1
|
Chakraborty R, Ray P, Barik S, Banik O, Mahapatra C, Banoth E, Kumar P. A Review on Microengineering of Epithelial Barriers for Biomedical and Pharmaceutical Research. ACS APPLIED BIO MATERIALS 2024; 7:8107-8125. [PMID: 39565389 DOI: 10.1021/acsabm.4c00813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Epithelial tissue forms a barrier around the human body and visceral organs, providing protection, permeation, sensation, and secretion. It is vital for our sustenance as it protects the tissue from harm and injury by restricting the entry of foreign bodies inside. Furthermore, it is a strong barrier to drugs, nutrients, and other essential deliverables. This layer also houses a large consortium of microbes, which thrive in tandem with human tissue, providing several health benefits. Moreover, the complex interplay of the microbiome with the barrier tissue is poorly understood. Therefore, replicating these barrier tissues on microdevices to generate physiological and pathophysiological models has been a huge interest for researchers over the last few decades. The artificially engineered reconstruction of these epithelial cellular barriers on microdevices could help underpin the host-microbe interaction, generating a physiological understanding of the tissue, tissue remodeling, receptor-based selective diffusion, drug testing, and others. In addition, these devices could reduce the burden of animal sacrifices for similar research and minimize the failure rate in drug discovery due to the use of primary human cells and others. This review discusses the nature of the epithelial barrier at different tissue sites, the recent developments in creating engineered barrier models, and their applications in pathophysiology, host-microbe interactions, drug discovery, and cytotoxicity. The review aims to provide know-how and knowledge behind engineered epithelial barrier tissue to bioengineers, biotechnologists, and scientists in allied fields.
Collapse
Affiliation(s)
- Ruchira Chakraborty
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Pragyan Ray
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Swagatika Barik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Oindrila Banik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Chinmaya Mahapatra
- Department of Biotechnology, National Institute of Technology, Raipur-492010 Chhattisgarh, India
| | - Earu Banoth
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Prasoon Kumar
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| |
Collapse
|
2
|
Kumar A, Vaiphei KK, Gulbake A. A nanotechnology driven effectual localized lung cancer targeting approaches using tyrosine kinases inhibitors: Recent progress, preclinical assessment, challenges, and future perspectives. Int J Pharm 2024; 666:124745. [PMID: 39321904 DOI: 10.1016/j.ijpharm.2024.124745] [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: 07/09/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
The higher incidence and mortality rate among all populations worldwide explains the unmet solutions in the treatment of lung cancer. The evolution of targeted therapies using tyrosine kinase inhibitors (TKI) has encouraged anticancer therapies. However, on-target and off-target effects and the development of drug resistance limited the anticancer potential of such targeted biologics. The advances in nanotechnology-driven-TKI embedded carriers that offered a new path toward lung cancer treatment. It is the inhalation route of administration known for its specific, precise, and efficient drug delivery to the lungs. The development of numerous TKI-nanocarriers through inhalation is proof of TKI growth. The future scopes involve using potential lung cancer biomarkers to achieve localized active cancer-targeting strategies. The adequate knowledge of in vitro absorption models usually helps establish better in vitro - in vivo correlation/extrapolation (IVIVC/E) to successfully evaluate inhalable drugs and drug products. The advanced in vitro and ex vivo lung tissue/ organ models offered better tumor heterogeneity, etiology, and microenvironment heterogeneity. The involvement of lung cancer organoids (LCOs), human organ chip models, and genetically modified mouse models (GEMMs) has resolved the challenges associated with conventional in vitro and in vivo models. To access potential inhalation-based drugtherapies, biological barriers, drug delivery, device-based challenges, and regulatory challenges must be encountered associated with their development. A proper understanding of material toxicity, size-based particle deposition at active disease sites, mucociliary clearance, phagocytosis, and the presence of enzymes and surfactants are required to achieve successful inhalational drug delivery (IDD). This article summarizes the future of lung cancer therapy using targeted drug-mediated inhalation using TKI.
Collapse
Affiliation(s)
- Ankaj Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Klaudi K Vaiphei
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India.
| |
Collapse
|
3
|
Liu Z, Anderson JD, Rose NR, Baker EH, Dowell AE, Ryan KJ, Acosta EP, Guimbellot JS. Differential distribution of ivacaftor and its metabolites in plasma and human airway epithelia. Pulm Pharmacol Ther 2024; 86:102314. [PMID: 38964603 DOI: 10.1016/j.pupt.2024.102314] [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] [Received: 04/09/2024] [Revised: 05/24/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Ivacaftor is the first clinically approved monotherapy potentiator to treat CFTR channel dysfunction in people with cystic fibrosis. Ivacaftor (Iva) is a critical component for all current modulator therapies, including highly effective modulator therapies. Clinical studies show that CF patients on ivacaftor-containing therapies present various clinical responses, off-target effects, and adverse reactions, which could be related to metabolites of the compound. In this study, we reported the concentrations of Iva and two of its major metabolites (M1-Iva and M6-Iva) in capillary plasma and estimated M1-Iva and M6-Iva metabolic activity via the metabolite parent ratio in capillary plasma over 12 h. We also used the ratio of capillary plasma versus human nasal epithelial cell concentrations to evaluate entry into epithelial cells in vivo. M6-Iva was rarely detected by LC-MS/MS in epithelial cells from participants taking ivacaftor, although it was detected in plasma. To further explore this discrepancy, we performed in vitro studies, which showed that M1-Iva, but not M6-Iva, readily crossed 16HBE cell membranes. Our studies also suggest that metabolism of these compounds is unlikely to occur in airway epithelia despite evidence of expression of metabolism enzymes. Overall, our data provide evidence that there are differences between capillary and cellular concentrations of these compounds that may inform future studies of clinical response and off-target effects.
Collapse
Affiliation(s)
- Zhongyu Liu
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Justin D Anderson
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Natalie R Rose
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth H Baker
- Department of Sociology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexander E Dowell
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kevin J Ryan
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Edward P Acosta
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennifer S Guimbellot
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
4
|
Ahmed MU, Li J, Zhou Q(T. Tobramycin Reduces Pulmonary Toxicity of Polymyxin B via Inhibiting the Megalin-Mediated Drug Uptake in the Human Lung Epithelial Cells. Pharmaceutics 2024; 16:389. [PMID: 38543283 PMCID: PMC10975719 DOI: 10.3390/pharmaceutics16030389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 04/01/2024] Open
Abstract
Accumulation of polymyxins in the lung epithelial cells can lead to increased mitochondrial oxidative stress and pulmonary toxicity. Aminoglycosides and polymyxins are used, via intravenous and pulmonary delivery, against multidrug-resistant Gram-negative pathogens. Our recent in vitro and animal studies demonstrated that the co-administration of polymyxins with aminoglycosides decreases polymyxin-induced pulmonary toxicity. The aim of this study was to investigate the in vitro transport and uptake of polymyxin B and tobramycin in human lung epithelial Calu-3 cells and the mechanism of reduced pulmonary toxicity resulting from this combination. Transport, intracellular localization, and accumulation of polymyxin B and tobramycin were investigated using doses of 30 mg/L polymyxin B, 70 mg/L tobramycin, and the combination of both. Adding tobramycin significantly (p < 0.05) decreased the polymyxin B-induced cytotoxicity in Calu-3 cells. The combination treatment significantly reduced the transport and uptake of polymyxin B and tobramycin in Calu-3 cells, compared to each drug alone, which supported the reduced pulmonary toxicity. We hypothesized that cellular uptake of polymyxin B and tobramycin shared a common transporter, megalin. We further investigated the megalin expression of Calu-3 cells using confocal microscopy and evaluated megalin activity using a megalin substrate, FITC-BSA, and a megalin inhibitor, sodium maleate. Both polymyxin B and tobramycin significantly inhibited FITC-BSA uptake by Calu-3 cells in a concentration-dependent manner. Sodium maleate substantially inhibited polymyxin B and tobramycin transport and cellular accumulation in the Calu-3 cell monolayer. Our study demonstrated that the significantly reduced uptake of polymyxin B and tobramycin in Calu-3 cells is attributed to the mechanism of action that determines that polymyxin B and tobramycin share a common transporter, megalin.
Collapse
Affiliation(s)
- Maizbha Uddin Ahmed
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Jian Li
- Monash Biomedicine Discovery Institute, Infection Program and Department of Microbiology, Monash University, Clayton, VIC 3800, Australia
| | - Qi (Tony) Zhou
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| |
Collapse
|
5
|
Chaudhary KR, Singh K, Singh C. Recent Updates in Inhalable Drug Delivery System against Various Pulmonary Diseases: Challenges and Future Perspectives. Curr Drug Deliv 2024; 21:1320-1345. [PMID: 37870055 DOI: 10.2174/0115672018265571231011093546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/22/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023]
Abstract
In the current scenario, pulmonary disease has become a prime burden for morbidity and mortality alongside tremendous social and economic crises throughout the world. Numerous conventional drug delivery system and treatment approach targeting the respiratory region has been driven out. However, effective and accurate recovery has not been achieved yet. In this regard, nanotechnological- based inhalable drug delivery strategy including polymeric, lipidic, or metallic-based respirable microparticles plays an indispensable role in circumventing numerous challenges faced during traditional treatment. Excellent aerodynamic performance leads to enhanced lung targetability, reduced dosing frequency and hence systemic toxicities, as well as improved pharmaceutical attributes, and therefore pharmacokinetic profiles are interminable factors associated with nanotechnologicalbased inhalable delivery. In this review, we comprehensively explored recent advancements in nanotechnologically engineered inhalable formulations targeting each of the mentioned pulmonary diseases. Moreover, we systematically discussed possible respiratory or systemic toxicities about the indeterminate and undefined physicochemical characteristics of inhaled particles.
Collapse
Affiliation(s)
- Kabi Raj Chaudhary
- Department of Pharmaceutics, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T Road, Moga, Punjab 142001, India
- Department of Research and Development, United Biotech [P] Ltd. Bagbania, Nalagarh, Solan, Himachal Pradesh, India
| | - Karanvir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Charan Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T Road, Moga, Punjab 142001, India
- Department of Pharmaceutical Sciences HNB Garhwal University, Madhi Chauras, Srinagar, Uttarakhand 246174, India
| |
Collapse
|
6
|
Mutual Effects of Single and Combined CFTR Modulators and Bacterial Infection in Cystic Fibrosis. Microbiol Spectr 2023; 11:e0408322. [PMID: 36625583 PMCID: PMC9927584 DOI: 10.1128/spectrum.04083-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) modulators improve clinical outcomes with varied efficacies in patients with CF. However, the mutual effects of CFTR modulators and bacterial adaptation, together with antibiotic regimens, can influence clinical outcomes. We evaluated the effects of ivacaftor (IVA), lumacaftor (LUM), tezacaftor, elexacaftor, and a three-modulator combination of elexacaftor, tezacaftor, and ivacaftor (ETI), alone or combined with antibiotics, on sequential CF isolates. IVA and ETI showed direct antimicrobial activities against Staphylococcus aureus but not against Pseudomonas aeruginosa. Additive effects or synergies were observed between the CFTR modulators and antibiotics against both species, independently of adaptation to the CF lung. IVA and LUM were the most effective in potentiating antibiotic activity against S. aureus, while IVA and ETI enhanced mainly polymyxin activity against P. aeruginosa. Next, we evaluated the effect of P. aeruginosa pneumonia on the pharmacokinetics of IVA in mice. IVA and its metabolites in plasma, lung, and epithelial lining fluid were increased by P. aeruginosa infection. Thus, CFTR modulators can have direct antimicrobial properties and/or enhance antibiotic activity against initial and adapted S. aureus and P. aeruginosa isolates. Furthermore, bacterial infection impacts airway exposure to IVA, potentially affecting its efficacy. Our findings suggest optimizing host- and pathogen-directed therapies to improve efficacy for personalized treatment. IMPORTANCE CFTR modulators have been developed to correct and/or enhance CFTR activity in patients with specific cystic fibrosis (CF) genotypes. However, it is of great importance to identify potential off-targets of these novel therapies to understand how they affect lung physiology in CF. Since bacterial infections are one of the hallmarks of CF lung disease, the effects (if any) of CFTR modulators on bacteria could impact their efficacy. This work highlights a mutual interaction between CFTR modulators and opportunistic bacterial infections; in particular, it shows that (i) CFTR modulators have an antibacterial activity per se and influence antibiotic efficacy, and (ii) bacterial airway infections affect levels of CFTR modulators in the airways. These findings may help optimize host- and pathogen-directed drug regimens to improve the efficacy of personalized treatment.
Collapse
|
7
|
Silver Nanoparticles Conjugated with Colistin Enhanced the Antimicrobial Activity against Gram-Negative Bacteria. Molecules 2022; 27:molecules27185780. [PMID: 36144516 PMCID: PMC9505607 DOI: 10.3390/molecules27185780] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Colistin is a potent peptide antibiotic that is effective against Gram-negative bacteria. However, nephrotoxicity limited its clinical use. Silver nanoparticles (AgNPs) have gained attention as a potential antimicrobial agent and nanodrug carrier. The conjugation of antibiotics and AgNPs has been found to increase the activity and decrease drug toxicity. In this study, colistin was conjugated with AgNPs (Col-AgNPs), which was confirmed by Fourier-transform infrared (FT-IR) and energy-dispersive X-ray (EDX) spectra. The optimized Col-AgNPs had the proper characteristics, including spherical shape, monodispersity, nanosized particle, high surface charge, and good stability. The powder X-ray diffraction (PXRD) pattern supported the crystallinity of Col-AgNPs and AgNPs. The drug loading of Col-AgNPs was 11.55 ± 0.93%. Col-AgNPs had higher activity against Gram-negative bacteria (Escherichia coli, Klebsiella pneumonia, and Pseudomonas aeruginosa) than AgNPs and colistin. The mechanism of actions of Col-AgNPs involved membrane disruption and genomic DNA damage. The Col-AgNPs and AgNPs were biocompatible with human red blood cells and renal cells at concentrations up to 16 µg/mL. Interestingly, Col-AgNPs exhibited higher cell survival than AgNPs and colistin at 32 µg/mL. Our results revealed that the Col-AgNPs could enhance the antimicrobial activity and cell biocompatibility more than colistin and AgNPs.
Collapse
|
8
|
Guan J, Yuan H, Yu S, Mao S, Tony Zhou Q. Spray dried inhalable ivacaftor co-amorphous microparticle formulations with leucine achieved enhanced in vitro dissolution and superior aerosol performance. Int J Pharm 2022; 622:121859. [PMID: 35643348 PMCID: PMC10017267 DOI: 10.1016/j.ijpharm.2022.121859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 11/28/2022]
Abstract
The present study aimed to develop inhalable powder formulations with both dissolution enhancement and superior aerodynamic properties for potential pulmonary delivery of a poorly water-soluble drug, ivacaftor (IVA). The IVA-leucine (LEU) microparticle formulations were produced by spray drying and the physicochemical, aerosolization and cytotoxicity properties were characterized. Co-amorphous microparticle formulation was formed at the IVA: LEU 3:1 M ratio with hydrogen bond interactions as indicated by Fourier transform infrared spectroscopy (FTIR) results. Dissolution rate of the co-spray dried formulations was significantly improved as compared with the IVA alone or physical mixtures. The co-spray dried formulations exhibited > 80% fine particle fraction (FPF) and > 95% emitted dose percentage (ED) values respectively, with superior physical and aerosolization stability under 40℃ at 75% RH for 30 days. The laser scanning confocal microscopy results demonstrated that more IVA was uptake by Calu-3 cell lines for the co-spray dried formulation. In summary, our results demonstrated that co-spray drying IVA with LEU could achieve enhanced in vitro release and superior aerodynamic properties for pulmonary delivery of IVA.
Collapse
Affiliation(s)
- Jian Guan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Huiya Yuan
- Department of Forensic Analytical Toxicology, China Medical University School of Forensic Medicine, Shenyang, China; Key Laboratory of Forensic Bio-evidence Sciences, Liaoning Province, China; China Medical University Center of Forensic Investigation, China
| | - Shihui Yu
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States; Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
9
|
Spray-freeze-dried inhalable composite microparticles containing nanoparticles of combinational drugs for potential treatment of lung infections caused by Pseudomonas aeruginosa. Int J Pharm 2021; 610:121160. [PMID: 34624446 DOI: 10.1016/j.ijpharm.2021.121160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/27/2021] [Accepted: 10/03/2021] [Indexed: 12/24/2022]
Abstract
The multi-drug resistance of Pseudomonas aeruginosa is an overwhelming cause of terminal and persistent lung infections in cystic fibrosis (CF) patients. Antimicrobial synergy has been shown for colistin and ivacaftor, and our study designed a relatively high drug-loading dry powder inhaler formulation containing nanoparticles of ivacaftor and colistin. The ivacaftor-colistin nanosuspensions (Iva-Col-NPs) were prepared by the anti-solvent method with different stabilizers. Based on the aggregation data, the formulation 7 (F7) with DSPG-PEG-OMe as the stabilizer was selected for further studies. The F7 consisted of ivacaftor, colistin and DSPG-PEG-OMe with a mass ratio of 1:1:1. The F7 powder formulation was developed using the ultrasonic spray-freeze-drying method and exhibited a rough surface with relatively high fine particle fraction values of 61.4 ± 3.4% for ivacaftor and 63.3 ± 3.3% for colistin, as well as superior emitted dose of 97.8 ± 0.3% for ivacaftor and 97.6 ± 0.5% for colistin. The F7 showed very significant dissolution improvement for poorly water soluble ivacaftor than the physical mixture. Incorporating two drugs in a single microparticle with synchronized dissolution and superior aerosol performance will maximize the synergy and bioactivity of those two drugs. Minimal cytotoxicity in Calu-3 human lung epithelial cells and enhanced antimicrobial activity against colistin-resistant P. aeruginosa suggested that our formulation has potential to improve the treatment of CF patients with lung infections.
Collapse
|