1
|
Padovano F, Villa C. The development of drug resistance in metastatic tumours under chemotherapy: An evolutionary perspective. J Theor Biol 2024; 595:111957. [PMID: 39369787 DOI: 10.1016/j.jtbi.2024.111957] [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: 07/02/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
We present a mathematical model of the evolutionary dynamics of a metastatic tumour under chemotherapy, comprising non-local partial differential equations for the phenotype-structured cell populations in the primary tumour and its metastasis. These equations are coupled with a physiologically-based pharmacokinetic model of drug administration and distribution, implementing a realistic delivery schedule. The model is carefully calibrated from the literature, focusing on BRAF-mutated melanoma treated with Dabrafenib as a case study. By means of long-time asymptotic and global sensitivity analyses, as well as numerical simulations, we explore the impact of cell migration from the primary to the metastatic site, physiological aspects of the tumour tissues and drug dose on the development of chemoresistance and treatment efficacy. Our findings provide a possible explanation for empirical evidence indicating that chemotherapy may foster metastatic spread and that metastases may be less impacted by the chemotherapeutic agent.
Collapse
Affiliation(s)
- Federica Padovano
- Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR 7598, 4 place Jussieu, 75005 Paris, France.
| | - Chiara Villa
- Sorbonne Université, CNRS, Université de Paris, Inria, Laboratoire Jacques-Louis Lions UMR 7598, 4 place Jussieu, 75005 Paris, France.
| |
Collapse
|
2
|
Stella J, Abdelaal MAME, Kamal MAM, Shehu K, Alhayek A, Haupenthal J, Hirsch AK, Schneider M. Spray drying of a zinc complexing agent for inhalation therapy of pulmonary fibrosis. Eur J Pharm Sci 2024; 202:106891. [PMID: 39233259 DOI: 10.1016/j.ejps.2024.106891] [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: 04/17/2024] [Revised: 08/24/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
Pulmonary fibrosis, a disabling lung disease, results from the fibrotic transformation of lung tissue. This fibrotic transformation leads to a deterioration of lung capacity, resulting in significant respiratory distress and a reduction in overall quality of life. Currently, the frontline treatment of pulmonary fibrosis remains limited, focusing primarily on symptom relief and slowing disease progression. Bacterial infections with Pseudomonas aeruginosa are contributing to a severe progression of idiopathic pulmonary fibrosis. Phytic acid, a natural chelator of zinc, which is essential for the activation of metalloproteinase enzymes involved in pulmonary fibrosis, shows potential inhibition of LasB, a virulence factor in P. aeruginosa, and mammalian metalloproteases (MMPs). In addition, phytic acid has anti-inflammatory properties believed to result from its ability to capture free radicals, inhibit certain inflammatory enzymes and proteins, and reduce the production of inflammatory cytokines, key signaling molecules that promote inflammation. To achieve higher local concentrations in the deep lung, phytic acid was spray dried into an inhalable powder. Challenges due to its hygroscopic and low melting (25 °C) nature were mitigated by converting it to sodium phytate to improve crystallinity and powder characteristics. The addition of leucine improved aerodynamic properties and reduced agglomeration, while mannitol served as carrier matrix. Size variation was achieved by modifying process parameters and were evaluated by tools such as the Next Generation Impactor (NGI), light diffraction methods, and scanning electron microscopy (SEM). An inhibition assay for human MMP-1 (collagenase-1) and MMP-2 (gelatinase A) allowed estimation of the biological effect on tissue remodeling enzymes. The activity was also assessed with respect to inhibition of bacterial LasB. The formulated phytic acid demonstrated an IC50 of 109.7 µg/mL for LasB with viabilities > 80 % up to 188 µg/mL on A549 cells. Therefore, inhalation therapy with phytic acid-based powder shows promise as a treatment for early-stage Pseudomonas-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Justin Stella
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, Saarbrücken 66123, Germany
| | | | - Mohamed Ashraf Mostafa Kamal
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, Saarbrücken 66123, Germany; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8 1, Saarbrücken 66123, Germany
| | - Kristela Shehu
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, Saarbrücken 66123, Germany; INM - Leibniz Institute for New Materials, Saarbrücken 66123, Germany
| | - Alaa Alhayek
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8 1, Saarbrücken 66123, Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8 1, Saarbrücken 66123, Germany
| | - Anna K Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8 1, Saarbrücken 66123, Germany; Department of Pharmacy, Medicinal Chemistry, Saarland University, Saarbrücken 66123, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, Saarbrücken 66123, Germany.
| |
Collapse
|
3
|
Hickey AJ, Maloney SE, Kuehl PJ, Phillips JE, Wolff RK. Practical Considerations in Dose Extrapolation from Animals to Humans. J Aerosol Med Pulm Drug Deliv 2024; 37:77-89. [PMID: 38237032 DOI: 10.1089/jamp.2023.0041] [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: 04/21/2024] Open
Abstract
Animal studies are an important component of drug product development and the regulatory review process since modern practices have been in place, for almost a century. A variety of experimental systems are available to generate aerosols for delivery to animals in both liquid and solid forms. The extrapolation of deposited dose in the lungs from laboratory animals to humans is challenging because of genetic, anatomical, physiological, pharmacological, and other biological differences between species. Inhaled drug delivery extrapolation requires scrutiny as the aerodynamic behavior, and its role in lung deposition is influenced not only by the properties of the drug aerosol but also by the anatomy and pulmonary function of the species in which it is being evaluated. Sources of variability between species include the formulation, delivery system, and species-specific biological factors. It is important to acknowledge the underlying variables that contribute to estimates of dose scaling between species.
Collapse
Affiliation(s)
- Anthony J Hickey
- Department of Technology Advancement and Commercialization, RTI International, Research Triangle Park, North Carolina, USA
| | - Sara E Maloney
- Department of Technology Advancement and Commercialization, RTI International, Research Triangle Park, North Carolina, USA
| | - Phillip J Kuehl
- Division: Scientific Core Laboratories; Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Jonathan E Phillips
- Amgen, Inc., Inflammation Discovery Research, Thousand Oaks, California, USA
| | | |
Collapse
|
4
|
Xia W, Kolli AR, Kuczaj AK, Szostak J, Lam S, Toh WW, Purwanti A, Tan WT, Ng R, Phillips B, Peitsch MC, Hoeng J. Aerosol delivery and spatiotemporal tissue distribution of hydroxychloroquine in rat lung. Eur J Pharm Sci 2024; 194:106693. [PMID: 38184016 DOI: 10.1016/j.ejps.2024.106693] [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: 11/14/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/08/2024]
Abstract
Inhalation enables the delivery of drugs directly to the lung, increasing the retention for prolonged exposure and maximizing the therapeutic index. However, the differential regional lung exposure kinetics and systemic pharmacokinetics are not fully known, and their estimation is critical for pulmonary drug delivery. The study evaluates the pharmacokinetics of hydroxychloroquine in different regions of the respiratory tract for multiple routes of administration. We also evaluated the influence of different inhaled formulations on systemic and lung pharmacokinetics by identifying suitable nebulizers followed by early characterization of emitted aerosol physicochemical properties. The salt- and freebase-based formulations required different nebulizers and generated aerosol with different physicochemical properties. An administration of hydroxychloroquine by different routes resulted in varied systemic and lung pharmacokinetics, with oral administration resulting in low tissue concentrations in all regions of the respiratory tract. A nose-only inhalation exposure resulted in higher and sustained lung concentrations of hydroxychloroquine with a lung parenchyma-to-blood ratio of 386 after 1440 min post-exposure. The concentrations of hydroxychloroquine in different regions of the respiratory tract (i.e., nasal epithelium, larynx, trachea, bronchi, and lung parenchyma) varied over time, indicating different retention kinetics. The spatiotemporal distribution of hydroxychloroquine in the lung is different due to the heterogeneity of cell types, varying blood perfusion rate, clearance mechanisms, and deposition of inhaled aerosol along the respiratory tract. In addition to highlighting the varied lung physiology, these results demonstrate the ability of the lung to retain increased levels of inhaled lysosomotropic drugs. Such findings are critical for the development of future inhalation-based therapeutics, aiming to optimize target site exposure, enable precision medicine, and ultimately enhance clinical outcomes.
Collapse
Affiliation(s)
- Wenhao Xia
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Aditya R Kolli
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchâtel CH-2000, Switzerland.
| | - Arkadiusz K Kuczaj
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchâtel CH-2000, Switzerland
| | - Justyna Szostak
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchâtel CH-2000, Switzerland
| | - Sharon Lam
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Wei Wen Toh
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Asef Purwanti
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Wei Teck Tan
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Raymond Ng
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchâtel CH-2000, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchâtel CH-2000, Switzerland
| |
Collapse
|
5
|
Himstedt A, Braun C, Wicha SG, Borghardt JM. Understanding the suitability of established antibiotics for oral inhalation from a pharmacokinetic perspective: an integrated model-based investigation based on rifampicin, ciprofloxacin and tigecycline in vivo data. J Antimicrob Chemother 2022; 77:2922-2932. [PMID: 35904005 DOI: 10.1093/jac/dkac240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/16/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Treating pulmonary infections by administering drugs via oral inhalation represents an attractive alternative to usual routes of administration. However, the local concentrations after inhalation are typically not known and the presumed benefits are derived from experiences with drugs specifically optimized for inhaled administration. OBJECTIVES A physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) model was developed to elucidate the pulmonary PK for ciprofloxacin, rifampicin and tigecycline and link it to bacterial PK/PD models. An exemplary sensitivity analysis was performed to potentially guide drug optimization regarding local efficacy for inhaled antibiotics. METHODS Detailed pulmonary tissue, endothelial lining fluid and systemic in vivo drug concentration-time profiles were simultaneously measured for all drugs in rats after intravenous infusion. Using this data, a PBPK/PD model was developed, translated to humans and adapted for inhalation. Simulations were performed comparing potential benefits of oral inhalation for treating bronchial infections, covering intracellular pathogens and bacteria residing in the bronchial epithelial lining fluid. RESULTS The PBPK/PD model was able to describe pulmonary PK in rats. Often applied optimization parameters for orally inhaled drugs (e.g. high systemic clearance and low oral bioavailability) showed little influence on efficacy and instead mainly increased pulmonary selectivity. Instead, low permeability, a high epithelial efflux ratio and a pronounced post-antibiotic effect represented the most impactful parameters to suggest a benefit of inhalation over systemic administration for locally acting antibiotics. CONCLUSIONS The present work might help to develop antibiotics for oral inhalation providing high pulmonary concentrations and fast onset of exposure coupled with lower systemic drug concentrations.
Collapse
Affiliation(s)
- Anneke Himstedt
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany.,Research DMPK, Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Clemens Braun
- Research DMPK, Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Sebastian Georg Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Jens Markus Borghardt
- Research DMPK, Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
6
|
Himstedt A, Borghardt JM, Wicha SG. Inferring pulmonary exposure based on clinical PK data: accuracy and precision of model-based deconvolution methods. J Pharmacokinet Pharmacodyn 2021; 49:135-149. [PMID: 34585333 PMCID: PMC8940815 DOI: 10.1007/s10928-021-09780-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 09/01/2021] [Indexed: 12/03/2022]
Abstract
Determining and understanding the target-site exposure in clinical studies remains challenging. This is especially true for oral drug inhalation for local treatment, where the target-site is identical to the site of drug absorption, i.e., the lungs. Modeling and simulation based on clinical pharmacokinetic (PK) data may be a valid approach to infer the pulmonary fate of orally inhaled drugs, even without local measurements. In this work, a simulation-estimation study was systematically applied to investigate five published model structures for pulmonary drug absorption. First, these models were compared for structural identifiability and how choosing an inadequate model impacts the inference on pulmonary exposure. Second, in the context of the population approach both sequential and simultaneous parameter estimation methods after intravenous administration and oral inhalation were evaluated with typically applied models. With an adequate model structure and a well-characterized systemic PK after intravenous dosing, the error in inferring pulmonary exposure and retention times was less than twofold in the majority of evaluations. Whether a sequential or simultaneous parameter estimation was applied did not affect the inferred pulmonary PK to a relevant degree. One scenario in the population PK analysis demonstrated biased pulmonary exposure metrics caused by inadequate estimation of systemic PK parameters. Overall, it was demonstrated that empirical modeling of intravenous and inhalation PK datasets provided robust estimates regarding accuracy and bias for the pulmonary exposure and pulmonary retention, even in presence of the high variability after drug inhalation.
Collapse
Affiliation(s)
- Anneke Himstedt
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstrasse 45, 20146, Hamburg, Germany.,Research DMPK, Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Jens Markus Borghardt
- Research DMPK, Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Sebastian Georg Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstrasse 45, 20146, Hamburg, Germany.
| |
Collapse
|
7
|
Michelet R, Ursino M, Boulet S, Franck S, Casilag F, Baldry M, Rolff J, van Dyk M, Wicha SG, Sirard JC, Comets E, Zohar S, Kloft C. The Use of Translational Modelling and Simulation to Develop Immunomodulatory Therapy as an Adjunct to Antibiotic Treatment in the Context of Pneumonia. Pharmaceutics 2021; 13:601. [PMID: 33922017 PMCID: PMC8143524 DOI: 10.3390/pharmaceutics13050601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
The treatment of respiratory tract infections is threatened by the emergence of bacterial resistance. Immunomodulatory drugs, which enhance airway innate immune defenses, may improve therapeutic outcome. In this concept paper, we aim to highlight the utility of pharmacometrics and Bayesian inference in the development of immunomodulatory therapeutic agents as an adjunct to antibiotics in the context of pneumonia. For this, two case studies of translational modelling and simulation frameworks are introduced for these types of drugs up to clinical use. First, we evaluate the pharmacokinetic/pharmacodynamic relationship of an experimental combination of amoxicillin and a TLR4 agonist, monophosphoryl lipid A, by developing a pharmacometric model accounting for interaction and potential translation to humans. Capitalizing on this knowledge and associating clinical trial extrapolation and statistical modelling approaches, we then investigate the TLR5 agonist flagellin. The resulting workflow combines expert and prior knowledge on the compound with the in vitro and in vivo data generated during exploratory studies in order to construct high-dimensional models considering the pharmacokinetics and pharmacodynamics of the compound. This workflow can be used to refine preclinical experiments, estimate the best doses for human studies, and create an adaptive knowledge-based design for the next phases of clinical development.
Collapse
Affiliation(s)
- Robin Michelet
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| | - Moreno Ursino
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, Université de Paris, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, F-75019 Paris, France;
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
| | - Sandrine Boulet
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Sebastian Franck
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Fiordiligie Casilag
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Mara Baldry
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Jens Rolff
- Department of Evolutionary Biology, Institute of Biology, Freie Universitaet Berlin, 14195 Berlin, Germany;
| | - Madelé van Dyk
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia;
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Jean-Claude Sirard
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Emmanuelle Comets
- INSERM, University Rennes-1, CIC 1414, F-35000 Rennes, France;
- INSERM, IAME, Université de Paris, F-75006 Paris, France
| | - Sarah Zohar
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Charlotte Kloft
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| |
Collapse
|
8
|
Yıldız-Peköz A, Ehrhardt C. Advances in Pulmonary Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12100911. [PMID: 32977672 PMCID: PMC7598662 DOI: 10.3390/pharmaceutics12100911] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 12/27/2022] Open
Abstract
Pulmonary drug delivery represents an attractive, non-invasive administration option. In addition to locally acting drugs, molecules that are intended to produce systemic effects can be delivered via the pulmonary route. Several factors need to be considered in the context of delivering drugs to or via the lungs—in addition to the drug itself, its formulation into an appropriate inhalable dosage form of sufficient stability is critical. It is also essential that this formulation is paired with a suitable inhaler device, which generates an aerosol of a particle/droplet size that ensures deposition in the desired region of the respiratory tract. Lastly, the patient’s (patho-) physiology and inhalation manoeuvre are of importance. This Special Issue brings together recent advances in the areas of inhalation device testing, aerosol formulation development, use of in vitro and in silico models in pulmonary drug deposition and drug disposition studies, and pulmonary delivery of complex drugs, such as vaccines, antibiotics and peptides, to or via the lungs.
Collapse
Affiliation(s)
- Ayca Yıldız-Peköz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, İstanbul University, İstanbul 34116, Turkey;
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Correspondence: ; Tel.: +353-1-896-2441
| |
Collapse
|