1
|
McCartan AJS, Mrsny RJ. In vitro modelling of intramuscular injection site events. Expert Opin Drug Deliv 2024; 21:1155-1173. [PMID: 39126130 DOI: 10.1080/17425247.2024.2388841] [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: 10/18/2023] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Intramuscular (IM) injections deliver a plethora of drugs. The majority of IM-related literature details dissolution and/or pharmacokinetic (PK) studies, using methods with limited assessments of post-injection events that can impact drug fate, and absorption parameters. Food and Drug Association guidelines no longer require preclinical in vivo modeling in the U.S.A. Preclinical animal models fail to correlate with clinical outcomes, highlighting the need to study, and understand, IM drug fate in vitro using bespoke models emulating human IM sites. Post-IM injection events, i.e. underlying processes that influence PK outcomes, remain unacknowledged, complicating the application of in vitro methods in preclinical drug development. Understanding such events could guide approaches to predict and modulate IM drug fate in humans. AREAS COVERED This article reviews challenges in biorelevant IM site modeling (i.e. modeling drug fate outcomes), the value of technologies available for developing IM injectables, methods for studying drug fate, and technologies for training in performing IM administrations. PubMed, Web-of-Science, and Lens databases provided papers published between 2014 and 2024. EXPERT OPINION IM drug research is expanding what injectable therapeutics can achieve. However, post-injection events that influence PK outcomes remain poorly understood. Until addressed, advances in IM drug development will not realize their full potential.
Collapse
Affiliation(s)
- Adam J S McCartan
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath, UK
| | - Randall J Mrsny
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath, UK
| |
Collapse
|
2
|
Mwema A, Muccioli GG, des Rieux A. Innovative drug delivery strategies to the CNS for the treatment of multiple sclerosis. J Control Release 2023; 364:435-457. [PMID: 37926243 DOI: 10.1016/j.jconrel.2023.10.052] [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: 07/05/2023] [Revised: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Disorders of the central nervous system (CNS), such as multiple sclerosis (MS) represent a great emotional, financial and social burden. Despite intense efforts, great unmet medical needs remain in that field. MS is an autoimmune, chronic inflammatory demyelinating disease with no curative treatment up to date. The current therapies mostly act in the periphery and seek to modulate aberrant immune responses as well as slow down the progression of the disease. Some of these therapies are associated with adverse effects related partly to their administration route and show some limitations due to their rapid clearance and inability to reach the CNS. The scientific community have recently focused their research on developing MS therapies targeting different processes within the CNS. However, delivery of therapeutics to the CNS is mainly limited by the presence of the blood-brain barrier (BBB). Therefore, there is a pressing need to develop new drug delivery strategies that ensure CNS availability to capitalize on identified therapeutic targets. Several approaches have been developed to overcome or bypass the BBB and increase delivery of therapeutics to the CNS. Among these strategies, the use of alternative routes of administration, such as the nose-to-brain (N2B) pathway, offers a promising non-invasive option in the scope of MS, as it would allow a direct transport of the drugs from the nasal cavity to the brain. Moreover, the combination of bioactive molecules within nanocarriers bring forth new opportunities for MS therapies, allowing and/or increasing their transport to the CNS. Here we will review and discuss these alternative administration routes as well as the nanocarrier approaches useful to deliver drugs for MS.
Collapse
Affiliation(s)
- Ariane Mwema
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium; Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium.
| | - Anne des Rieux
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium.
| |
Collapse
|
3
|
Pastorin G, Benetti C, Wacker MG. From in vitro to in vivo: A comprehensive guide to IVIVC development for long-acting therapeutics. Adv Drug Deliv Rev 2023; 199:114906. [PMID: 37286087 DOI: 10.1016/j.addr.2023.114906] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/22/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Affiliation(s)
- Giorgia Pastorin
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | - Camillo Benetti
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| |
Collapse
|
4
|
Liang Z, Giles MB, Stenslik MJ, Marsales M, Ormes JD, Seto R, Zhong W. Direct visualization of the drug release process of non-conductive polymeric implants via molecular imaging. Anal Chim Acta 2022; 1230:340395. [DOI: 10.1016/j.aca.2022.340395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/12/2022] [Accepted: 09/11/2022] [Indexed: 11/15/2022]
|
5
|
Villa Nova M, Gan K, Wacker MG. Biopredictive tools for the development of injectable drug products. Expert Opin Drug Deliv 2022; 19:671-684. [PMID: 35603724 DOI: 10.1080/17425247.2022.2081682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Biopredictive release tests are commonly used in the evaluation of oral medicines. They support decision-making in formulation development and allow predictions of the expected in-vivo performances. So far, there is limited experience in the application of these methodologies to injectable drug products. AREAS COVERED Parenteral drug products cover a variety of dosage forms and administration sites including subcutaneous, intramuscular, and intravenous injections. In this area, developing biopredictive and biorelevant methodologies often confronts us with unique challenges and knowledge gaps. Here, we provide a formulation-centric approach and explain the key considerations and workflow when designing biopredictive assays. Also, we outline the key role of computational methods in achieving clinical relevance and put all considerations into context using liposomal nanomedicines as an example. EXPERT OPINION Biopredictive tools are the need of the hour to exploit the tremendous opportunities of injectable drug products. A growing number of biopharmaceuticals such as peptides, proteins, and nucleic acids require different strategies and a better understanding of the influences on drug absorption. Here, our design strategy must maintain the balance of robustness and complexity required for effective formulation development.
Collapse
Affiliation(s)
- Mônica Villa Nova
- State University of Maringá, Department of Pharmacy, Maringá, Paraná, Brazil
| | - Kennard Gan
- National University of Singapore, Department of Pharmacy, Singapore
| | | |
Collapse
|
6
|
Villa Nova M, Lin TP, Shanehsazzadeh S, Jain K, Ng SCY, Wacker R, Chichakly K, Wacker MG. Nanomedicine Ex Machina: Between Model-Informed Development and Artificial Intelligence. Front Digit Health 2022; 4:799341. [PMID: 35252958 PMCID: PMC8894322 DOI: 10.3389/fdgth.2022.799341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Today, a growing number of computational aids and simulations are shaping model-informed drug development. Artificial intelligence, a family of self-learning algorithms, is only the latest emerging trend applied by academic researchers and the pharmaceutical industry. Nanomedicine successfully conquered several niche markets and offers a wide variety of innovative drug delivery strategies. Still, only a small number of patients benefit from these advanced treatments, and the number of data sources is very limited. As a consequence, “big data” approaches are not always feasible and smart combinations of human and artificial intelligence define the research landscape. These methodologies will potentially transform the future of nanomedicine and define new challenges and limitations of machine learning in their development. In our review, we present an overview of modeling and artificial intelligence applications in the development and manufacture of nanomedicines. Also, we elucidate the role of each method as a facilitator of breakthroughs and highlight important limitations.
Collapse
Affiliation(s)
- Mônica Villa Nova
- Department of Pharmacy, State University of Maringá, Maringá, Brazil
| | - Tzu Ping Lin
- Wacker Research Lab, Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Saeed Shanehsazzadeh
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Kinjal Jain
- Wacker Research Lab, Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Samuel Cheng Yong Ng
- Wacker Research Lab, Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | | | | | - Matthias G. Wacker
- Wacker Research Lab, Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
- *Correspondence: Matthias G. Wacker
| |
Collapse
|
7
|
Drug release from in situ forming implants and advances in release testing. Adv Drug Deliv Rev 2021; 178:113912. [PMID: 34363860 DOI: 10.1016/j.addr.2021.113912] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 12/29/2022]
Abstract
In situ forming implants, defined as liquid formulations that generate solid or semisolid depots following administration, have shown a range of advantages in drug delivery. This drug delivery strategy allows localized delivery, sustained drug release over periods of days to months, and is a less invasive option compared to traditional solid implants which typically require surgical implantation. Unfortunately, there are a number of quality control challenges in terms of drug release testing of these delivery systems which is likely to have contributed to the relatively few commercially available in situ forming implant products. This article reviews current marketed in situ forming implant products, FDA guidance on in vitro release testing, and formulation and environmental parameters influencing drug release from in situ forming implants. Formulation considerations for development of biological agents loaded in situ forming implants are also discussed. The advantages and limitations of typically used in vitro release testing methods are summarized. Difficulties in the development of in vitro-in vivo correlations (IVIVCs) for in situ forming implant are discussed. The knowledge presented will be helpful for the development of in situ forming implants, as well as for the development of appropriate in vitro testing methods and IVIVCs.
Collapse
|
8
|
Simulate SubQ: The Methods and the Media. J Pharm Sci 2021; 112:1492-1508. [PMID: 34728176 DOI: 10.1016/j.xphs.2021.10.031] [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: 08/16/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022]
Abstract
For decades, there has been a growing interest in injectable subcutaneous formulations to improve the absorption of drugs into the systemic circulation and to prolong their release over a longer period. However, fluctuations in the blood plasma levels together with bioavailability issues often limit their clinical success. This warrants a closer look at the performance of long-acting depots, for example, and their dependence on the complex interplay between the dosage form and the physiological microenvironment. For this, biopredictive performance testing is used for a thorough understanding of the biophysical processes affecting the absorption of compounds from the injection site in vivo and their simulation in vitro. In the present work, we discuss in vitro methodologies including methods and media developed for the subcutaneous route of administration on the background of the most relevant absorption mechanisms. Also, we highlight some important knowledge gaps and shortcomings of the existing methodologies to provide the reader with a better understanding of the scientific evidence underlying these models.
Collapse
|
9
|
Huang Y, Yu Q, Chen Z, Wu W, Zhu Q, Lu Y. In vitro and in vivo correlation for lipid-based formulations: Current status and future perspectives. Acta Pharm Sin B 2021; 11:2469-2487. [PMID: 34522595 PMCID: PMC8424225 DOI: 10.1016/j.apsb.2021.03.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/03/2021] [Accepted: 01/15/2021] [Indexed: 12/17/2022] Open
Abstract
Lipid-based formulations (LBFs) have demonstrated a great potential in enhancing the oral absorption of poorly water-soluble drugs. However, construction of in vitro and in vivo correlations (IVIVCs) for LBFs is quite challenging, owing to a complex in vivo processing of these formulations. In this paper, we start with a brief introduction on the gastrointestinal digestion of lipid/LBFs and its relation to enhanced oral drug absorption; based on the concept of IVIVCs, the current status of in vitro models to establish IVIVCs for LBFs is reviewed, while future perspectives in this field are discussed. In vitro tests, which facilitate the understanding and prediction of the in vivo performance of solid dosage forms, frequently fail to mimic the in vivo processing of LBFs, leading to inconsistent results. In vitro digestion models, which more closely simulate gastrointestinal physiology, are a more promising option. Despite some successes in IVIVC modeling, the accuracy and consistency of these models are yet to be validated, particularly for human data. A reliable IVIVC model can not only reduce the risk, time, and cost of formulation development but can also contribute to the formulation design and optimization, thus promoting the clinical translation of LBFs.
Collapse
Key Words
- ANN, artificial neural network
- AUC, area under the curve
- Absorption
- BCS, biopharmaceutics classification system
- BE, bioequivalence
- CETP, cholesterol ester transfer protein
- Cmax, peak plasma concentration
- DDS, drug delivery system
- FDA, US Food and Drug Administration
- GI, gastrointestinal
- HLB, hydrophilic–lipophilic balance
- IVIVC, in vitro and in vivo correlation
- IVIVR, in vitro and in vivo relationship
- In silico prediction
- In vitro and in vivo correlations
- LBF, lipid-based formulation
- LCT, long-chain triglyceride
- Lipid-based formulation
- Lipolysis
- MCT, medium-chain triglyceride
- Model
- Oral delivery
- PBPK, physiologically based pharmacokinetic
- PK, pharmacokinetic
- Perspectives
- SCT, short-chain triglyceride
- SEDDS, self-emulsifying drug delivery system
- SGF, simulated gastric fluid
- SIF, simulated intestinal fluid
- SLS, sodium lauryl sulfate
- SMEDDS, self-microemulsifying drug delivery system
- SNEDDS, self-nanoemulsifying drug delivery system
- TIM, TNO gastrointestinal model
- TNO, Netherlands Organization for Applied Scientific Research
- Tmax, time to reach the peak plasma concentration
Collapse
|
10
|
A Bidirectional Permeability Assay for beyond Rule of 5 Compounds. Pharmaceutics 2021; 13:pharmaceutics13081146. [PMID: 34452112 PMCID: PMC8400635 DOI: 10.3390/pharmaceutics13081146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/02/2022] Open
Abstract
Bidirectional permeability measurement with cellular models grown on Transwell inserts is widely used in pharmaceutical research since it not only provides information about the passive permeability of a drug, but also about transport proteins involved in the active transport of drug substances across physiological barriers. With the increasing number of investigative drugs coming from chemical space beyond Lipinski’s Rule of 5, it becomes more and more challenging to provide meaningful data with the standard permeability assay. This is exemplified here by the difficulties we encountered with the cyclic depsipeptides emodepside and its close analogs with molecular weight beyond 1000 daltons and cLogP beyond 5. The aim of this study is to identify potential reasons for these challenges and modify the permeability assays accordingly. With the modified assay, intrinsic permeability and in vitro efflux of depsipeptides could be measured reliably. The improved correlation to in vivo bioavailability and tissue distribution data indicated the usefulness of the modified permeability assay for the in vitro screening of compounds beyond the Rule of 5.
Collapse
|
11
|
An Imaging Toolkit for Physical Characterization of Long-Acting Pharmaceutical Implants. J Pharm Sci 2020; 109:2798-2811. [DOI: 10.1016/j.xphs.2020.05.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
|
12
|
Fodor-Kardos A, Kiss ÁF, Monostory K, Feczkó T. Sustained in vitro interferon-beta release and in vivo toxicity of PLGA and PEG-PLGA nanoparticles. RSC Adv 2020; 10:15893-15900. [PMID: 35493658 PMCID: PMC9052435 DOI: 10.1039/c9ra09928j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/05/2020] [Indexed: 11/21/2022] Open
Abstract
Interferon-beta-1a (IFN-β-1a) can diminish the symptoms of relapsing-remitting multiple sclerosis. Herein, we prepared sustained drug delivery IFN-β-1a-loaded nanoparticles by a double emulsion solvent evaporation method. Bovine serum albumin (BSA) model drug was used to optimize the preparation of nanoparticles composed of four types of poly(lactic-co-glycolic acid) (PLGA) polymers and two pegylated PLGA (PEG-PLGA) polymers. Via optimization, selected PLGA and PEG-PLGA polymers were able to entrap IFN-β-1a with high encapsulation efficiency (>95%) and low size (145 nm and 163 nm, respectively). In vitro release kinetics of BSA and IFN-β showed similar tendency for PLGA and PEG-PLGA nanoparticles, respectively. Although the drug loaded nanoparticles did not show toxicity in hepatocyte cells, mild toxic effects such as pale kidney and pyelectasis were observed in the in vivo studies.
Collapse
Affiliation(s)
- Andrea Fodor-Kardos
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences Magyar Tudósok Körútja 2 H-1117 Budapest Hungary +36-88-624000 ext. 3508
- Research Institute of Biomolecular and Chemical Engineering, University of Pannonia Egyetem u. 10 H-8200 Veszprém Hungary
| | - Ádám Ferenc Kiss
- Institute of Enzymology, Research Centre for Natural Sciences Magyar Tudósok Körútja 2 H-1117 Budapest Hungary
| | - Katalin Monostory
- Institute of Enzymology, Research Centre for Natural Sciences Magyar Tudósok Körútja 2 H-1117 Budapest Hungary
| | - Tivadar Feczkó
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences Magyar Tudósok Körútja 2 H-1117 Budapest Hungary +36-88-624000 ext. 3508
- Research Institute of Biomolecular and Chemical Engineering, University of Pannonia Egyetem u. 10 H-8200 Veszprém Hungary
| |
Collapse
|
13
|
Rasool A, Ata S, Islam A, Rizwan M, Azeem MK, Mehmood A, Khan RU, Qureshi AUR, Mahmood HA. Kinetics and controlled release of lidocaine from novel carrageenan and alginate-based blend hydrogels. Int J Biol Macromol 2020; 147:67-78. [PMID: 31926227 DOI: 10.1016/j.ijbiomac.2020.01.073] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 12/30/2019] [Accepted: 01/07/2020] [Indexed: 12/13/2022]
Abstract
The controlled release of drug from drug carrier has been a point of concern for the researchers to ensure the bioavailability of drug with reduced side effects. The formulation in this study is based upon biopolymers; carrageenan (CG), sodium alginate (SA) and various molecular weights of polyethylene glycol (PEG), cross-linked with (3-Aminopropyl)triethoxysilane, APTES for the sustained release of model drug (lidocaine). The physicochemical properties of the formulated hydrogel blends include bonding pattern (using Fourier Transform Infra-Red Spectroscopy (FTIR), Thermogravimetric analysis (TGA), X-ray diffraction (XRD), swelling study, antimicrobial activity and morphology of hydrogel films was analyzed by scanning electron microscopy (SEM). The as-prepared hydrogels show an improved cell compatibility against 3T3 cell line as well as cell proliferation and kinetics of drug release showed that these hydrogels are potential for controlled release of lidocaine, a numbing agent. GAP 60 exhibited maximum swelling percent (910%) and was employed to load the drug. By using in vitro model, the drug release was studied in PBS solution. Non-Fickian and other kinetic models (Zero order, Higuchi, Hixson, Korsmeyer Peppas and Baker-Lonsdale) for diffusion were followed in results. The improved properties showed that the formulated hydrogels can easily be used for the sustain drug release studies.
Collapse
Affiliation(s)
- Atta Rasool
- Institute of Chemistry, University of the Punjab, 54590 Lahore, Pakistan; Department of Polymer Engineering and Technology, University of the Punjab, 54590 Lahore, Pakistan
| | - Sadia Ata
- Institute of Chemistry, University of the Punjab, 54590 Lahore, Pakistan.
| | - Atif Islam
- Department of Polymer Engineering and Technology, University of the Punjab, 54590 Lahore, Pakistan.
| | - Muhammad Rizwan
- Department of Chemistry, The University of Lahore, Lahore, Pakistan
| | - Muhammad Khalid Azeem
- Department of Polymer Engineering and Technology, University of the Punjab, 54590 Lahore, Pakistan
| | - Azra Mehmood
- Centre of Excellence in Molecular Biology (CEMB), University of Punjab, Lahore, Pakistan
| | - Rafi Ullah Khan
- Department of Polymer Engineering and Technology, University of the Punjab, 54590 Lahore, Pakistan
| | | | - Hafiz Arshad Mahmood
- Department of Polymer Engineering and Technology, University of the Punjab, 54590 Lahore, Pakistan
| |
Collapse
|
14
|
Marques MR, Choo Q, Ashtikar M, Rocha TC, Bremer-Hoffmann S, Wacker MG. Nanomedicines - Tiny particles and big challenges. Adv Drug Deliv Rev 2019; 151-152:23-43. [PMID: 31226397 DOI: 10.1016/j.addr.2019.06.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 02/08/2023]
Abstract
After decades of research, nanotechnology has been used in a broad array of biomedical products including medical devices, drug products, drug substances, and pharmaceutical-grade excipients. But like many great achievements in science, there is a fine balance between the risks and opportunities of this new technology. Some materials and surface structures in the nanosize range can exert unexpected toxicities and merit a more detailed safety assessment. Regulatory agencies such as the United States Food and Drug Administration or the European Medicines Agency have started dealing with the potential risks posed by nanomaterials. Considering that a thorough characterization is one of the key aspects of controlling such risks this review presents the regulatory background of nanosafety assessment and provides some practical advice on how to characterize nanomaterials and drug formulations. Further, the challenges of how to maintain and monitor pharmaceutical quality through a highly complex production processes will be discussed.
Collapse
|
15
|
Nothnagel L, Wacker MG. How to measure release from nanosized carriers? Eur J Pharm Sci 2018; 120:199-211. [PMID: 29751101 DOI: 10.1016/j.ejps.2018.05.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/13/2018] [Accepted: 05/07/2018] [Indexed: 12/28/2022]
Abstract
Novel drug delivery systems exhibit great potential in the formulation of poorly soluble compounds but have also been applied to reduce side effects of highly active drug molecules. Despite all efforts, there are only few technologies available to investigate the in vitro release of next-generation nanotherapeutics. In the following, different approaches for testing the drug release from nanoparticles in the fields of formulation development and quality control will be discussed. A variety of methods is available, starting from dialysis-based equipment, in situ measurements, flow-through devices and sample and separate setups. If possible, these methods should enable a more rapid formulation development and quality control of nanosized carriers as well as improve the prediction of in vivo performance and clinical outcomes.
Collapse
Affiliation(s)
- Lisa Nothnagel
- Department of Pharmaceutical Technology and Nanosciences, Fraunhofer-Institute for Molecular Biology and Applied Ecology (IME), 60438 Frankfurt am Main, Germany
| | - Matthias G Wacker
- Department of Pharmaceutical Technology and Nanosciences, Fraunhofer-Institute for Molecular Biology and Applied Ecology (IME), 60438 Frankfurt am Main, Germany; Institute of Pharmaceutical Technology, Goethe University, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
16
|
Hofmann MCJ, Schmidt M, Arne O, Geisslinger G, Parnham MJ, de Bruin NMWJ. Non-invasive bioluminescence imaging as a standardized assessment measure in mouse models of dermal inflammation. J Dermatol Sci 2018; 91:S0923-1811(18)30187-7. [PMID: 29706245 DOI: 10.1016/j.jdermsci.2018.04.013] [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: 01/24/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Myeloperoxidase is used as a marker and diagnostic tool for inflammatory processes. Hypochlorous acid produced by myeloperoxidase oxidizes luminol to produce light. By injecting luminol into experimental animals, inflammatory processes can be tracked in real-time by bioluminescence imaging (BLI). OBJECTIVE We aimed to establish BLI as a standardized assessment measure in three mouse models of dermal inflammation. METHODS Oxazolone-induced delayed-type-hypersensitivity (DTH) (acute), a model for dermatitis, imiquimod (IMQ) (sub-chronic) model for psoriasis and the (chronic) bleomycin model for scleroderma were used. In the first two models, dexamethasone and clobetasol, respectively, were used as reference compounds. In all cases, classical readouts such as dermal swelling, severity scores and histological analyses were compared with in- vivo bioluminescence. RESULTS In DTH, bioluminescence peaked earlier than ear swelling, reflecting early cell infiltration. Dexamethasone blocked both ear swelling and bioluminescence. In the IMQ model, bioluminescence closely reflected the psoriasis scores and histology and revealed a relapse-remitting course of the disease. Clobetasol partially decreased the disease severity. After stopping IMQ and clobetasol treatment, BLI adopted a rhythmic pattern during resolution. Bleomycin induced an increase in bioluminescence and in collagen thickness. BLI revealed a time-course of the effects of bleomycin that was not reflected by histology alone. CONCLUSION For drug discovery and translational purposes, it is important that disease processes be tracked in vivo and possibly over a long period. We conclude that BLI is a valuable and reliable method for in-vivo measurement of dermal inflammation and potentially for inflammation resolution.
Collapse
Affiliation(s)
- Martine Catharina Josephine Hofmann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| | - Mike Schmidt
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Olga Arne
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Michael John Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Natasja Maria Wilhelmina Johanna de Bruin
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
17
|
Hernandez C, Exner AA. Predicting in vivo behavior of injectable, in situ-forming drug-delivery systems. Ther Deliv 2017; 8:479-483. [PMID: 28350230 PMCID: PMC10072068 DOI: 10.4155/tde-2017-0007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Christopher Hernandez
- Department of Radiology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| |
Collapse
|