1
|
Marlin A, Cao M, El Hamouche J, Glaser O, Boros E. Decoding growth inhibitory associated pathways of xenometal-siderophore antibiotic conjugates in S. aureus. Chem Sci 2025; 16:7039-7050. [PMID: 40144493 PMCID: PMC11934059 DOI: 10.1039/d4sc08509d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Pathogenic Staphylococcus aureus causes most infectious disease related deaths in the developed world. Continuously evolving resistance to clinically approved antibiotics and combination therapies limits treatment efficacy; new strategies that evade and slow resistance or produce resistant mutants with reduced fitness are needed. We employ antibiotics conjugated to bacterially recognized siderophores to potentiate their efficacy. Acting as a Trojan horse, the siderophore antibiotic conjugates efficiently deliver the antibiotic inside the bacterial cytoplasm by hijacking the iron transport system pathways which are crucial for bacterial survival. Here, we investigated the mechanism of action of gallium xenometallomycins (siderophore antibiotic conjugates incorporating non-endogenous metal ions), Ga-DFO-Cip and Ga-LDFC-Cip, which have demonstrated high potency compared to the parent antibiotic's efficacy in vitro in S. aureus infection. Employing physicochemical, synthetic and transcriptomic analysis studies, this work reveals that kinetically inert, gallium-containing xenometallomycins targeting cytoplasmic bacterial targets impart differential resistance and gene expression profiles when compared to their parent antibiotic in S. aureus bacterial strains. Both Ga-DFO-Cip and Ga-LDFC-Cip effectively disrupt iron-siderophore biosynthesis and uptake machinery. We affirm our results with the radioactive surrogate 67/68Ga-DFO-Cip and demonstrate that the bacterial uptake in Ga-DFO-Cip-resistant S. aureus strains is impaired, leading to diminished compound accumulation in vitro and in vivo.
Collapse
Affiliation(s)
- Axia Marlin
- Department of Chemistry, Stony Brook University Stony Brook New York 11790 USA
- Department of Chemistry, University of Wisconsin-Madison Madison Wisconsin 53706 USA
| | - Minhua Cao
- Department of Chemistry, Stony Brook University Stony Brook New York 11790 USA
- Department of Chemistry, University of Wisconsin-Madison Madison Wisconsin 53706 USA
| | - Joelle El Hamouche
- Department of Chemistry, Stony Brook University Stony Brook New York 11790 USA
| | - Owen Glaser
- Department of Chemistry, Stony Brook University Stony Brook New York 11790 USA
- Department of Chemistry, University of Wisconsin-Madison Madison Wisconsin 53706 USA
| | - Eszter Boros
- Department of Chemistry, Stony Brook University Stony Brook New York 11790 USA
- Department of Chemistry, University of Wisconsin-Madison Madison Wisconsin 53706 USA
| |
Collapse
|
2
|
Loo CY, Traini D, Young PM, Yeung S, Leong CR, Lee WH. Evaluation of curcumin nanoparticles of various sizes for targeting multidrug-resistant lung cancer cells via inhalation. Nanomedicine (Lond) 2025; 20:141-153. [PMID: 39660666 PMCID: PMC11731332 DOI: 10.1080/17435889.2024.2439241] [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: 07/30/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Inhalation drug delivery can deliver high doses of chemotherapeutic drugs to the lung tumor. This study evaluates the efficacy and the mechanistic pathways of nebulized Cur NPs at various sizes to treat multidrug resistant lung cancer. METHODS AND RESULTS Cur-NPs (30 nm and 200 nm) were nebulized separately onto the multidrug-resistant lung cancer cells (H69AR). Smaller NPs induced significantly higher cell death owing to a higher rate of particle internalization via dynamin-dependent clathrin-mediated endocytosis. Owing to the higher lysosome trafficking of Cur-NP30 nm compared to Cur-NP200 nm, oxidation of lysosome was higher (0.47 ± 0.08 vs 0.38 ± 0.08), contributing to significantly higher mitochondrial membrane potential loss (1.57 ± 0.17 vs 1.30 ± 0.11). MRP1 level in H69AR cells was reduced from 352 ± 12.3 ng/µg of protein (untreated cells) to 287 ± 12 ng/µg of protein (Cur-NP30 nm) and 303 ± 13.4 ng/µg of protein (Cur-NP200 nm). NF-κB, and various cytokine expressions were reduced after treatment with nebulized Cur-NPs. CONCLUSIONS Nebulized Cur-NPs formulations could be internalized into the H69AR cells. The Cur-NPs toxicity toward the H69AR was size and time-dependent. Cur-NP30 nm was more effective than Cur-NP200 nm to retain within the cells to exert higher oxidative stresss-induced cell death.
Collapse
Affiliation(s)
- Ching-Yee Loo
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), Ipoh, Malaysia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Paul M. Young
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
- Department of Marketing, Macquarie Business School, Macquarie University, Sydney, Australia
| | - Stewart Yeung
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Chean Ring Leong
- Malaysian Institute of Chemical and Bioengineering Technology, Universiti Kuala Lumpur, Melaka, Malaysia
| | - Wing-Hin Lee
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), Ipoh, Malaysia
| |
Collapse
|
3
|
Laborda P, Lolle S, Hernando-Amado S, Alcalde-Rico M, Aanæs K, Martínez JL, Molin S, Johansen HK. Mutations in the efflux pump regulator MexZ shift tissue colonization by Pseudomonas aeruginosa to a state of antibiotic tolerance. Nat Commun 2024; 15:2584. [PMID: 38519499 PMCID: PMC10959964 DOI: 10.1038/s41467-024-46938-w] [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: 09/15/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Mutations in mexZ, encoding a negative regulator of the expression of the mexXY efflux pump genes, are frequently acquired by Pseudomonas aeruginosa at early stages of lung infection. Although traditionally related to resistance to the first-line drug tobramycin, mexZ mutations are associated with low-level aminoglycoside resistance when determined in the laboratory, suggesting that their selection during infection may not be necessarily, or only, related to tobramycin therapy. Here, we show that mexZ-mutated bacteria tend to accumulate inside the epithelial barrier of a human airway infection model, thus colonising the epithelium while being protected against diverse antibiotics. This phenotype is mediated by overexpression of lecA, a quorum sensing-controlled gene, encoding a lectin involved in P. aeruginosa tissue invasiveness. We find that lecA overexpression is caused by a disrupted equilibrium between the overproduced MexXY and another efflux pump, MexAB, which extrudes quorum sensing signals. Our results indicate that mexZ mutations affect the expression of quorum sensing-regulated pathways, thus promoting tissue invasiveness and protecting bacteria from the action of antibiotics within patients, something unnoticeable using standard laboratory tests.
Collapse
Affiliation(s)
- Pablo Laborda
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark.
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | - Signe Lolle
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | | | - Manuel Alcalde-Rico
- Centro Nacional de Biotecnología, CSIC, Madrid, Spain
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen Macarena, CSIC, Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Kasper Aanæs
- Department of Otorhinolaryngology, Head and Neck Surgery & Audiology, Rigshospitalet, Copenhagen, Denmark
| | | | - Søren Molin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Helle Krogh Johansen
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Rano S, Bhaduri A, Singh M. Nanoparticle-based platforms for targeted drug delivery to the pulmonary system as therapeutics to curb cystic fibrosis: A review. J Microbiol Methods 2024; 217-218:106876. [PMID: 38135160 DOI: 10.1016/j.mimet.2023.106876] [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: 10/17/2021] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023]
Abstract
Cystic fibrosis (CF) is a genetic disorder of the respiratory system caused by mutation of the Cystic Fibrosis Trans-Membrane Conductance Regulator (CFTR) gene that affects a huge number of people worldwide. It results in difficulty breathing due to a large accumulation of mucus in the respiratory tract, resulting in serious bacterial infections, and subsequent death. Traditional drug-based treatments face hindered penetration at the site of action due to the thick mucus layer. Nanotechnology offers possibilities for developing advanced and effective treatment platforms by focusing on drugs that can penetrate the dense mucus layer, fighting against the underlying bacterial infections, and targeting the genetic cause of the disease. In this review, current nanoparticle-mediated drug delivery platforms for CF, challenges in therapeutics, and future prospects have been highlighted. The effectiveness of the different types of nano-based systems conjugated with various drugs to combat the symptoms and the challenges of treating CF are brought into focus. The toxic effects of these nano-medicines and the various factors that are responsible for their effectiveness are also highlighted.
Collapse
Affiliation(s)
- Sujoy Rano
- Department of Biotechnology, Haldia Institute of Technology, HIT Campus, Purba Medinipur, Haldia 721657, West Bengal, India; In-vitro Biology, Aragen Life Sciences, Hyderabad 500076, Telangana, India
| | - Ahana Bhaduri
- Department of Biotechnology, Haldia Institute of Technology, HIT Campus, Purba Medinipur, Haldia 721657, West Bengal, India
| | - Mukesh Singh
- Department of Biotechnology, Haldia Institute of Technology, HIT Campus, Purba Medinipur, Haldia 721657, West Bengal, India; Department of Botany, Kabi Nazrul College, Murarai, Birbhum 731219 (West Bengal), India.
| |
Collapse
|
5
|
Tewes F, Lamy B, Laroche J, Lamarche I, Marchand S. PK-PD Evaluation of Inhaled Microparticles loaded with Ciprofloxacin-Copper complex in a Rat Model of Chronic Pseudomonas aeruginosa Lung Infection. Int J Pharm X 2023; 5:100178. [PMID: 36970713 PMCID: PMC10033950 DOI: 10.1016/j.ijpx.2023.100178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
The potential gain in efficacy of pulmonary administration over IV administration of some antibiotics such as ciprofloxacin (CIP) may be limited by the short residence time of the drug at the site of infection after nebulization. Complexation of CIP with copper reduced its apparent permeability in vitro through a Calu-3 cell monolayer and greatly increased its pulmonary residence time after aerosolisation in healthy rats. Chronic P. aeruginosa lung infections in cystic fibrosis patients result in airway and alveolar inflammation that may increase the permeability of inhaled antibiotics and alter their fate in the lung after inhalation compared to what was seen in healthy conditions. The objective of this study was to compare the pharmacokinetics and efficacy of CIP-Cu2+ complex-loaded microparticles administered by pulmonary route with a CIP solution administered by IV to model rats with chronic lung infection. After a single pulmonary administration of microparticles loaded with CIP-Cu2+ complex, pulmonary exposure to CIP was increased 2077-fold compared to IV administration of CIP solution. This single lung administration significantly reduced the lung burden of P. aeruginosa expressed as CFU/lung measured 24 h after administration by 10-fold while IV administration of the same dose of CIP was ineffective compared to the untreated control. This better efficacy of inhaled microparticles loaded with CIP-Cu2+ complex compared with CIP solution can be attributed to the higher pulmonary exposure to CIP obtained with inhaled CIP-Cu2+ complex-loaded microparticles than that obtained with IV solution.
Collapse
Affiliation(s)
- Frederic Tewes
- Université de Poitiers, INSERM U1070, Poitiers, France
- Corresponding author.
| | - Barbara Lamy
- Université de Poitiers, INSERM U1070, Poitiers, France
| | - Julian Laroche
- CHU de Poitiers, laboratoire de Toxicologie et de Pharmacocinetique, Poitiers, France
| | | | - Sandrine Marchand
- Université de Poitiers, INSERM U1070, Poitiers, France
- CHU de Poitiers, laboratoire de Toxicologie et de Pharmacocinetique, Poitiers, France
| |
Collapse
|
6
|
The development of a 3D-printed in vitro integrated oro-pharyngeal air-liquid interface cellular throat model for drug transport. Drug Deliv Transl Res 2023; 13:1405-1419. [PMID: 36786980 DOI: 10.1007/s13346-023-01302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/15/2023]
Abstract
To simulate the deposition of drugs in the oro-pharynx region, several in vitro models are available such as the United States Pharmacopeia-Induction Port (USP-IP) throat and the Virginia Commonwealth University (VCU) models. However, currently, there is no such in vitro model that incorporates a biological barrier to elucidate drug transport across the pharyngeal cells. Cellular models such as in vitro air-liquid interface (ALI) models of human respiratory epithelial cell lines are extensively used to study drug transport. To date, no studies have yet been performed to optimise the ALI culture conditions of the human pharyngeal cell line Detroit 562 and determine whether it could be used for drug transport. Therefore, this study aimed to develop a novel 3D-printed throat model integrated with an ALI cellular model of Detroit 562 cells and optimise the culture conditions to investigate whether the combined model could be used to study drug transport, using Lidocaine as a model drug. Differentiating characteristics specific to airway epithelia were assessed using 3 seeding densities (30,000, 60,000, and 80,000 cells/well (c/w), respectively) over 21 days. The results showed that Detroit 562 cells completely differentiates on day 18 of ALI for both 60,000 and 80,000 c/w with significant mucus production, showing response to bacterial and viral stimuli and development of functional tight junctions and Lidocaine transport with no significant differences observed between the ALI models with the 2 cell seeding densities. Results showed the suitability of the Low density (60,000 c/w or 1.8 × 105 cells/cm2) ALI model to study drug transport. Importantly, the developed novel 3D-printed throat model integrated with our optimised in vitro Detroit 562 ALI model showed transport of Lidocaine throat spray. Overall, the study highlights the potential of the novel 3D-printed bio-throat integrated model as a promising in vitro system to investigate the transport of inhalable drug therapies targeted at the oro-pharyngeal region.
Collapse
|
7
|
Hamed MM, Abdelsamie AS, Rox K, Schütz C, Kany AM, Röhrig T, Schmelz S, Blankenfeldt W, Arce‐Rodriguez A, Borrero‐de Acuña JM, Jahn D, Rademacher J, Ringshausen FC, Cramer N, Tümmler B, Hirsch AKH, Hartmann RW, Empting M. Towards Translation of PqsR Inverse Agonists: From In Vitro Efficacy Optimization to In Vivo Proof-of-Principle. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204443. [PMID: 36596691 PMCID: PMC9929129 DOI: 10.1002/advs.202204443] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/28/2022] [Indexed: 06/17/2023]
Abstract
Pseudomonas aeruginosa (PA) is an opportunistic human pathogen, which is involved in a wide range of dangerous infections. It develops alarming resistances toward antibiotic treatment. Therefore, alternative strategies, which suppress pathogenicity or synergize with antibiotic treatments are in great need to combat these infections more effectively. One promising approach is to disarm the bacteria by interfering with their quorum sensing (QS) system, which regulates the release of various virulence factors as well as biofilm formation. Herein, this work reports the rational design, optimization, and in-depth profiling of a new class of Pseudomonas quinolone signaling receptor (PqsR) inverse agonists. The resulting frontrunner compound features a pyrimidine-based scaffold, high in vitro and in vivo efficacy, favorable pharmacokinetics as well as clean safety pharmacology characteristics, which provide the basis for potential pulmonary as well as systemic routes of administration. An X-ray crystal structure in complex with PqsR facilitated further structure-guided lead optimization. The compound demonstrates potent pyocyanin suppression, synergizes with aminoglycoside antibiotic tobramycin against PA biofilms, and is active against a panel of clinical isolates from bronchiectasis patients. Importantly, this in vitro effect translated into in vivo efficacy in a neutropenic thigh infection model in mice providing a proof-of-principle for adjunctive treatment scenarios.
Collapse
Affiliation(s)
- Mostafa M. Hamed
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Ahmed S. Abdelsamie
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of Chemistry of Natural and Microbial ProductsInstitute of Pharmaceutical and Drug Industries ResearchNational Research CentreEl‐Buhouth St.DokkiCairo12622Egypt
| | - Katharina Rox
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of Chemical Biology (CBIO)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
| | - Christian Schütz
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Andreas M. Kany
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Teresa Röhrig
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Stefan Schmelz
- Department of Structure and Function of Proteins (SFPR)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
| | - Wulf Blankenfeldt
- Department of Structure and Function of Proteins (SFPR)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
- Institute for BiochemistryBiotechnology and BioinformaticsTechnische Universität BraunschweigBraunschweigGermany
| | | | - José Manuel Borrero‐de Acuña
- Institute of MicrobiologyTechnische Universität Braunschweig38106BraunschweigGermany
- Braunschweig Integrated Centre of Systems Biology (BRICS)Technische Universität Braunschweig38106BraunschweigGermany
- Departamento de MicrobiologíaFacultad de BiologíaUniversidad de SevillaAv. de la Reina Mercedesno. 6SevillaCP 41012Spain
| | - Dieter Jahn
- Institute of MicrobiologyTechnische Universität Braunschweig38106BraunschweigGermany
- Braunschweig Integrated Centre of Systems Biology (BRICS)Technische Universität Braunschweig38106BraunschweigGermany
| | - Jessica Rademacher
- Department for Respiratory MedicineMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
| | - Felix C. Ringshausen
- Department for Respiratory MedicineMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
- European Reference Network on Rare and Complex Respiratory Diseases (ERN‐ LUNG)FrankfurtGermany
| | - Nina Cramer
- Department for Pediatric PneumologyAllergology and NeonatologyMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Burkhard Tümmler
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
- Department for Pediatric PneumologyAllergology and NeonatologyMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Anna K. H. Hirsch
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| | - Rolf W. Hartmann
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| | - Martin Empting
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| |
Collapse
|
8
|
Nguyen PTN, Le NV, Dinh HMN, Nguyen BQP, Nguyen TVA. Lung penetration and pneumococcal target binding of antibiotics in lower respiratory tract infection. Curr Med Res Opin 2022; 38:2085-2095. [PMID: 36189961 DOI: 10.1080/03007995.2022.2131304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To achieve the therapeutic effects, antibiotics must penetrate rapidly into infection sites and bind to targets. This study reviewed updated knowledge on the ability of antibiotics to penetrate into the lung, their physicochemical properties influencing the pulmonary penetration and their ability to bind to targets on pneumococci. METHODS A search strategy was developed using PubMED, Web of Science, and ChEMBL. Data on serum protein binding, drug concentration, target binding ability, drug transporters, lung penetration, physicochemical properties of antibiotics in low respiratory tract infection (LRTI) were collected. RESULTS It was seen that infection site-to-serum concentration ratios of most antibiotics are >1 at different time points except for ceftriaxone, clindamycin and vancomycin. Most agents have proper physicochemical properties that facilitate antibiotic penetration. In antimicrobial-resistant Streptococcus pneumoniae, the binding affinity of antibiotics to targets mostly decreases compared to that in susceptible strains. The data on binding affinity of linezolid, clindamycin and vancomycin were insufficient. The higher drug concentration at the infection sites compared to that in the blood can be associated with inflammation conditions. Little evidence showed the effect of drug transporters on the clinical efficacy of antibiotics against LRTI. CONCLUSIONS Data on antibiotic penetration into the lung in LRTI patients and binding affinity of antibiotics for pneumococcal targets are still limited. Further studies are required to clarify the associations of the lung penetration and target binding ability of antibitotics with therapeutic efficacy to help propose the right antibiotics for LRTI.
Collapse
Affiliation(s)
| | - Nho Van Le
- Danang University of Medical Technology and Pharmacy, Da Nang, Vietnam
| | | | | | - Thi Van Anh Nguyen
- Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
9
|
Li Z, Du X, Tian S, Fan S, Zuo X, Li Y, Wang R, Wang B, Huang Y. Pharmacokinetic herb-drug interactions: Altered systemic exposure and tissue distribution of ciprofloxacin, a substrate of multiple transporters, after combined treatment with Polygonum capitatum Buch.-Ham. ex D. Don extracts. Front Pharmacol 2022; 13:1033667. [PMID: 36386188 PMCID: PMC9640990 DOI: 10.3389/fphar.2022.1033667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Combination of Polygonum capitatum Buch.-Ham. ex D. Don extract (PCE) and ciprofloxacin (CIP) was commonly prescribed in the treatment of urinary tract infections. Their pharmacokinetic herb-drug interactions (HDIs) were focused in this study to assess potential impact on the safety and effectiveness. Methods: A randomized, three-period, crossover trial was designed to study the pharmacokinetic HDI between PCE and CIP in healthy humans. Their pharmacokinetic- and tissue distribution-based HDIs were also evaluated in rats. Gallic acid (GA) and protocatechuic acid (PCA) were chosen as PK-markers of PCE in humans and rats. Potential drug interaction mechanisms were revealed by assessing the effects of PCE on the activity and expression of multiple transporters, including OAT1/3, OCT2, MDR1, and BCRP. Results: Concurrent use of PCE substantially reduced circulating CIP (approximately 40%–50%) in humans and rats, while CIP hardly changed circulating GA and PCA. PCE significantly increased the tissue distribution of CIP in the prostate and testis of rats, but decreased in liver and lungs. Meanwhile, CIP significantly increased the tissue distribution of GA or PCA in the prostate and testis of rats, but decreased in kidney and heart. In the transporter-mediated in vitro HDI, GA and PCA presented inhibitory effects on OAT1/3 and inductive effects on MDR1 and BCRP. Conclusion: Multiple transporter-mediated HDI contributes to effects of PCE on the reduced systemic exposure and altered tissue distribution of CIP. More attention should be paid on the potential for PCE-perpetrated interactions.
Collapse
Affiliation(s)
- Ziqiang Li
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xi Du
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuang Tian
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shanshan Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xurui Zuo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanfen Li
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruihua Wang
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Baohe Wang
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Huang
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Yuhong Huang,
| |
Collapse
|
10
|
Shi C, Ignjatović J, Wang J, Guo Y, Zhang L, Cvijić S, Cun D, Yang M. Evaluating the pharmacokinetics of intrapulmonary administered ciprofloxacin solution for respiratory infections using in vivo and in silico PBPK rat model studies. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.04.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
11
|
Danis-Wlodarczyk KM, Cai A, Chen A, Gittrich MR, Sullivan MB, Wozniak DJ, Abedon ST. Friends or Foes? Rapid Determination of Dissimilar Colistin and Ciprofloxacin Antagonism of Pseudomonas aeruginosa Phages. Pharmaceuticals (Basel) 2021; 14:1162. [PMID: 34832944 PMCID: PMC8624478 DOI: 10.3390/ph14111162] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 12/14/2022] Open
Abstract
Phage therapy is a century-old technique employing viruses (phages) to treat bacterial infections, and in the clinic it is often used in combination with antibiotics. Antibiotics, however, interfere with critical bacterial metabolic activities that can be required by phages. Explicit testing of antibiotic antagonism of phage infection activities, though, is not a common feature of phage therapy studies. Here we use optical density-based 'lysis-profile' assays to assess the impact of two antibiotics, colistin and ciprofloxacin, on the bactericidal, bacteriolytic, and new-virion-production activities of three Pseudomonas aeruginosa phages. Though phages and antibiotics in combination are more potent in killing P. aeruginosa than either acting alone, colistin nevertheless substantially interferes with phage bacteriolytic and virion-production activities even at its minimum inhibitory concentration (1× MIC). Ciprofloxacin, by contrast, has little anti-phage impact at 1× or 3× MIC. We corroborate these results with more traditional measures, particularly colony-forming units, plaque-forming units, and one-step growth experiments. Our results suggest that ciprofloxacin could be useful as a concurrent phage therapy co-treatment especially when phage replication is required for treatment success. Lysis-profile assays also appear to be useful, fast, and high-throughput means of assessing antibiotic antagonism of phage infection activities.
Collapse
Affiliation(s)
| | - Alice Cai
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Anna Chen
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Marissa R. Gittrich
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Matthew B. Sullivan
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
- Department of Civil, Environmental and Geodetic Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA;
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Stephen T. Abedon
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| |
Collapse
|
12
|
Development and in vitro characterization of a novel pMDI diclofenac formulation as an inhalable anti-inflammatory therapy for cystic fibrosis. Int J Pharm 2021; 596:120319. [PMID: 33540036 DOI: 10.1016/j.ijpharm.2021.120319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 11/24/2022]
Abstract
Anti-inflammatory treatment options for cystic fibrosis (CF) patients are currently limited and as such, there is an imperative need to develop new anti-inflammatory agents to reduce the persistent inflammation present within CF lungs. This study explored the potential of Diclofenac (DICLO) as a novel inhaled anti-inflammatory drug for CF treatment. The anti-inflammatory activity of DICLO on an air-liquid interface (ALI) cell culture model of healthy (NuLi-1) and CF (CuFi-1) airways showed a significant reduction in the secretion of pro-inflammatory cytokines, IL-6 and IL-8. Therefore, pressurized metered dose inhaler (pMDI) DICLO formulations were developed to allow targeted DICLO delivery to CF airways. As such, two pMDI DICLO formulations with varying ethanol concentrations: 5% (w/w) equating to 150 µg of DICLO per dose (Low dose), and 15% (w/w) equating to 430 µg of DICLO per dose (High dose) were developed and characterized to determine the optimum formulation. The Low dose pMDI DICLO formulation showed a significantly smaller particle diameter with uniform distribution resulting in a greater aerosol performance when compared to High dose formulation. Consequently, the Low dose pMDI DICLO formulation was further evaluated in terms of in vitro transport characteristics and anti-inflammatory activity. Importantly, the DICLO pMDI displayed anti-inflammatory activity in both healthy and CF in vitro models, highlighting the potential of an aerosolized low-dose DICLO formulation as a promising inhaled anti-inflammatory therapy for CF treatment.
Collapse
|
13
|
Montefusco-Pereira CV, Carvalho-Wodarz CDS, Seeger J, Kloft C, Michelet R, Lehr CM. Decoding (patho-)physiology of the lung by advanced in vitro models for developing novel anti-infectives therapies. Drug Discov Today 2020; 26:148-163. [PMID: 33232842 DOI: 10.1016/j.drudis.2020.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/11/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Advanced lung cell culture models provide physiologically-relevant and complex data for mathematical models to exploit host-pathogen responses during anti-infective drug testing.
Collapse
Affiliation(s)
- Carlos Victor Montefusco-Pereira
- Department of Pharmacy, Saarland University, Saarbruecken, Germany; Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | | | - Johanna Seeger
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbruecken, Germany; Department of Pharmacy, Saarland University, Saarbruecken, Germany
| |
Collapse
|
14
|
Lababidi N, Montefusco-Pereira CV, de Souza Carvalho-Wodarz C, Lehr CM, Schneider M. Spray-dried multidrug particles for pulmonary co-delivery of antibiotics with N-acetylcysteine and curcumin-loaded PLGA-nanoparticles. Eur J Pharm Biopharm 2020; 157:200-210. [PMID: 33222771 DOI: 10.1016/j.ejpb.2020.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
Nowadays, the resistance of bacterial biofilms towards the available antibiotics is a severe problem. Therefore, many efforts were devoted to develop new formulations using nanotechnology. We have developed an inhalable microparticle formulation using spray-drying combining multiple drugs: an antibiotic (tobramycin, ciprofloxacin or azithromycin), N-acetylcysteine (NAC), and curcumin (Cur). The use of PLGA nanoparticles (NP) also allowed incorporating curcumin to facilitate spray drying and modify the release of some compounds. The aerosolizable microparticles formulations were characterized in terms of size, morphology, and aerodynamic properties. Biocompatibility when tested on macrophage-like cells was acceptable after 20 h exposure for concentrations up to at least 32 µg/mL. Antibacterial activity of free drugs versus drugs in the multiple drug formulations was evaluated on P. aeruginosa in the same range. When co-delivered the efficacy of tobramycin was enhanced compared to the free drug for the 1 µg/mL concentration. The combinations of azithromycin and ciprofloxacin with NAC and Cur did not show an improved antibacterial activity. Bacteria-triggered cytokine release was not inhibited by free antibiotics, except for TNF-α. In contrast, the application of NAC and the addition of curcumin-loaded PLGA NPs showed a higher potential to inhibit TNF-α, IL-8, and IL-1β release. Overall, the approach described here allows simultaneous delivery of antibacterial, mucolytic, and anti-inflammatory compounds in a single inhalable formulation and may therefore pave the way for a more efficient therapy of pulmonary infections.
Collapse
Affiliation(s)
- Nashrawan Lababidi
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany
| | - Carlos Victor Montefusco-Pereira
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany; Helmholtz Institute for Pharmaceutical Research Saarland, Campus E8 1, 66123 Saarbrücken, Germany
| | | | - Claus-Michael Lehr
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany; Helmholtz Institute for Pharmaceutical Research Saarland, Campus E8 1, 66123 Saarbrücken, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany.
| |
Collapse
|
15
|
Luscher A, Simonin J, Falconnet L, Valot B, Hocquet D, Chanson M, Resch G, Köhler T, van Delden C. Combined Bacteriophage and Antibiotic Treatment Prevents Pseudomonas aeruginosa Infection of Wild Type and cftr- Epithelial Cells. Front Microbiol 2020; 11:1947. [PMID: 32983005 PMCID: PMC7479825 DOI: 10.3389/fmicb.2020.01947] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
With the increase of infections due to multidrug resistant bacterial pathogens and the shortage of antimicrobial molecules with novel targets, interest in bacteriophages as a therapeutic option has regained much attraction. Before the launch of future clinical trials, in vitro studies are required to better evaluate the efficacies and potential pitfalls of such therapies. Here we studied in an ex vivo human airway epithelial cell line model the efficacy of phage and ciprofloxacin alone and in combination to treat infection by Pseudomonas aeruginosa. The Calu-3 cell line and the isogenic CFTR knock down cell line (cftr-) infected apically with P. aeruginosa strain PAO1 showed a progressive reduction in transepithelial resistance during 24 h. Administration at 6 h p.i. of single phage, phage cocktails or ciprofloxacin alone prevented epithelial layer destruction at 24 h p.i. Bacterial regrowth, due to phage resistant mutants harboring mutations in LPS synthesis genes, occurred thereafter both in vitro and ex vivo. However, co-administration of two phages combined with ciprofloxacin efficiently prevented PAO1 regrowth and maintained epithelial cell integrity at 72 p.i. The phage/ciprofloxacin treatment did not induce an inflammatory response in the tested cell lines as determined by nanoString® gene expression analysis. We conclude that combination of phage and ciprofloxacin efficiently protects wild type and cftr- epithelial cells from infection by P. aeruginosa and emergence of phage resistant mutants without inducing an inflammatory response. Hence, phage-antibiotic combination should be a safe and promising anti-Pseudomonas therapy for future clinical trials potentially including cystic fibrosis patients.
Collapse
Affiliation(s)
- Alexandre Luscher
- Transplant Infectious Diseases Unit, Geneva University Hospitals, Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Juliette Simonin
- Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Léna Falconnet
- Transplant Infectious Diseases Unit, Geneva University Hospitals, Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Benoît Valot
- UMR CNRS 6249 Chrono-Environnement, University of Franche-Comté-Bourgogne, Besançon, France
- Bioinformatique et Big Data au Service de la Santé, UFR Santé, Université de Bourgogne Franche-Comté, Besançon, France
| | - Didier Hocquet
- UMR CNRS 6249 Chrono-Environnement, University of Franche-Comté-Bourgogne, Besançon, France
- Bioinformatique et Big Data au Service de la Santé, UFR Santé, Université de Bourgogne Franche-Comté, Besançon, France
- Department of Infection Control, University Hospital of Besançon, Besançon, France
| | - Marc Chanson
- Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Grégory Resch
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Thilo Köhler
- Transplant Infectious Diseases Unit, Geneva University Hospitals, Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Christian van Delden
- Transplant Infectious Diseases Unit, Geneva University Hospitals, Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
16
|
Strzempek W, Korzeniowska A, Kowalczyk A, Roth WJ, Gil B. Detemplated and Pillared 2-Dimensional Zeolite ZSM-55 with Ferrierite Layer Topology as a Carrier for Drugs. Molecules 2020; 25:molecules25153501. [PMID: 32752039 PMCID: PMC7435734 DOI: 10.3390/molecules25153501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 01/26/2023] Open
Abstract
The present studies were conducted to show the potential of 2D zeolites as effective and non-toxic carriers of drugs. Layered zeolites exhibit adjustable interlayer porosity which can be exploited for controlled drug delivery allowing detailed investigation of the drug release because the structure of the carrier is known exactly. This study was conducted with model drugs ciprofloxacin and piracetam, and ZSM-55 with ca 1 nm thick layers, in detemplated and pillared forms. The release profiles differed from the commercial, crystalline forms of drugs-the release rate increased for ciprofloxacin and decreased for piracetam. To understand the dissolution mechanisms the release data were fitted to Korsmeyer-Peppas equation, showing Fickian (for pillared) and anomalous (for detemplated sample) transport. FT-IR studies showed that strong interaction carrier-drug may be responsible for the modified, slowed down release of piracetam while better solubility and faster release of ciprofloxacin was attributed to formation of the protonated form resulting in weaker interaction with the zeolite than in the pure crystalline form. Two independent tests on L929 mice fibroblasts (ToxiLight and PrestoBlue) showed that ZSM-55, in moderate concentrations may be safely used as a carrier of drug molecules, not having negative effect on the cells viability or proliferation rate.
Collapse
|
17
|
Inhaled rapamycin solid lipid nano particles for the treatment of Lymphangioleiomyomatosis. Eur J Pharm Sci 2020; 142:105098. [DOI: 10.1016/j.ejps.2019.105098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 01/03/2023]
|
18
|
Chai G, Park H, Yu S, Zhou F, Li J, Xu Q, Zhou QT. Evaluation of co-delivery of colistin and ciprofloxacin in liposomes using an in vitro human lung epithelial cell model. Int J Pharm 2019; 569:118616. [PMID: 31415873 DOI: 10.1016/j.ijpharm.2019.118616] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 01/15/2023]
Abstract
Respiratory tract infections caused by multidrug-resistant Gram-negative bacteria are serious burdens to the public. Our previous findings indicated that co-loading of colistin and ciprofloxacin via liposomes improved in vitro antimicrobial activities against multidrug resistant Pseudomonas aeruginosa as compared to the monotherapies. The current study aims to investigate the transport behavior of colistin and ciprofloxacin in liposomes using the in vitro Calu-3 cell monolayer, which is a lung epithelial model cultured under the air-interfaced condition. The cell viability results demonstrated that there was no obvious toxicity of cells exposed to single or co-administered drugs at the concentration ≤500 μg/mL. Transport of ciprofloxacin into the cells was easier than that of colistin, which reached a plateau rapidly. Colistin was less trapped in the mucus or adhered to the apical cell membrane, and less transported across the cell monolayer than ciprofloxacin. The deposition of ciprofloxacin on the apical side increased over time (from 1 to 4 h). There was no drug-drug interaction observed during the transport of ciprofloxacin and colistin across the cell monolayer, when they were dosed together in the solution form. The amount of drug transported across the cell monolayer was decreased in both agents when loaded in liposomes. Both drugs were more trapped in the mucus or more adhered to the apical side cell membrane of the cell monolayer when they were in liposomes. This study demonstrated that co-delivery of colistin and ciprofloxacin in a single liposome can reduce transport capacity of both drugs across the lung epithelial cell monolayer and enhance drug retention on the lung epithelial surfaces; therefore, it is a promising approach to treat the respiratory infections caused by multidrug resistant Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Guihong Chai
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Heejun Park
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Shihui Yu
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Fanfan Zhou
- School of Pharmacy, The University of Sydney, NSW 2006, Australia
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Qingguo Xu
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
19
|
Farhangi M, Mahboubi A, Kobarfard F, Vatanara A, Mortazavi SA. Optimization of a dry powder inhaler of ciprofloxacin-loaded polymeric nanomicelles by spray drying process. Pharm Dev Technol 2019; 24:584-592. [DOI: 10.1080/10837450.2018.1545237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Mahdieh Farhangi
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Mahboubi
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Food Safety Research Center, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Vatanara
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Alireza Mortazavi
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
In Vitro Mechanistic Study of the Distribution of Lascufloxacin into Epithelial Lining Fluid. Antimicrob Agents Chemother 2019; 63:AAC.02208-18. [PMID: 30718243 DOI: 10.1128/aac.02208-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/25/2019] [Indexed: 11/20/2022] Open
Abstract
The present study aimed to clarify the mechanism underlying the high distribution of lascufloxacin in epithelial lining fluid (ELF). Involvement of transporters was examined by transcellular transport across Calu-3 and transporter-overexpressing cells; the binding of lascufloxacin to ELF components was examined by an organic solvent-water partitioning system that employed pulmonary surfactant and phospholipids. Transcellular transport across the transporter-overexpressing cells indicated lascufloxacin to be a substrate of both P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP); therefore, its transport across Calu-3 cells was inhibited by P-gp and BCRP inhibitors. However, permeability and efflux ratios of lascufloxacin were similar to those of the other quinolones with relatively low ELF distribution, indicating the existence of another mechanism for lascufloxacin distribution in ELF. Amongst pulmonary surfactants, which are a primary component of ELF, lascufloxacin preferentially bound to phosphatidylserine (PhS) from several phospholipids, and the binding was significantly greater than that for other quinolones. This binding was saturable with two apparent classes of binding sites and inhibited by some weakly basic drugs, indicating the presence of an ionic bond. In conclusion, the results of this study suggest that the binding of lascufloxacin to PhS in the pulmonary surfactant is the major mechanism of the high distribution of lascufloxacin in the ELF.
Collapse
|
21
|
Tomas A, Stilinović N, Sabo A, Tomić Z. Use of microdialysis for the assessment of fluoroquinolone pharmacokinetics in the clinical practice. Eur J Pharm Sci 2019; 131:230-242. [PMID: 30811969 DOI: 10.1016/j.ejps.2019.02.032] [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: 10/18/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
Antibacterial drugs, including fluoroquinolones, can exert their therapeutic action only with adequate penetration at the infection site. Multiple factors, such as rate of protein binding, drug liposolubility and organ blood-flow all influence ability of antibiotics to penetrate target tissues. Microdialysis is an in vivo sampling technique that has been successfully applied to measure the distribution of fluoroquinolones in the interstitial fluid of different tissues both in animal studies and clinical setting. Tissue concentrations need to be interpreted within the context of the pathogenesis and causative agents implicated in infections. Integration of microdialysis -derived tissue pharmacokinetics with pharmacodynamic data offers crucial information for correlating exposure with antibacterial effect. This review explores these concepts and provides an overview of tissue concentrations of fluoroquinolones derived from microdialysis studies and explores the therapeutic implications of fluoroquinolone distribution at various target tissues.
Collapse
Affiliation(s)
- Ana Tomas
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Serbia.
| | - Nebojša Stilinović
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Serbia
| | - Ana Sabo
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Serbia
| | - Zdenko Tomić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Serbia
| |
Collapse
|
22
|
Zimmermann ES, de Miranda Silva C, Neris C, Torres BGDS, Schmidt S, Dalla Costa T. Population pharmacokinetic modeling to establish the role of P-glycoprotein on ciprofloxacin distribution to lung and prostate following intravenous and intratracheal administration to Wistar rats. Eur J Pharm Sci 2018; 127:319-329. [PMID: 30423435 DOI: 10.1016/j.ejps.2018.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/03/2018] [Accepted: 11/07/2018] [Indexed: 02/06/2023]
Abstract
Ciprofloxacin (CIP) is indicated for clinical treatment of urinary and respiratory tract infections. Poor infection site penetration and consequent insufficient exposure to the antimicrobial agent may be the reason for some therapeutic failures. Ciprofloxacin is reported as a substrate for efflux transporters, such as P-glycoprotein, which could be related to the presence of sub-therapeutic drug concentration at the infection site. In the present work we evaluated CIP pharmacokinetics (PK) in plasma and lung and prostate tissues of Wistar rats after intravenous (i.v.) and intratracheal (i.t.) dosing (7 mg/Kg) in the presence and absence of P-gp inhibitor tariquidar (TAR, 15 mg/Kg). Microdialysis was applied to determine free tissue concentration-time profiles and the obtained data were analyzed by non-compartmental and population PK (popPK) analysis. A sequential strategy was used to develop the popPK model: characterization of CIP PK in tissues (Tissue model) was performed subsequently to CIP PK modeling in plasma (Plasma model). Two and three compartmental models were used to simultaneously characterize plasma concentrations after i.t. and i.v. dosing; the distribution model was developed by separating the central compartment into venous and arterial compartment and by adding lung and prostate; TAR was identified as a significant covariate for clearance and volume of distribution of central compartment as well as for inter-compartmental clearance. Our results indicate an impact of P-gp on plasma PK, likely by acting on renal active secretion of CIP. Regarding CIP exposure in lung and prostate tissues, our results suggest a complex interplay between drug transporters; P-gp inhibition by TAR was likely counterbalanced by the activity of other efflux/influx transporters, which could not be fully characterized by our model.
Collapse
Affiliation(s)
- Estevan Sonego Zimmermann
- Pharmaceutical Sciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Carolina de Miranda Silva
- Center for Pharmacometrics & Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, USA
| | - Camila Neris
- Pharmaceutical Sciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Stephan Schmidt
- Center for Pharmacometrics & Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, USA
| | - Teresa Dalla Costa
- Pharmaceutical Sciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
23
|
In Vitro Models for Studying Transport Across Epithelial Tissue Barriers. Ann Biomed Eng 2018; 47:1-21. [DOI: 10.1007/s10439-018-02124-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022]
|
24
|
Zaichik S, Steinbring C, Menzel C, Knabl L, Orth-Höller D, Ellemunter H, Niedermayr K, Bernkop-Schnürch A. Development of self-emulsifying drug delivery systems (SEDDS) for ciprofloxacin with improved mucus permeating properties. Int J Pharm 2018; 547:282-290. [DOI: 10.1016/j.ijpharm.2018.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/30/2018] [Accepted: 06/03/2018] [Indexed: 12/11/2022]
|
25
|
Antimicrobial molecules in the lung: formulation challenges and future directions for innovation. Future Med Chem 2018; 10:575-604. [PMID: 29473765 DOI: 10.4155/fmc-2017-0162] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inhaled antimicrobials have been extremely beneficial in treating respiratory infections, particularly chronic infections in a lung with cystic fibrosis. The pulmonary delivery of antibiotics has been demonstrated to improve treatment efficacy, reduce systemic side effects and, critically, reduce drug exposure to commensal bacteria compared with systemic administration, reducing selective pressure for antimicrobial resistance. This review will explore the specific challenges of pulmonary delivery of a number of differing antimicrobial molecules, and the formulation and technological approaches that have been used to overcome these difficulties. It will also explore the future challenges being faced in the development of inhaled products and respiratory infection treatment, and identify future directions of innovation, with a particular focus on respiratory infections caused by multiple drug-resistant pathogens.
Collapse
|
26
|
Loo CY, Lee WH, Lauretani G, Scalia S, Cipolla D, Traini D, Young P, Ong HX. Sweetening Inhaled Antibiotic Treatment for Eradication of Chronic Respiratory Biofilm Infection. Pharm Res 2018; 35:50. [PMID: 29417313 DOI: 10.1007/s11095-018-2350-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 01/17/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE The failure of chronic therapy with antibiotics to clear persistent respiratory infection is the key morbidity and mortality factor for patients with chronic lung diseases, primarily due to the presence of biofilm in the lungs. It is hypothesised that carbon sources, such as mannitol, could stimulate the metabolic activity of persister cells within biofilms and restore their susceptibility to antibiotics. The aims of the current study are to: (1) establish a representative in vitro model of Pseudomonas aeruginosa biofilm lung infection, and (2) investigate the effects of nebulised mannitol on antibiotic efficacy, focusing on ciprofloxacin, in the eradication of biofilm. METHOD Air interface biofilm was cultured onto Snapwell inserts incorporated into a modified pharmacopeia deposition apparatus, the Anderson Cascade Impactor (ACI). Three different formulations including mannitol only, ciprofloxacin only and combined ciprofloxacin and mannitol were nebulised onto the P. aeruginosa biofilm using the modified ACI. Antibacterial effectiveness was evaluated using colony-forming units counts, biofilm penetration and scanning electron microscopy. RESULTS Nebulised mannitol promotes the dispersion of bacteria from the biofilm and demonstrated a synergistic enhancement of the antibacterial efficacy of ciprofloxacin compared to delivery of antibiotic alone. CONCLUSIONS The combination of ciprofloxacin and mannitol may provide an important new strategy to improve antibiotic therapy for the treatment of chronic lung infections. Furthermore, the development of a representative lung model of bacterial biofilm could potentially be used as a platform for future new antimicrobial pre-clinical screening.
Collapse
Affiliation(s)
- Ching-Yee Loo
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2037, Australia
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (RCMP UniKL), Ipoh, Perak, Malaysia
| | - Wing-Hin Lee
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2037, Australia
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (RCMP UniKL), Ipoh, Perak, Malaysia
| | - Gianluca Lauretani
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2037, Australia
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Santo Scalia
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - David Cipolla
- Pharmaceutical Sciences, Aradigm Corporation, Hayward, California, USA
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2037, Australia
- Discipline of Pharmacology, Sydney Medical School, Camperdown, NSW, 2006, Australia
| | - Paul Young
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2037, Australia
- Discipline of Pharmacology, Sydney Medical School, Camperdown, NSW, 2006, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2037, Australia.
- Discipline of Pharmacology, Sydney Medical School, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
27
|
Darweesh RS, Sakagami M. In vitro lung epithelial cell transport and anti-interleukin-8 releasing activity of liposomal ciprofloxacin. Eur J Pharm Sci 2018; 115:68-76. [PMID: 29337216 DOI: 10.1016/j.ejps.2018.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 11/28/2017] [Accepted: 01/08/2018] [Indexed: 01/14/2023]
Abstract
As a promising long-acting inhaled formulation, liposomal ciprofloxacin (Lipo-CPFX) was characterized in the in vitro human lung epithelial Calu-3 cell monolayer system, compared to ciprofloxacin in solution (CPFX). Its modulated absorptive transport and uptake, and sustained inhibitory activity against induced pro-inflammatory interleukin-8 (IL-8) release were examined. The absorptive transport and uptake kinetics for Lipo-CPFX and CPFX were determined at 0.1-50 mg/ml in the Transwell system. The Lipo-CPFX transport was then challenged for mechanistic exploration via cell energy depletion, a reduced temperature, endocytosis and/or lipid fusion inhibition, and addition of excess non-loaded liposomes. The inhibitory activities of Lipo-CPFX and CPFX against lipopolysaccharide (LPS)-induced IL-8 release were assessed in a co-incubation or pre-incubation mode. In the tight Calu-3 cell monolayers, Lipo-CPFX yielded 15-times slower ciprofloxacin flux of absorptive transport and 5-times lower cellular drug uptake than CPFX. Its transport appeared to be transcellular; kinetically linear, proportional to encapsulated ciprofloxacin concentration; and consistent with the cell energy-independent lipid bilayer fusion mechanism. Lipo-CPFX was equipotent to CPFX in the anti-IL-8 releasing activity upon 24 h co-incubation with LPS. Additionally, Lipo-CPFX, but not CPFX, retained the anti-IL-8 releasing activity even 24 h after pre-incubation. In conclusion, Lipo-CPFX enabled slower absorptive lung epithelial cell transport and uptake of ciprofloxacin, apparently via the lipid bilayer fusion mechanism, and the sustained inhibitory activity against LPS-induced IL-8 release, compared to CPFX.
Collapse
Affiliation(s)
- Ruba S Darweesh
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, 410 N. 12th Street, Richmond, Virginia, 23298, USA.; Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22,110, Jordan
| | - Masahiro Sakagami
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, 410 N. 12th Street, Richmond, Virginia, 23298, USA..
| |
Collapse
|
28
|
Intravenous anti-MRSA phosphatiosomes mediate enhanced affinity to pulmonary surfactants for effective treatment of infectious pneumonia. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:215-225. [PMID: 29128664 DOI: 10.1016/j.nano.2017.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/26/2017] [Accepted: 10/16/2017] [Indexed: 12/25/2022]
Abstract
The aim of this study was to develop PEGylated phosphatidylcholine (PC)-rich nanovesicles (phosphatiosomes) carrying ciprofloxacin (CIPX) for lung targeting to eradicate extracellular and intracellular methicillin-resistant Staphylococcus aureus (MRSA). Soyaethyl morphonium ethosulfate (SME) was intercalated in the nanovesicle surface with the dual goals of achieving strengthened bactericidal activity of CIPX-loaded phosphatiosomes and delivery to the lungs. The isothermal titration calorimetry (ITC) results proved the strong association of SME phosphatiosomes with pulmonary surfactant. We demonstrated a superior anti-MRSA activity of SME phosphatiosomes compared to plain phosphatiosomes and to free CIPX. A synergistic effect of CIPX and SME nanocarriers was found in the biofilm eradication. SME phosphatiosomes were readily engulfed by the macrophages, restricting the intracellular MRSA count by 1-2 log units. SME phosphatiosomes efficiently accumulated in the lungs after intravenous injection. In a rat model of lung infection, the MRSA burden in the lungs could be decreased by 8-fold after SME nanosystem application.
Collapse
|
29
|
Akdag Cayli Y, Sahin S, Buttini F, Balducci AG, Montanari S, Vural I, Oner L. Dry powders for the inhalation of ciprofloxacin or levofloxacin combined with a mucolytic agent for cystic fibrosis patients. Drug Dev Ind Pharm 2017; 43:1378-1389. [DOI: 10.1080/03639045.2017.1318902] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - Selma Sahin
- Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | | | | | | | - Imran Vural
- Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Levent Oner
- Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
30
|
Sadiq MW, Nielsen EI, Khachman D, Conil JM, Georges B, Houin G, Laffont CM, Karlsson MO, Friberg LE. A whole-body physiologically based pharmacokinetic (WB-PBPK) model of ciprofloxacin: a step towards predicting bacterial killing at sites of infection. J Pharmacokinet Pharmacodyn 2017; 44:69-79. [PMID: 27578330 PMCID: PMC5376394 DOI: 10.1007/s10928-016-9486-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/18/2016] [Indexed: 11/26/2022]
Abstract
The purpose of this study was to develop a whole-body physiologically based pharmacokinetic (WB-PBPK) model for ciprofloxacin for ICU patients, based on only plasma concentration data. In a next step, tissue and organ concentration time profiles in patients were predicted using the developed model. The WB-PBPK model was built using a non-linear mixed effects approach based on data from 102 adult intensive care unit patients. Tissue to plasma distribution coefficients (Kp) were available from the literature and used as informative priors. The developed WB-PBPK model successfully characterized both the typical trends and variability of the available ciprofloxacin plasma concentration data. The WB-PBPK model was thereafter combined with a pharmacokinetic-pharmacodynamic (PKPD) model, developed based on in vitro time-kill data of ciprofloxacin and Escherichia coli to illustrate the potential of this type of approach to predict the time-course of bacterial killing at different sites of infection. The predicted unbound concentration-time profile in extracellular tissue was driving the bacterial killing in the PKPD model and the rate and extent of take-over of mutant bacteria in different tissues were explored. The bacterial killing was predicted to be most efficient in lung and kidney, which correspond well to ciprofloxacin's indications pneumonia and urinary tract infections. Furthermore, a function based on available information on bacterial killing by the immune system in vivo was incorporated. This work demonstrates the development and application of a WB-PBPK-PD model to compare killing of bacteria with different antibiotic susceptibility, of value for drug development and the optimal use of antibiotics .
Collapse
Affiliation(s)
- Muhammad W Sadiq
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden
- CVMD iMED, DMPK, Astrazeneca, Mölndal, Sweden
| | - Elisabet I Nielsen
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden
| | - Dalia Khachman
- INRA, Toxalim, Toulouse, France
- Universite de Toulouse, Toulouse, France
| | - Jean-Marie Conil
- Laboratoire de Pharmacocinetique et Toxicologie Clinique, Hospital Purpan, Institut Federatif de Biologie, Toulouse, France
- Pole d'Anesthesie-Reanimation, Hopital Rangueil, Toulouse, France
| | - Bernard Georges
- Laboratoire de Pharmacocinetique et Toxicologie Clinique, Hospital Purpan, Institut Federatif de Biologie, Toulouse, France
- Pole d'Anesthesie-Reanimation, Hopital Rangueil, Toulouse, France
| | - Georges Houin
- Laboratoire de Pharmacocinetique et Toxicologie Clinique, Hospital Purpan, Institut Federatif de Biologie, Toulouse, France
| | - Celine M Laffont
- INRA, Toxalim, Toulouse, France
- Universite de Toulouse, Toulouse, France
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden
| | - Lena E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden.
| |
Collapse
|
31
|
Ufuk A, Assmus F, Francis L, Plumb J, Damian V, Gertz M, Houston JB, Galetin A. In Vitro and in Silico Tools To Assess Extent of Cellular Uptake and Lysosomal Sequestration of Respiratory Drugs in Human Alveolar Macrophages. Mol Pharm 2017; 14:1033-1046. [PMID: 28252969 DOI: 10.1021/acs.molpharmaceut.6b00908] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accumulation of respiratory drugs in human alveolar macrophages (AMs) has not been extensively studied in vitro and in silico despite its potential impact on therapeutic efficacy and/or occurrence of phospholipidosis. The current study aims to characterize the accumulation and subcellular distribution of drugs with respiratory indication in human AMs and to develop an in silico mechanistic AM model to predict lysosomal accumulation of investigated drugs. The data set included 9 drugs previously investigated in rat AM cell line NR8383. Cell-to-unbound medium concentration ratio (Kp,cell) of all drugs (5 μM) was determined to assess the magnitude of intracellular accumulation. The extent of lysosomal sequestration in freshly isolated human AMs from multiple donors (n = 5) was investigated for clarithromycin and imipramine (positive control) using an indirect in vitro method (±20 mM ammonium chloride, NH4Cl). The AM cell parameters and drug physicochemical data were collated to develop an in silico mechanistic AM model. Three in silico models differing in their description of drug membrane partitioning were evaluated; model (1) relied on octanol-water partitioning of drugs, model (2) used in vitro data to account for this process, and model (3) predicted membrane partitioning by incorporating AM phospholipid fractions. In vitro Kp,cell ranged >200-fold for respiratory drugs, with the highest accumulation seen for clarithromycin. A good agreement in Kp,cell was observed between human AMs and NR8383 (2.45-fold bias), highlighting NR8383 as a potentially useful in vitro surrogate tool to characterize drug accumulation in AMs. The mean Kp,cell of clarithromycin (81, CV = 51%) and imipramine (963, CV = 54%) were reduced in the presence of NH4Cl by up to 67% and 81%, respectively, suggesting substantial contribution of lysosomal sequestration and intracellular binding in the accumulation of these drugs in human AMs. The in vitro data showed variability in drug accumulation between individual human AM donors due to possible differences in lysosomal abundance, volume, and phospholipid content, which may have important clinical implications. Consideration of drug-acidic phospholipid interactions significantly improved the performance of the in silico models; use of in vitro Kp,cell obtained in the presence of NH4Cl as a surrogate for membrane partitioning (model (2)) captured the variability in clarithromycin and imipramine Kp,cell observed in vitro and showed the best ability to predict correctly positive and negative lysosomotropic properties. The developed mechanistic AM model represents a useful in silico tool to predict lysosomal and cellular drug concentrations based on drug physicochemical data and system specific properties, with potential application to other cell types.
Collapse
Affiliation(s)
- Ayşe Ufuk
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester , Manchester, U.K
| | - Frauke Assmus
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester , Manchester, U.K
| | - Laura Francis
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester , Manchester, U.K
| | - Jonathan Plumb
- Respiratory and Allergy Clinical Research Facility, University Hospital of South Manchester , Manchester, U.K
| | - Valeriu Damian
- Computational Modeling Sciences, DDS, GlaxoSmithKline , Upper Merion, Pennsylvania 19406, United States
| | - Michael Gertz
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester , Manchester, U.K.,Pharmaceutical Sciences, pRED, Roche Innovation Center , Basel, Switzerland
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester , Manchester, U.K
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester , Manchester, U.K
| |
Collapse
|
32
|
Potent Antiedematous and Protective Effects of Ciprofloxacin in Pulmonary Ricinosis. Antimicrob Agents Chemother 2016; 60:7153-7158. [PMID: 27645243 DOI: 10.1128/aac.01696-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/08/2016] [Indexed: 12/15/2022] Open
Abstract
The plant toxin ricin is considered a biological threat agent of concern and is most toxic when inhaled. Pulmonary exposure to a lethal dose of ricin can be redressed by treatment with antiricin antibodies; however, late antitoxin intervention is of limited efficacy. This limitation is associated with overt lung damage, clinically manifested as severe pulmonary inflammation, which develops over time. Increased evidence indicates that ciprofloxacin, a broad-spectrum antimicrobial agent, possesses immunomodulatory properties. Here we demonstrate that while antiricin antibody administration at late hours after intranasal exposure to ricin confers limited protection to mice, highly efficient protection can be achieved by adding ciprofloxacin to the antibody treatment. We further demonstrate that parameters associated with lung injury, in particular, pulmonary proinflammatory cytokine production, neutrophil migration, and edema, are sharply reduced in ricin-intoxicated mice that were treated with ciprofloxacin. The presented data highlight the potential clinical application of ciprofloxacin as a beneficial immunomodulatory agent in the course of ricin intoxication.
Collapse
|
33
|
Goodfriend AC, Welch TR, Thomas CE, Nguyen KT, Johnson RF, Forbess JM. Bacterial sensitivity assessment of multifunctional polymeric coatings for airway stents. J Biomed Mater Res B Appl Biomater 2016; 105:2153-2161. [PMID: 27424845 DOI: 10.1002/jbm.b.33754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/27/2016] [Accepted: 06/26/2016] [Indexed: 11/08/2022]
Abstract
Current interventional technology for pediatric airway obstruction consists of cardiovascular stents and silicon tubes. These devices are composed of permanent materials that have limitations in biocompatibility and mechanical properties that make them controversial for used in pediatrics. Bioresorbable stents offer a temporary intervention that dissolves in the body over time and can serve as a platform for local drug delivery. Here we investigate a novel approach to use an antibiotic, ciprofloxacin, as a polymerization initiator to synthesize poly(ciprofloxacin fumaric acid) (PCFA) and then a second polymer using gadodiamide as an initiator to synthesize poly(gadodiamide ciprofloxacin fumaric acid) (PGCFA). Polymer structure, degradation, thermal properties, and rheological behavior were analyzed. Ciprofloxacin released was determined and polymer degradation extracts were used in bacterial sensitivity assessments with four common airway pathogens. PCFA and PGCFA polymers and drug release properties were compared to our previously published polymer poly(fumaric acid) (PFA). These novel polymers enable new possibilities as coatings for bioresorbable biomedical applications that require antibiotic resistance and imaging capabilities. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2153-2161, 2017.
Collapse
Affiliation(s)
- Amy C Goodfriend
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, 75390-9130
| | - Tré R Welch
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, 75390-9130
| | - Collin E Thomas
- Office of Research, Technology Translation Laboratory, University of Texas at Dallas, Richardson, Texas, 75080-3021
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas Arlington, Arlington, Texas, 76019
| | - Romaine F Johnson
- Department of Otolaryngology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390-9130
| | - Joseph M Forbess
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, 75390-9130
| |
Collapse
|
34
|
Nickel S, Clerkin CG, Selo MA, Ehrhardt C. Transport mechanisms at the pulmonary mucosa: implications for drug delivery. Expert Opin Drug Deliv 2016; 13:667-90. [DOI: 10.1517/17425247.2016.1140144] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Sabrina Nickel
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Caoimhe G. Clerkin
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mohammed Ali Selo
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Faculty of Pharmacy, Kufa University, Al-Najaf, Iraq
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
35
|
Stigliani M, Haghi M, Russo P, Young PM, Traini D. Antibiotic transport across bronchial epithelial cells: Effects of molecular weight, LogP and apparent permeability. Eur J Pharm Sci 2016; 83:45-51. [DOI: 10.1016/j.ejps.2015.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/24/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
|
36
|
Hittinger M, Juntke J, Kletting S, Schneider-Daum N, de Souza Carvalho C, Lehr CM. Preclinical safety and efficacy models for pulmonary drug delivery of antimicrobials with focus on in vitro models. Adv Drug Deliv Rev 2015; 85:44-56. [PMID: 25453270 DOI: 10.1016/j.addr.2014.10.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/30/2014] [Accepted: 10/07/2014] [Indexed: 12/11/2022]
Abstract
New pharmaceutical formulations must be proven as safe and effective before entering clinical trials. Also in the context of pulmonary drug delivery, preclinical models allow testing of novel antimicrobials, reducing risks and costs during their development. Such models allow reducing the complexity of the human lung, but still need to reflect relevant (patho-) physiological features. This review focuses on preclinical pulmonary models, mainly in vitro models, to assess drug safety and efficacy of antimicrobials. Furthermore, approaches to investigate common infectious diseases of the respiratory tract, are emphasized. Pneumonia, tuberculosis and infections occurring due to cystic fibrosis are in focus of this review. We conclude that especially in vitro models offer the chance of an efficient and detailed analysis of new antimicrobials, but also draw attention to the advantages and limitations of such currently available models and critically discuss the necessary steps for their future development.
Collapse
|
37
|
Bharatwaj B, Mohammad AK, Dimovski R, Cassio FL, Bazito RC, Conti D, Fu Q, Reineke J, da Rocha SRP. Dendrimer nanocarriers for transport modulation across models of the pulmonary epithelium. Mol Pharm 2015; 12:826-38. [PMID: 25455560 PMCID: PMC4350608 DOI: 10.1021/mp500662z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The purpose of this study was to
determine the effect of PEGylation
on the interaction of poly(amidoamine) (PAMAM) dendrimer nanocarriers
(DNCs) with in vitro and in vivo models of the pulmonary epithelium. Generation-3 PAMAM dendrimers
with varying surface densities of PEG 1000 Da were synthesized and
characterized. The results revealed that the apical to basolateral
transport of DNCs across polarized Calu-3 monolayers increases with
an increase in PEG surface density. DNC having the greatest number
of PEG groups (n = 25) on their surface traversed
at a rate 10-fold greater than its non-PEGylated counterpart, in spite
of their larger size. This behavior was attributed to a significant
reduction in charge density upon PEGylation. We also observed that
PEGylation can be used to modulate cellular internalization. The total
uptake of PEG-free DNC into polarized Calu-3 monolayers was 12% (w/w)
vs 2% (w/w) for that with 25 PEGs. Polarization is also shown to be
of great relevance in studying this in vitro model
of the lung epithelium. The rate of absorption of DNCs administered
to mice lungs increased dramatically when conjugated with 25 PEG groups,
thus supporting the in vitro results. The exposure
obtained for the DNC with 25PEG was determined to be very high, with
peak plasma concentrations reaching 5 μg·mL–1 within 3 h. The combined in vitro and in
vivo results shown here demonstrate that PEGylation can be
potentially used to modulate the internalization and transport of
DNCs across the pulmonary epithelium. Modified dendrimers thereby
may serve as a valuable platform that can be tailored to target the
lung tissue for treating local diseases, or the circulation, using
the lung as pathway to the bloodstream, for systemic delivery.
Collapse
Affiliation(s)
- Balaji Bharatwaj
- Department of Chemical Engineering and Materials Science, and ‡Department of Pharmaceutical Sciences, Wayne State University , Detroit, Michigan 48202, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Haghi M, Ong HX, Traini D, Young P. Across the pulmonary epithelial barrier: Integration of physicochemical properties and human cell models to study pulmonary drug formulations. Pharmacol Ther 2014; 144:235-52. [DOI: 10.1016/j.pharmthera.2014.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/30/2014] [Indexed: 11/16/2022]
|
39
|
Ong HX, Benaouda F, Traini D, Cipolla D, Gonda I, Bebawy M, Forbes B, Young PM. In vitro and ex vivo methods predict the enhanced lung residence time of liposomal ciprofloxacin formulations for nebulisation. Eur J Pharm Biopharm 2014; 86:83-9. [DOI: 10.1016/j.ejpb.2013.06.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 10/26/2022]
|
40
|
Ong HX, Traini D, Salama R, Anderson SD, Daviskas E, Young PM. The Effects of Mannitol on the Transport of Ciprofloxacin across Respiratory Epithelia. Mol Pharm 2013; 10:2915-24. [DOI: 10.1021/mp400030n] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Hui Xin Ong
- Respiratory
Technology, Woolcock
Institute of Medical Research and Discipline of Pharmacology, Sydney
Medical School, The University of Sydney, NSW 2037, Australia
| | - Daniela Traini
- Respiratory
Technology, Woolcock
Institute of Medical Research and Discipline of Pharmacology, Sydney
Medical School, The University of Sydney, NSW 2037, Australia
| | - Rania Salama
- Respiratory
Technology, Woolcock
Institute of Medical Research and Discipline of Pharmacology, Sydney
Medical School, The University of Sydney, NSW 2037, Australia
| | - Sandra D. Anderson
- Department of Respiratory and
Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South
Wales, Australia
| | - Evangelia Daviskas
- Department of Respiratory and
Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South
Wales, Australia
| | - Paul M. Young
- Respiratory
Technology, Woolcock
Institute of Medical Research and Discipline of Pharmacology, Sydney
Medical School, The University of Sydney, NSW 2037, Australia
| |
Collapse
|
41
|
Hutter V, Chau DYS, Hilgendorf C, Brown A, Cooper A, Zann V, Pritchard DI, Bosquillon C. Digoxin net secretory transport in bronchial epithelial cell layers is not exclusively mediated by P-glycoprotein/MDR1. Eur J Pharm Biopharm 2013; 86:74-82. [PMID: 23816640 DOI: 10.1016/j.ejpb.2013.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 05/24/2013] [Accepted: 06/10/2013] [Indexed: 01/05/2023]
Abstract
The impact of P-glycoprotein (MDR1, ABCB1) on drug disposition in the lungs as well as its presence and activity in in vitro respiratory drug absorption models remain controversial to date. Hence, we characterised MDR1 expression and the bidirectional transport of the common MDR1 probe (3)H-digoxin in air-liquid interfaced (ALI) layers of normal human bronchial epithelial (NHBE) cells and of the Calu-3 bronchial epithelial cell line at different passage numbers. Madin-Darby Canine Kidney (MDCKII) cells transfected with the human MDR1 were used as positive controls. (3)H-digoxin efflux ratio (ER) was low and highly variable in NHBE layers. In contrast, ER=11.4 or 3.0 were measured in Calu-3 layers at a low or high passage number, respectively. These were, however, in contradiction with increased MDR1 protein levels observed upon passaging. Furthermore, ATP depletion and the two MDR1 inhibitory antibodies MRK16 and UIC2 had no or only a marginal impact on (3)H-digoxin net secretory transport in the cell line. Our data do not support an exclusive role of MDR1 in (3)H-digoxin apparent efflux in ALI Calu-3 layers and suggest the participation of an ATP-independent carrier. Identification of this transporter might provide a better understanding of drug distribution in the lungs.
Collapse
Affiliation(s)
- Victoria Hutter
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, UK
| | - David Y S Chau
- Allergy Research Group, School of Molecular Medical Sciences, University of Nottingham, UK
| | | | - Alan Brown
- Immune Modulation Group, Division of Molecular and Cellular Science, School of Pharmacy, University of Nottingham, UK
| | | | | | - David I Pritchard
- Immune Modulation Group, Division of Molecular and Cellular Science, School of Pharmacy, University of Nottingham, UK
| | - Cynthia Bosquillon
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, UK.
| |
Collapse
|