accepted: 05/03/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVES New drugs and methods to efficiently fight carbapenem-resistant Gram-negative pathogens are sorely needed. In this study we characterized the preclinical pharmacokinetics and pharmacodynamics of the clinical-stage drug candidate apramycin in time kill and mouse lung infection models. Based on in vitro and in vivo data, we developed a mathematical model to predict human efficacy. METHODS Three pneumonia-inducing Gram-negative species Acinetobacter baumannii, Pseudomonas aeruginosa, and Klebsiella pneumoniae were studied. Bactericidal kinetics were evaluated with time-kill curves; in vivo pharmacokinetics were studied in healthy and infected mice, with sampling in plasma and epithelial lining fluid after subcutaneous administration; in vivo efficacy was measured in a neutropenic mouse pneumonia model. A pharmacokinetic-pharmacodynamic model, integrating all the data, was developed and simulations were performed. RESULTS Good lung penetration of apramycin in epithelial lining fluid (ELF) was shown (AUCELF/AUCplasma = 88%). Plasma clearance was 48% lower in lung infected mice compared to healthy mice. For two out of five strains studied, a delay in growth (∼5h) was observed in vivo but not in vitro. The mathematical model enabled integration of lung pharmacokinetics to drive mouse PKPD. Simulations predicted that 30 mg/kg of apramycin once daily would result in bacteriostasis in patients. CONCLUSION Apramycin is a candidate for treatment of carbapenem-resistant Gram-negative pneumonia as demonstrated in an integrated modeling framework for three bacterial species. We show that mathematical modelling is a useful tool for simultaneous inclusion of multiple data sources, notably plasma and lung in vivo PK and simulation of expected scenarios in a clinical setting, notably lung infections.
Collapse
|