1
|
Dangkoub F, Bemani Naeini M, Akar S, Badiee A, Jaafari MR, Sankian M, Tafaghodi M, Mousavi Shaegh SA. Preparation of atorvastatin calcium-loaded liposomes using thin-film hydration and coaxial micromixing methods: A comparative study. Int J Pharm X 2024; 8:100309. [PMID: 39697814 PMCID: PMC11653151 DOI: 10.1016/j.ijpx.2024.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Development of techniques to produce nanoformulations in a controlled and reproducible manner is of great importance for research, clinical trials, and industrial scale-up. This research aimed to introduce a cost-effective micromixing approach for the nanoassembly of liposomes and compared with thin-film hydration (TFH) method. Numerical simulations and design of experiments (DOE) by response surface methodology (RSM) were used to evaluate the effects of input parameters on liposome properties, aiming to identify optimal conditions. Anionic liposomes without or with atorvastatin calcium (ATC) produced using TFH and the micromixing methods showed similar characteristics in size (150-190 nm), PDI (<0.2), and zeta potential (-50 to -60 mV). Both methods achieved about 70 % encapsulation efficiency with similar drug release profile for ATC-containing liposomes. Analysis of stability and DSC thermograms revealed comparable outcomes for liposomes prepared using both techniques. Nanoliposomes produced via both approaches indicated similar in vitro biological performance regarding cellular uptake and cell viability. The micromixing approach presented an alternative method to produce nanoliposomes in a one-step manner with high controllability and reproducibility without requiring specialized equipment. Compatibility of the micromixer with various solvents, including those detrimental to conventional microfluidic materials like PDMS and thermoplastics, enables exploration of a wide range of formulations.
Collapse
Affiliation(s)
- Faezeh Dangkoub
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehri Bemani Naeini
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Akar
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Tafaghodi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Unit of Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Gu Y, Jin L, Wang L, Ma X, Tian M, Sohail A, Wang J, Wang D. Preparation of Baicalin Liposomes Using Microfluidic Technology and Evaluation of Their Antitumor Activity by a Zebrafish Model. ACS OMEGA 2024; 9:41289-41300. [PMID: 39398129 PMCID: PMC11465266 DOI: 10.1021/acsomega.4c03356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024]
Abstract
Baicalin (BCL), a well-known flavonoid molecule, has numerous therapeutic applications. However, its low water solubility and bioavailability limit its applicability. Microfluidics is a new method for liposome preparation that provides efficient and rapid control of the process, improving the stability and controllability. This study used microfluidic techniques to create baicalin liposomes (BCL-LPs), first screening for optimal total flow rates (TFR) and flow rate ratios (FRR), and then optimizing the phospholipid concentration, phospholipid-to-cholesterol ratio, and Tween-80 concentration using univariate and response surface methodology approaches. The study found that the ideal phospholipid content was 9.5%, the phospholipid-to-cholesterol ratio was 9:1 (w:w), and the Tween-80 concentration was 15%. BCL-LPs achieved 95.323% ± 0.481% encapsulation efficiency under the optimum circumstances. Characterization indicated that the BCL-LPs were spherical and uniform in size, with a mean diameter of 62.32 nm ± 0.42, a polydispersity index of 0.092 ± 0.009, and a zeta potential of -25.000 mV ± 0.216. In vitro experiments found that BCL-LPs had a better slow-release effect and stability than the BCL monomer. In zebrafish bioassays, BCL-LPs performed better than BCL monomer in terms of biological activity and bioavailability. The established method provided a feasible medicine delivery platform for BCL and could apply for the transport and encapsulation of more natural compounds, expanding the applications of drug delivery systems in healthcare and cancer therapies.
Collapse
Affiliation(s)
- Yuhao Gu
- School
of Light Industry Science and Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Heze
Branch of Qilu University of Technology (Shandong Academy of Sciences), Heze 274000, China
| | - Liqiang Jin
- School
of Light Industry Science and Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Li Wang
- Jinan
Vocational College of Engineering Department: Youth League Committee, Jinan 250200, China
| | - Xianzheng Ma
- Heze
Branch of Qilu University of Technology (Shandong Academy of Sciences), Heze 274000, China
| | - Mingfa Tian
- Heze
Branch of Qilu University of Technology (Shandong Academy of Sciences), Heze 274000, China
| | | | - Jianchun Wang
- Shandong
Giant E-Tech Co., Ltd., Jinan 250102, China
| | - Daijie Wang
- Heze
Branch of Qilu University of Technology (Shandong Academy of Sciences), Heze 274000, China
| |
Collapse
|
3
|
Yu Y, Li S, Kong L, Du Y, Liu Y, Zang J, Guo R, Zhang L, Zhao Z, Ju R, Li X. Development of a brain-targeted nano drug delivery system to enhance the treatment of neurodegenerative effects of resveratrol. J Liposome Res 2024; 34:435-451. [PMID: 38032385 DOI: 10.1080/08982104.2023.2290050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023]
Abstract
As the aging population continues to increase, aging-related inflammation, oxidative stress, and neurodegenerative diseases have become serious global health threats. Resveratrol, a star molecule in natural polyphenols, has been widely reported to have physiological activities such as anti-aging, anti-inflammatory, antioxidant, and neuroprotection. However, its poor water solubility, rapid metabolism, low bioavailability and poor targeting ability, which limits its application. Accordingly, a brain-targeted resveratrol liposome (ANG-RES-LIP) was developed to solve these issues. Experimental results showed that ANG-RES-LIP has a uniform size distribution, good biocompatibility, and a drug encapsulation rate of over 90%. Furthermore, in vitro cell experiments showed that the modification of the targeting ligand ANG significantly increased the capability of RES to cross the BBB and neuronal uptake. Compared with free RES, ANG-RES-LIP demonstrated stronger antioxidant activity and the ability to rescue oxidatively damaged cells from apoptosis. Additionally, ANG-RES-LIP showed the ability to repair damaged neuronal mitochondrial membrane potential. In vivo experiments further demonstrated that ANG-RES-LIP improved cognitive function by reducing oxidative stress and inflammation levels in the brains of aging model mice, repairing damaged neurons and glial cells, and increasing brain-derived neurotrophic factor. In summary, this study not only provides a new method for further development and application of resveratrol but also a promising strategy for preventing and treating age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Shutong Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Yumeng Du
- Department of Pharmaceutical Engineering, Beijing Institute of Petrochemical Technology, Beijing, China
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Ziyue Zhao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| | - Ruijun Ju
- Department of Pharmaceutical Engineering, Beijing Institute of Petrochemical Technology, Beijing, China
| | - Xuetao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine - Dalian Campus, Dalian, China
| |
Collapse
|
4
|
Eugster R, Ganguin AA, Seidi A, Aleandri S, Luciani P. 3D printing injectable microbeads using a composite liposomal ink for local treatment of peritoneal diseases. Drug Deliv Transl Res 2024; 14:1567-1581. [PMID: 38006449 PMCID: PMC11052830 DOI: 10.1007/s13346-023-01472-y] [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] [Accepted: 11/05/2023] [Indexed: 11/27/2023]
Abstract
The peritoneal cavity offers an attractive administration route for challenging-to-treat diseases, such as peritoneal carcinomatosis, post-surgical adhesions, and peritoneal fibrosis. Achieving a uniform and prolonged drug distribution throughout the entire peritoneal space, though, is difficult due to high clearance rates, among others. To address such an unmet clinical need, alternative drug delivery approaches providing sustained drug release, reduced clearance rates, and a patient-centric strategy are required. Here, we describe the development of a 3D-printed composite platform for the sustained release of the tyrosine kinase inhibitor gefitinib (GEF), a small molecule drug with therapeutic applications for peritoneal metastasis and post-surgical adhesions. We present a robust method for the production of biodegradable liposome-loaded hydrogel microbeads that can overcome the pharmacokinetic limitations of small molecules with fast clearance rates, a current bottleneck for the intraperitoneal (IP) administration of these therapeutics. By means of an electromagnetic droplet printhead, we 3D printed microbeads employing an alginate-based ink loaded with GEF-containing multilamellar vesicles (MLVs). The sustained release of GEF from microbeads was demonstrated. In vitro studies on an immortalized human hepatic cancer cell line (Huh-7) proved concentration-dependent cell death. These findings demonstrate the potential of 3D-printed alginate microbeads containing liposomes for delivering small drug compounds into the peritoneum, overcoming previous limitations of IP drug delivery.
Collapse
Affiliation(s)
- Remo Eugster
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Aymar Abel Ganguin
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Amirmohammad Seidi
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012, Bern, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012, Bern, Switzerland.
| |
Collapse
|
5
|
Eş I, Thakur A, Mousavi Khaneghah A, Foged C, de la Torre LG. Engineering aspects of lipid-based delivery systems: In vivo gene delivery, safety criteria, and translation strategies. Biotechnol Adv 2024; 72:108342. [PMID: 38518964 DOI: 10.1016/j.biotechadv.2024.108342] [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: 01/06/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024]
Abstract
Defects in the genome cause genetic diseases and can be treated with gene therapy. Due to the limitations encountered in gene delivery, lipid-based supramolecular colloidal materials have emerged as promising gene carrier systems. In their non-functionalized form, lipid nanoparticles often demonstrate lower transgene expression efficiency, leading to suboptimal therapeutic outcomes, specifically through reduced percentages of cells expressing the transgene. Due to chemically active substituents, the engineering of delivery systems for genetic drugs with specific chemical ligands steps forward as an innovative strategy to tackle the drawbacks and enhance their therapeutic efficacy. Despite intense investigations into functionalization strategies, the clinical outcome of such therapies still needs to be improved. Here, we highlight and comprehensively review engineering aspects for functionalizing lipid-based delivery systems and their therapeutic efficacy for developing novel genetic cargoes to provide a full snapshot of the translation from the bench to the clinics. We outline existing challenges in the delivery and internalization processes and narrate recent advances in the functionalization of lipid-based delivery systems for nucleic acids to enhance their therapeutic efficacy and safety. Moreover, we address clinical trials using these vectors to expand their clinical use and principal safety concerns.
Collapse
Affiliation(s)
- Ismail Eş
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Headington, Oxford OX3 7DQ, UK.
| | - Aneesh Thakur
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Amin Mousavi Khaneghah
- Faculty of Biotechnologies (BioTech), ITMO University 191002, 9 Lomonosova Street, Saint Petersburg, Russia.
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Lucimara Gaziola de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
6
|
Liu A, He M, Liu C, Ye Z, Tan CP, Liu Y, Gong J, Lei J, He Y, Zhu S, Zhao J, Xu YJ, Liu Y. Prevention of Hypercholesterolemia with "Liposomes in Microspheres" Composite Carriers: A Promising Approach for Intestinal-Targeted Oral Delivery of Astaxanthin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6118-6132. [PMID: 38477232 DOI: 10.1021/acs.jafc.3c08697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Cardiovascular diseases are caused by hypercholesterolemia. Astaxanthin (AST) has been reported to exhibit antioxidant and anti-inflammatory properties. However, its bioavailability is poor because of low solubility and instability. In order to improve the bioavailability of AST, we developed an intestinal-responsive composite carrier termed as "liposomes in micropheres" incorporating N-succinyl-chitosan (NSC)-poly(ethylene glycol) (PEG) liposomes that functionalized by neonatal Fc receptors (FcRn) into hydrogels of sodium alginate (SA) and carboxymethyl chitosan (CMCS). In the AST NSC/HSA-PEG liposomes@SA/CMCS microspheres, the AST's encapsulation efficiency (EE) was 96.26% (w/w) and its loading capacity (LC) was 6.47% (w/w). AST NSC/HSA-PEG liposomes had stability in the gastric conditions and achieved long-term release of AST in intestinal conditions. Then, AST NSC/HSA-PEG liposomes@SA/CMCS bind to intestinal epithelial cell targets by the neonatal Fc receptor. In vitro permeation studies show that there was a 4-fold increase of AST NSC/HSA-PEG liposomes@SA/CMCS in AST permeation across the intestinal epithelium. Subsequent in vivo experiments demonstrated that the composite carrier exhibited a remarkable mucoadhesive capacity, allowing for extended intestinal retention of up to 12 h, and it displayed deep penetration through the mucus layer, efficiently entering the intestinal villi epithelial cells, and enhancing the absorption of AST and its bioavailability in vivo. And oral administration of AST NSC/HSA-PEG liposomes@SA/CMCS could effectively prevent hypercholesterolemia caused by a high-fat, high-cholesterol diet (HFHCD). These advancements highlight the potential of NSC/HSA-PEG liposomes@SA/CMCS composite carriers for targeted and oral uptake of hydrophobic bioactives.
Collapse
Affiliation(s)
- Aiyang Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Mengxue He
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Chunhuan Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Zhan Ye
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Chin-Ping Tan
- Department of Food Technology, Faculty of Food Science and Technology, University Putra Malaysia, Selangor 410500, Malaysia
| | - Yanjun Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jiajia Gong
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jingnan Lei
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yuan He
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Shuang Zhu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jialiang Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| |
Collapse
|
7
|
Giordani S, Marassi V, Zattoni A, Roda B, Reschiglian P. Liposomes characterization for market approval as pharmaceutical products: Analytical methods, guidelines and standardized protocols. J Pharm Biomed Anal 2023; 236:115751. [PMID: 37778202 DOI: 10.1016/j.jpba.2023.115751] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Liposomes are nano-sized lipid-based vesicles widely studied for their drug delivery capabilities. Compared to standard carries they exhibit better properties such as improved site-targeting and drug release, protection of drugs from degradation and clearance, and lower toxic side effects. At present, scientific literature is rich of studies regarding liposomes-based systems, while 14 types of liposomal products have been authorized to the market by EMA and FDA and many others have been approved by national agencies. Although the interest in nanodevices and nanomedicine has steadily increased in the last two decades the development of documentation regulating and standardizing all the phases of their development and quality control still suffers from major inadequacy due to the intrinsic complexity of nano-systems characterization. Many generic documents (Type 1) discussing guidelines for the study of nano-systems (lipidic and not) have been proposed while there is a lack of robust and standardized methods (Type 2 documents). As a result, a widespread of different techniques, approaches and methodologies are being used, generating results of variable quality and hard to compare with each other. Additionally, such documents are often subject to updates and rewriting further complicating the topic. Within this context the aim of this work is focused on bridging the gap in liposome characterization: the most recent standardized methodologies suitable for liposomes characterization are here reported (with the corresponding Type 2 documents) and revised in a short and pragmatical way focused on providing the reader with a practical background of the state of the art. In particular, this paper will put the accent on the methodologies developed to evaluate the main critical quality attributes (CQAs) necessary for liposomes market approval.
Collapse
Affiliation(s)
- Stefano Giordani
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy
| | - Valentina Marassi
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy.
| | - Andrea Zattoni
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy
| | - Barbara Roda
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy.
| | - Pierluigi Reschiglian
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy
| |
Collapse
|
8
|
Bordon G, Ramakrishna SN, Edalat SG, Eugster R, Arcifa A, Vermathen M, Aleandri S, Bertoncelj MF, Furrer J, Vermathen P, Isa L, Crockett R, Distler O, Luciani P. Liposomal aggregates sustain the release of rapamycin and protect cartilage from friction. J Colloid Interface Sci 2023; 650:1659-1670. [PMID: 37494862 DOI: 10.1016/j.jcis.2023.07.087] [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: 03/30/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Liposomes show promise as biolubricants for damaged cartilage, but their small size results in low joint and cartilage retention. We developed a zinc ion-based liposomal drug delivery system for local osteoarthritis therapy, focusing on sustained release and tribological protection from phospholipid lubrication properties. Our strategy involved inducing aggregation of negatively charged liposomes with zinc ions to extend rapamycin (RAPA) release and improve cartilage lubrication. Liposomal aggregation occurred within 10 min and was irreversible, facilitating excess cation removal. The aggregates extended RAPA release beyond free liposomes and displayed irregular morphology influenced by RAPA. At nearly 100 µm, the aggregates were large enough to exceed the previously reported size threshold for increased joint retention. Tribological assessment on silicon surfaces and ex vivo porcine cartilage revealed the system's excellent protective ability against friction at both nano- and macro-scales. Moreover, RAPA was shown to attenuate the fibrotic response in human OA synovial fibroblasts. Our findings suggest the zinc ion-based liposomal drug delivery system has potential to enhance OA therapy through extended release and cartilage tribological protection, while also illustrating the impact of a hydrophobic drug like RAPA on liposome aggregation and morphology.
Collapse
Affiliation(s)
- Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Shivaprakash N Ramakrishna
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zurich, Vladimir- Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Sam G Edalat
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Remo Eugster
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Andrea Arcifa
- Laboratory for Surface Science and Coating Technologies, EMPA, Uberlandstrasse 129, 8600 Dubendorf, Switzerland
| | - Martina Vermathen
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | | | - Julien Furrer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Peter Vermathen
- Magnetic Resonance Methodology, Institute of Diagnostic and Interventional Neuroradiology, University & Inselspital Bern, sitem-insel AG, Freiburgstrasse 3, 3010 Bern, Switzerland
| | - Lucio Isa
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zurich, Vladimir- Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Rowena Crockett
- Laboratory for Surface Science and Coating Technologies, EMPA, Uberlandstrasse 129, 8600 Dubendorf, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| |
Collapse
|
9
|
Skorup I, Valentino G, Aleandri S, Gelli R, Ganguin AA, Felli E, Selicean SE, Marxer RA, Teworte S, Lucić A, Gracia-Sancho J, Berzigotti A, Ridi F, Luciani P. Polyenylphosphatidylcholine as bioactive excipient in tablets for the treatment of liver fibrosis. Int J Pharm 2023; 646:123473. [PMID: 37788730 DOI: 10.1016/j.ijpharm.2023.123473] [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: 04/23/2023] [Revised: 08/24/2023] [Accepted: 09/30/2023] [Indexed: 10/05/2023]
Abstract
Liver fibrosis is a condition characterized by the accumulation of extracellular matrix (ECM) arising from the myofibroblastic transdifferentiation of hepatic stellate cells (HSCs) occurring as the natural response to liver damage. To date, no pharmacological treatments have been specifically approved for liver fibrosis. We recently reported a beneficial effect of polyenylphosphatidylcholines (PPCs)-rich formulations in reverting fibrogenic features of HSCs. However, unsaturated phospholipids' properties pose a constant challenge to the development of tablets as preferred patient-centric dosage form. Profiting from the advantageous physical properties of the PPCs-rich Soluthin® S 80 M, we developed a tablet formulation incorporating 70% w/w of this bioactive lipid. Tablets were characterized via X-ray powder diffraction, thermogravimetry, and Raman confocal imaging, and passed the major compendial requirements. To mimic physiological absorption after oral intake, phospholipids extracted from tablets were reconstituted as protein-free chylomicron (PFC)-like emulsions and tested on the fibrogenic human HSC line LX-2 and on primary cirrhotic rat hepatic stellate cells (PRHSC). Lipids extracted from tablets and reconstituted in buffer or as PFC-like emulsions exerted the same antifibrotic effect on both activated LX-2 and PRHSCs as observed with plain S 80 M liposomes, showing that the manufacturing process did not interfere with the bioactivity of PPCs.
Collapse
Affiliation(s)
- Ivo Skorup
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Gina Valentino
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Rita Gelli
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Aymar Abel Ganguin
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland
| | - Sonia Emilia Selicean
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland
| | - Rosanne Angela Marxer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Sarah Teworte
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Ana Lucić
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Jordi Gracia-Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland; Liver Vascular Biology Research Group, CIBEREHD, IDIBAPS Research Institute, Barcelona, Spain
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland
| | - Francesca Ridi
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
10
|
Sharma R, Yadav S, Yadav V, Akhtar J, Katari O, Kuche K, Jain S. Recent advances in lipid-based long-acting injectable depot formulations. Adv Drug Deliv Rev 2023; 199:114901. [PMID: 37257756 DOI: 10.1016/j.addr.2023.114901] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/28/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Long-acting injectable (LAIs) delivery systems sustain the drug therapeutic action in the body, resulting in reduced dosage regimen, toxicity, and improved patient compliance. Lipid-based depots are biocompatible, provide extended drug release, and improve drug stability, making them suitable for systemic and localized treatment of various chronic ailments, including psychosis, diabetes, hormonal disorders, arthritis, ocular diseases, and cancer. These depots include oil solutions, suspensions, oleogels, liquid crystalline systems, liposomes, solid lipid nanoparticles, nanostructured lipid carriers, phospholipid phase separation gel, vesicular phospholipid gel etc. This review summarizes recent advancements in lipid-based LAIs for delivering small and macromolecules, and their potential in managing chronic diseases. It also provides an overview of the lipid depots available in market or clinical phase, as well as patents for lipid-based LAIs. Furthermore, this review critically discusses the current scenario of using in vitro release methods to establish IVIVC and highlights the challenges involved in developing lipid-based LAIs.
Collapse
Affiliation(s)
- Reena Sharma
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Sheetal Yadav
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Vivek Yadav
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Junia Akhtar
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Oly Katari
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Kaushik Kuche
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Sanyog Jain
- Department of Pharmaceutics, Centre for Pharmaceutical Nanotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), Punjab 160062, India.
| |
Collapse
|
11
|
Zhuang L, Ye Z, Li L, Yang L, Gong W. Next-Generation TB Vaccines: Progress, Challenges, and Prospects. Vaccines (Basel) 2023; 11:1304. [PMID: 37631874 PMCID: PMC10457792 DOI: 10.3390/vaccines11081304] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), is a prevalent global infectious disease and a leading cause of mortality worldwide. Currently, the only available vaccine for TB prevention is Bacillus Calmette-Guérin (BCG). However, BCG demonstrates limited efficacy, particularly in adults. Efforts to develop effective TB vaccines have been ongoing for nearly a century. In this review, we have examined the current obstacles in TB vaccine research and emphasized the significance of understanding the interaction mechanism between MTB and hosts in order to provide new avenues for research and establish a solid foundation for the development of novel vaccines. We have also assessed various TB vaccine candidates, including inactivated vaccines, attenuated live vaccines, subunit vaccines, viral vector vaccines, DNA vaccines, and the emerging mRNA vaccines as well as virus-like particle (VLP)-based vaccines, which are currently in preclinical stages or clinical trials. Furthermore, we have discussed the challenges and opportunities associated with developing different types of TB vaccines and outlined future directions for TB vaccine research, aiming to expedite the development of effective vaccines. This comprehensive review offers a summary of the progress made in the field of novel TB vaccines.
Collapse
Affiliation(s)
- Li Zhuang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
- Hebei North University, Zhangjiakou 075000, China
| | - Zhaoyang Ye
- Hebei North University, Zhangjiakou 075000, China
| | - Linsheng Li
- Hebei North University, Zhangjiakou 075000, China
| | - Ling Yang
- Hebei North University, Zhangjiakou 075000, China
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| |
Collapse
|
12
|
Liu A, Chai X, Zhu S, Chin PT, He M, Xu YJ, Liu Y. Effects of N-succinyl-chitosan coating on properties of astaxanthin-loaded PEG-liposomes: Environmental stability, antioxidant/antibacterial activities, and in vitro release. Int J Biol Macromol 2023:125311. [PMID: 37302627 DOI: 10.1016/j.ijbiomac.2023.125311] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/22/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Astaxanthin (AST) has outstanding antioxidant and anti-inflammation bioactivities, but the low biocompatibility and stability limit its application in foods. In this study, N-succinyl-chitosan (NSC)-coated AST polyethylene glycol (PEG)-liposomes were constructed to enhance the biocompatibility, stability, and intestinal-targeted migration of AST. The AST NSC/PEG-liposomes were uniform in size, had larger particles, greater encapsulation efficiency, and better storage, pH, and temperature stability than the AST PEG-liposomes. AST NSC/PEG-liposomes exerted stronger antibacterial and antioxidant activities against Escherichia coli and Staphylococcus aureus than AST PEG-liposomes. The NSC coating not only protects AST PEG-liposomes from gastric acid but also prolongs the retention and sustained release of AST NSC/PEG-liposomes depending on the intestinal pH. Moreover, caco-2 cellular uptake studies showed that AST NSC/PEG-liposomes had higher cellular uptake efficiency than AST PEG-liposomes. And AST NSC/PEG-liposomes were taken up by caco-2 cells through clathrin mediated endocytic, macrophage pathways and paracellular transport pathway. These results further proved that AST NSC/PEG-liposomes delayed the release and promoted the intestinal absorption of AST. Hence, AST PEG-liposomes coated with NSC could potentially be used as an efficient delivery system for therapeutic AST.
Collapse
Affiliation(s)
- Aiyang Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Xiuhang Chai
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Shuang Zhu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Ping-Tan Chin
- Department of Food Technology, Faculty of Food Science and Technology, University Putra Malaysia, Selangor 410500, Malaysia
| | - Mengxue He
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
13
|
Starosta R, Santos TC, Dinis de Sousa AF, Santos MS, Corvo ML, Tomaz AI, de Almeida RFM. Assessing the role of membrane lipids in the action of ruthenium(III) anticancer compounds. Front Mol Biosci 2023; 9:1059116. [PMID: 36660430 PMCID: PMC9845782 DOI: 10.3389/fmolb.2022.1059116] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
This work addresses the possible role of the cell membrane in the molecular mechanism of action of two salan-type ruthenium complexes that were previously shown to be active against human tumor cells, namely [Ru(III)(L1)(PPh3)Cl] and [Ru(III)(L2)(PPh3)Cl] (where L1 is 6,6'-(1R,2R)-cyclohexane-1,2-diylbis(azanediyl)bis(methylene)bis(3-methoxyphenol); and L2 is 2,2'-(1R,2R)-cyclohexane-1,2-diylbis(azanediyl)bis(methylene)bis(4-methoxyphenol)). One-component membrane models were first used, a disordered fluid bilayer of dioleoylphosphatodylcholine (DOPC), and an ordered rigid gel bilayer of dipalmitoylphosphatidylcholine. In addition, two quaternary mixtures of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin and cholesterol were used to mimic the lipid composition either of mammalian plasma membrane (1:1:1:1 mol ratio) or of a cancer cell line membrane (36.2:23.6:6.8:33.4 mol ratio). The results show that both salan ligands L1 and L2 bind relatively strongly to DOPC bilayers, but without significantly affecting their structure. The ruthenium complexes have moderate affinity for DOPC. However, their impact on the membranes was notable, leading to a significant increase in the permeability of the lipid vesicles. None of the compounds compromised liposome integrity, as revealed by dynamic light scattering. Fluorescence spectroscopy studies revealed changes in the biophysical properties of all membrane models analyzed in the presence of the two complexes, which promoted an increased fluidity and water penetration into the lipid bilayer in the one-component systems. In the quaternary mixtures, one of the complexes had an analogous effect (increasing water penetration), whereas the other complex reorganized the liquid ordered and liquid disordered domains. Thus, small structural differences in the metal ligands may lead to different outcomes. To better understand the effect of these complexes in cancer cells, the membrane dipole potential was also measured. For both Ru complexes, an increase in the dipole potential was observed for the cancer cell membrane model, while no alteration was detected on the non-cancer plasma membrane model. Our results show that the action of the Ru(III) complexes tested involves changes in the biophysical properties of the plasma membrane, and that it also depends on membrane lipid composition, which is frequently altered in cancer cells when compared to their normal counterparts.
Collapse
Affiliation(s)
- Radoslaw Starosta
- Faculty of Chemistry, University of Wroclaw, Wroclaw, Poland,Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Telma C. Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia F. Dinis de Sousa
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Soledade Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - M. Luisa Corvo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Isabel Tomaz
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal,*Correspondence: Rodrigo F. M. de Almeida, ; Ana Isabel Tomaz,
| | - Rodrigo F. M. de Almeida
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal,*Correspondence: Rodrigo F. M. de Almeida, ; Ana Isabel Tomaz,
| |
Collapse
|
14
|
Aleandri S, Rahnfeld L, Chatzikleanthous D, Bergadano A, Bühr C, Detotto C, Fuochi S, Weber-Wilk K, Schürch S, van Hoogevest P, Luciani P. Development and in vivo validation of phospholipid-based depots for the sustained release of bupivacaine. Eur J Pharm Biopharm 2022; 181:300-309. [PMID: 36427675 DOI: 10.1016/j.ejpb.2022.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
By direct deposition of the drug at the local site of action, injectable depot formulations - intended for treatment of a local disease or for local intervention - are designed to limit the immediate exposure of the active principle at a systemic level and to reduce the frequency of administration. To overcome known drawbacks in the production of some marketed phospholipid-based depots, here we propose to manufacture drug-loaded negatively charged liposomes through conventional technologies and to control their aggregation mixing a solution of divalent cations prior to administration. We identified phosphatidylglycerol (PG) as the most suitable phospholipid for controlled aggregation of the liposomes and to modulate the release of the anesthetic bupivacaine (BUP) from liposomal depots. In vivo imaging of the fluorescently-labelled liposomes showed a significantly higher retention of the PG liposomes at the injection site with respect to zwitterionic ones. In situ mixing of PG liposomes with calcium salts significantly extended the area under the curve of BUP in plasma compared to the non-depot system. Overall, controlling the aggregation of negatively charged liposomes with divalent cations not only modulated the particle clearance from the injection site but also the release in vivo of a small amphipathic drug such as BUP.
Collapse
Affiliation(s)
- Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland
| | - Lisa Rahnfeld
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland; Institute of Pharmacy, Faculty of Biosciences, Friedrich Schiller University, Jena, Germany
| | - Despo Chatzikleanthous
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland
| | | | - Claudia Bühr
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland
| | - Carlotta Detotto
- Experimental Animal Center (EAC), University of Bern, Switzerland
| | - Sara Fuochi
- Experimental Animal Center (EAC), University of Bern, Switzerland
| | - Kevin Weber-Wilk
- Experimental Animal Center (EAC), University of Bern, Switzerland
| | - Stefan Schürch
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland
| | | | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland; Institute of Pharmacy, Faculty of Biosciences, Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
15
|
Tracking matricellular protein SPARC in extracellular vesicles as a non-destructive method to evaluate lipid-based antifibrotic treatments. Commun Biol 2022; 5:1155. [PMID: 36310239 PMCID: PMC9618575 DOI: 10.1038/s42003-022-04123-z] [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: 12/07/2021] [Accepted: 10/17/2022] [Indexed: 11/10/2022] Open
Abstract
Uncovering the complex cellular mechanisms underlying hepatic fibrogenesis could expedite the development of effective treatments and noninvasive diagnosis for liver fibrosis. The biochemical complexity of extracellular vesicles (EVs) and their role in intercellular communication make them an attractive tool to look for biomarkers as potential alternative to liver biopsies. We developed a solid set of methods to isolate and characterize EVs from differently treated human hepatic stellate cell (HSC) line LX-2, and we investigated their biological effect onto naïve LX-2, proving that EVs do play an active role in fibrogenesis. We mined our proteomic data for EV-associated proteins whose expression correlated with HSC treatment, choosing the matricellular protein SPARC as proof-of-concept for the feasibility of fluorescence nanoparticle-tracking analysis to determine an EV-based HSCs’ fibrogenic phenotype. We thus used EVs to directly evaluate the efficacy of treatment with S80, a polyenylphosphatidylcholines-rich lipid, finding that S80 reduces the relative presence of SPARC-positive EVs. Here we correlated the cellular response to lipid-based antifibrotic treatment to the relative presence of a candidate protein marker associated with the released EVs. Along with providing insights into polyenylphosphatidylcholines treatments, our findings pave the way for precise and less invasive diagnostic analyses of hepatic fibrogenesis. A method is developed to isolate and characterize extracellular vesicles (EVs) from human hepatic stellate cells and proteomics reveals that the matricellular protein SPARC may be used as an EV marker after lipid-based antifibrotic treatment.
Collapse
|
16
|
Rofeal M, Abdelmalek F, Steinbüchel A. Naturally-Sourced Antibacterial Polymeric Nanomaterials with Special Reference to Modified Polymer Variants. Int J Mol Sci 2022; 23:4101. [PMID: 35456918 PMCID: PMC9030380 DOI: 10.3390/ijms23084101] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advancements in treating bacterial infections, antibiotic resistance (AR) is still an emerging issue. However, polymeric nanocarriers have offered unconventional solutions owing to their capability of exposing more functional groups, high encapsulation efficiency (EE) and having sustained delivery. Natural polymeric nanomaterials (NMs) are contemplated one of the most powerful strategies in drug delivery (DD) in terms of their safety, biodegradability with almost no side effects. Every nanostructure is tailored to enhance the system functionality. For example, cost-effective copper NPs could be generated in situ in cellulose sheets, demonstrating powerful antibacterial prospects for food safety sector. Dendrimers also have the capacity for peptide encapsulation, protecting them from proteolytic digestion for prolonged half life span. On the other hand, the demerits of naturally sourced polymers still stand against their capacities in DD. Hence, Post-synthetic modification of natural polymers could play a provital role in yielding new hybrids while retaining their biodegradability, which could be suitable for building novel super structures for DD platforms. This is the first review presenting the contribution of natural polymers in the fabrication of eight polymeric NMs including particulate nanodelivery and nanofabrics with antibacterial and antibiofilm prospects, referring to modified polymer derivatives to explore their full potential for obtaining sustainable DD products.
Collapse
Affiliation(s)
- Marian Rofeal
- International Center for Research on Innovative Biobased Materials (ICRI-BioM)—International Research Agenda, Lodz University of Technology, Zeromskiego 116, 90–924 Lodz, Poland;
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria 21521, Egypt
| | - Fady Abdelmalek
- International Center for Research on Innovative Biobased Materials (ICRI-BioM)—International Research Agenda, Lodz University of Technology, Zeromskiego 116, 90–924 Lodz, Poland;
| | - Alexander Steinbüchel
- International Center for Research on Innovative Biobased Materials (ICRI-BioM)—International Research Agenda, Lodz University of Technology, Zeromskiego 116, 90–924 Lodz, Poland;
| |
Collapse
|
17
|
Wang J, Gong J, Wei Z. Strategies for Liposome Drug Delivery Systems to Improve Tumor Treatment Efficacy. AAPS PharmSciTech 2021; 23:27. [PMID: 34907483 DOI: 10.1208/s12249-021-02179-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
In the advancement of tumor therapy, in addition to the search for new antitumor compounds, the development of nano-drug delivery systems has opened up new pathways for tumor treatment by addressing some of the limitations of traditional drugs. Liposomes have received much attention for their high biocompatibility, low toxicity, high inclusivity, and improved drug bioavailability. They are one of the most studied nanocarriers, changing the size and surface characteristics of liposomes to better fit the tumor environment by taking advantage of the unique pathophysiology of tumors. They can also be designed as tumor targeting drug delivery vehicles for the precise delivery of active drugs into tumor cells. This paper reviews the current development of liposome formulations, summarizes the characterization methods of liposomes, and proposes strategies to improve the effectiveness of tumor treatment. Finally, it provides an outlook on the challenges and future directions of the field. Graphical abstract.
Collapse
|
18
|
McCartan AJS, Curran DW, Mrsny RJ. Evaluating parameters affecting drug fate at the intramuscular injection site. J Control Release 2021; 336:322-335. [PMID: 34153375 DOI: 10.1016/j.jconrel.2021.06.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022]
Abstract
Intramuscular (IM) injections are a well-established method of delivering a variety of therapeutics formulated for parenteral administration. While the wide range of commercial IM pharmaceuticals provide a wealth of pharmacokinetic (PK) information following injection, there remains an inadequate understanding of drug fate at the IM injection site that could dictate these PK outcomes. An improved understanding of injection site events could improve approaches taken by formulation scientists to identify therapeutically effective and consistent drug PK outcomes. Interplay between the typically non-physiological aspects of drug formulations and the homeostatic IM environment may provide insights into the fate of drugs at the IM injection site, leading to predictions of how a drug will behave post-injection in vivo. Immune responses occur by design after e.g. vaccine administration, however immune responses post-injection are not in the scope of this article. Taking cues from existing in vitro modelling technologies, the purpose of this article is to propose "critical parameters" of the IM environment that could be examined in hypothesis-driven studies. Outcomes of such studies might ultimately be useful in predicting and improving in vivo PK performance of IM injected drugs.
Collapse
Affiliation(s)
- Adam J S McCartan
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, Avon BA2 7AY, UK
| | - David W Curran
- CMC Analytical, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Randall J Mrsny
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, Avon BA2 7AY, UK.
| |
Collapse
|
19
|
Craciunescu O, Icriverzi M, Florian PE, Roseanu A, Trif M. Mechanisms and Pharmaceutical Action of Lipid Nanoformulation of Natural Bioactive Compounds as Efficient Delivery Systems in the Therapy of Osteoarthritis. Pharmaceutics 2021; 13:1108. [PMID: 34452068 PMCID: PMC8399940 DOI: 10.3390/pharmaceutics13081108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease. An objective of the nanomedicine and drug delivery systems field is to design suitable pharmaceutical nanocarriers with controllable properties for drug delivery and site-specific targeting, in order to achieve greater efficacy and minimal toxicity, compared to the conventional drugs. The aim of this review is to present recent data on natural bioactive compounds with anti-inflammatory properties and efficacy in the treatment of OA, their formulation in lipid nanostructured carriers, mainly liposomes, as controlled release systems and the possibility to be intra-articularly (IA) administered. The literature regarding glycosaminoglycans, proteins, polyphenols and their ability to modify the cell response and mechanisms of action in different models of inflammation are reviewed. The advantages and limits of using lipid nanoformulations as drug delivery systems in OA treatment and the suitable route of administration are also discussed. Liposomes containing glycosaminoglycans presented good biocompatibility, lack of immune system activation, targeted delivery of bioactive compounds to the site of action, protection and efficiency of the encapsulated material, and prolonged duration of action, being highly recommended as controlled delivery systems in OA therapy through IA administration. Lipid nanoformulations of polyphenols were tested both in vivo and in vitro models that mimic OA conditions after IA or other routes of administration, recommending their clinical application.
Collapse
Affiliation(s)
- Oana Craciunescu
- National Institute of R&D for Biological Sciences, 296 Splaiul Independentei, 060031 Bucharest, Romania;
| | - Madalina Icriverzi
- The Institute of Biochemistry of the Romanian Academy, 296 Splaiul Independentei, 060031 Bucharest, Romania; (M.I.); (P.E.F.); (A.R.)
| | - Paula Ecaterina Florian
- The Institute of Biochemistry of the Romanian Academy, 296 Splaiul Independentei, 060031 Bucharest, Romania; (M.I.); (P.E.F.); (A.R.)
| | - Anca Roseanu
- The Institute of Biochemistry of the Romanian Academy, 296 Splaiul Independentei, 060031 Bucharest, Romania; (M.I.); (P.E.F.); (A.R.)
| | - Mihaela Trif
- The Institute of Biochemistry of the Romanian Academy, 296 Splaiul Independentei, 060031 Bucharest, Romania; (M.I.); (P.E.F.); (A.R.)
| |
Collapse
|
20
|
Zhang T, Su M, Liu M, Tao M, Yang Y, Liu C, Zeng X, Pan D, Wu Z, Guo Y. Optimization of Encapsulation Using Milk Polar Lipid Liposomes with S-Layer Protein and Transport Study of the ACE-Inhibitory Peptide RLSFNP. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7049-7056. [PMID: 34132090 DOI: 10.1021/acs.jafc.1c02216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The purpose of this study is to develop a new type of nanodrug delivery material by modifying milk polar lipid (MPL) liposomes with the S-layer protein. LIP-RLSFNP (MPL liposomes encapsulating RLSFNP (Arg-Leu-Ser-Phe-Asn-Pro)) and SLP-LIP-RLSFNP (S-layer protein-modified LIP-RLSFNP) were prepared and characterized by transmission electron microscopy, Fourier transform infrared spectroscopy, confocal laser scanning microscopy, surface plasmon resonance, and mastersizer dynamic light scattering measurements. The results showed that the S-layer protein could modify the surface of MPL liposomes, stabilize the shape of the vesicles, and improve the resistance to external interference. Furthermore, SLP-LIP-RLSFNP showed better performance in in vitro and in vivo experiments compared with LIP-RLSFNP in terms of promoting absorption and delayed release. The findings suggested that MPL liposomes modified with the S-layer protein have potential for use as an effective delivery system for therapeutic proteins and peptides.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Mi Su
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Mingzhen Liu
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Mingxuan Tao
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Yao Yang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Chen Liu
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Xiaoqun Zeng
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Daodong Pan
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Zhen Wu
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Yuxing Guo
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| |
Collapse
|
21
|
Hoogevest P, Tiemessen H, Metselaar JM, Drescher S, Fahr A. The Use of Phospholipids to Make Pharmaceutical Form Line Extensions. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Peter Hoogevest
- Phospholipid Research Center Im Neuenheimer Feld 515 Heidelberg 69120D‐69120 Germany
| | - Harry Tiemessen
- Technical & Research Development PHAD PDU Specialty Novartis Campus Physical Garden (WSJ 177) 2.14 Basel CH‐4002 Switzerland
| | - Josbert M. Metselaar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic Aachen D‐52074 Germany
- Institute for Biomedical Engineering, Faculty of Medicine RWTH Aachen University Aachen D‐52074 Germany
| | - Simon Drescher
- Phospholipid Research Center Im Neuenheimer Feld 515 Heidelberg D‐69120 Germany
| | - Alfred Fahr
- Professor Emeritus, Pharmaceutical Technology Friedrich‐Schiller‐University Jena Jena Germany
| |
Collapse
|
22
|
Drescher S, van Hoogevest P. The Phospholipid Research Center: Current Research in Phospholipids and Their Use in Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12121235. [PMID: 33353254 PMCID: PMC7766331 DOI: 10.3390/pharmaceutics12121235] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the research on phospholipids and their use for drug delivery related to the Phospholipid Research Center Heidelberg (PRC). The focus is on projects that have been approved by the PRC since 2017 and are currently still ongoing or have recently been completed. The different projects cover all facets of phospholipid research, from basic to applied research, including the use of phospholipids in different administration forms such as liposomes, mixed micelles, emulsions, and extrudates, up to industrial application-oriented research. These projects also include all routes of administration, namely parenteral, oral, and topical. With this review we would like to highlight possible future research directions, including a short introduction into the world of phospholipids.
Collapse
|
23
|
Weber F, Rahnfeld L, Luciani P. Analytical profiling and stability evaluation of liposomal drug delivery systems: A rapid UHPLC-CAD-based approach for phospholipids in research and quality control. Talanta 2020; 220:121320. [DOI: 10.1016/j.talanta.2020.121320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 01/26/2023]
|
24
|
Regenold M, Steigenberger J, Siniscalchi E, Dunne M, Casettari L, Heerklotz H, Allen C. Determining critical parameters that influence in vitro performance characteristics of a thermosensitive liposome formulation of vinorelbine. J Control Release 2020; 328:551-561. [DOI: 10.1016/j.jconrel.2020.08.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023]
|
25
|
Salehi B, Mishra AP, Nigam M, Kobarfard F, Javed Z, Rajabi S, Khan K, Ashfaq HA, Ahmad T, Pezzani R, Ramírez-Alarcón K, Martorell M, Cho WC, Ayatollahi SA, Sharifi-Rad J. Multivesicular Liposome (Depofoam) in Human Diseases. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:9-21. [PMID: 33224207 PMCID: PMC7667536 DOI: 10.22037/ijpr.2020.112291.13663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Drug development is a key point in the research of new therapeutic treatments for increasing maximum drug loading and prolonged drug effect. Encapsulation of drugs into multivesicular liposomes (DepoFoam) is a nanotechnology that allow delivery of the active constituent at a sufficient concentration during the entire treatment period. This guarantees the reduction of drug administration frequency, a very important factor in a prolonged treatment. Currently, diverse DepoFoam drugs are approved for clinical use against neurological diseases and for post-surgical pain management while other are under development for reducing surgical bleeding and for post-surgical analgesia. Also, on pre-clinical trials on cancer DepoFoam can improve bioavailability and stability of the drug molecules minimizing side effects by site-specific targeted delivery. In the current work, available literature on structure, preparation and pharmacokinetics of DepoFoam are reviewed. Moreover, we investigated approved DepoFoam formulations and preclinical studies with this nanotechnology.
Collapse
Affiliation(s)
- Bahare Salehi
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran.,Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Abhay P Mishra
- Department of Pharmaceutical Chemistry, H. N. B. Garhwal (A Central) University, Srinagar Garhwal, 246174, Uttarakhand, India
| | - Manisha Nigam
- Department of Biochemistry, H. N. B. Garhwal (A Central) University, Srinagar Garhwal, 246174, Uttarakhand, India
| | - Farzad Kobarfard
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeeshan Javed
- Office for Research innovation and commercialization (ORIC) Lahore garrison University, sector-c phase VI, DHA, Lahore Pakistan
| | - Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khushbukhat Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Hafiz Ahsan Ashfaq
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Toqeer Ahmad
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Raffaele Pezzani
- OU Endocrinology, Dept. Medicine (DIMED), University of Padova, via Ospedale 105, Padova 35128, Italy.,AIROB, Associazione Italiana per la Ricerca Oncologica di Base, Padova, Italy
| | - Karina Ramírez-Alarcón
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepcion, Concepcion 4070386, Chile
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepcion, Concepcion 4070386, Chile.,Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, Concepcion 4070386, Chile
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong, China
| | - Seyed Abdulmajid Ayatollahi
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Pharmacognosy and Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Zeng Q, Cai X, Cao Y, Zhou C, Yu L, Chen J. Preparation, characterization, and pharmacodynamic study on deep second degree burns of total flavonoids composite phospholipids liposome gel of Oxytropis falcata Bunge. Drug Dev Ind Pharm 2020; 46:2000-2009. [PMID: 33095085 DOI: 10.1080/03639045.2020.1841787] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Wound healing is the treatment problem after deep second degree (II°) burns. The p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor-κB/inhibitory factor-κB (NF-κB/IκB) signal pathways play significant role in angiogenesis and wound repair after burns.This study aimed to investigate the preparation, characterization and pharmacodynamics of the total flavonoids composite phospholipids liposome of Oxytropis falcata Bunge (TFOFB-CPL) on deep II° burns to research its biological activity and underlying mechanism. The TFOFB-CPL was prepared by thin-film dispersion method and the preparation process was optimized via central composite design. The TFOFB-CPL was then characterized by using particle size, polydispersity indexes (PDIs), zeta potential, encapsulation efficiency (EE) and morphology. Moerover, in vitro transdermal test and in vivo pharmacodynamic study included wound healing rate, hematoxylin-eosin (HE) staining, masson staning, western blotting and RT-PCR. The results showed that the therapeutic effects of TFOFB-CPL gel on deep II° burns, especially during wound healing were significant. TFOFB-CPL gel has a sustained-release effect during the treatment of deep II° burns with forming drug depot in the dermis layer. The wound healing rate of TFOFB-CPL gel group was near positive group and better than the other groups. TFOFB-CPL gel could promote the growth of epidermis, skin appendages, fibrovascular and collagen fibers, and had obvious anti-inflammatory effects. Moreover, TFOFB-CPL gel inhibited the activation of p38MAPK and the degradation of IκBα, and promoted the neonatal wounds during the early stage. Therefore, TFOFB-CPL gel could be considered as a novel preparation for treating deep II° burns.
Collapse
Affiliation(s)
- Qiping Zeng
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Xiaohui Cai
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Yixiang Cao
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Chengfang Zhou
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Le Yu
- Department of Pathology, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Jinshan Chen
- Department of Pharmacy, the 909th Hospital of PLA/the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| |
Collapse
|
27
|
Fan Y, Marioli M, Zhang K. Analytical characterization of liposomes and other lipid nanoparticles for drug delivery. J Pharm Biomed Anal 2020; 192:113642. [PMID: 33011580 DOI: 10.1016/j.jpba.2020.113642] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022]
Abstract
Lipid nanoparticles, especially liposomes and lipid/nucleic acid complexed nanoparticles have shown great success in the pharmaceutical industry. Their success is attributed to stable drug loading, extended pharmacokinetics, reduced off-target side effects, and enhanced delivery efficiency to disease targets with formidable blood-brain or plasma membrane barriers. Therefore, they offer promising formulation options for drugs limited by low therapeutic indexes in traditional dosage forms and current "undruggable" targets. Recent development of siRNA, antisense oligonucleotide, or the CRISPR complex-loaded lipid nanoparticles and liposomal vaccines also shed light on their potential in enabling versatile formulation platforms for new pharmaceutical modalities. Analytical characterization of these nanoparticles is critical to drug design, formulation development, understanding in vivo performance, as well as quality control. The multi-lipid excipients, unique core-bilayer structure, and nanoscale size all underscore their complicated critical quality attributes, including lipid species, drug encapsulation efficiency, nanoparticle characteristics, product stability, and drug release. To address these challenges and facilitate future applications of lipid nanoparticles in drug development, we summarize available analytical approaches for physicochemical characterizations of lipid nanoparticle-based pharmaceutical modalities. Furthermore, we compare advantages and challenges of different techniques, and highlight the promise of new strategies for automated high-throughput screening and future development.
Collapse
Affiliation(s)
- Yuchen Fan
- Research and Early Development, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Maria Marioli
- Pharma Technical Development Europe Analytics, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Kelly Zhang
- Research and Early Development, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
28
|
Nicardipine Loaded Solid Phospholipid Extrudates for the Prevention of Cerebral Vasospasms: In Vitro Characterization. Pharmaceutics 2020; 12:pharmaceutics12090817. [PMID: 32872184 PMCID: PMC7557531 DOI: 10.3390/pharmaceutics12090817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/29/2022] Open
Abstract
The aim of the study was to develop nicardipine loaded phospholipid extrudates as an alternative for PLA/PLGA-based implants for the prevention of cerebral vasospasms. Extrudates of different mixtures of saturated and unsaturated phosphatidylcholine (PC) were produced and characterized by DSC, microscopy and texture analysis. Single phospholipid components were identified by ELSD-HPLC. Extrudates of 2 mm diameter were obtained by twin screw extrusion temperatures below 50 °C. The ratio of unsaturated and saturated phosphatidylcholine components determines the physicochemical properties of the extrudates as well as the rate of erosion. Nicardipine loaded phospholipids extrudates released the drug over several weeks in vitro. The phospholipid composition of the remaining extrudate changed during the release, the content of unsaturated phospholipids decreased faster compared to the saturated ones. In conclusion, solid phospholipid extrudates are promising materials for the development of new parenteral controlled release systems.
Collapse
|
29
|
Rahnfeld L, Luciani P. Injectable Lipid-Based Depot Formulations: Where Do We Stand? Pharmaceutics 2020; 12:E567. [PMID: 32575406 PMCID: PMC7356974 DOI: 10.3390/pharmaceutics12060567] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 01/18/2023] Open
Abstract
The remarkable number of new molecular entities approved per year as parenteral drugs, such as biologics and complex active pharmaceutical ingredients, calls for innovative and tunable drug delivery systems. Besides making these classes of drugs available in the body, injectable depot formulations offer the unique advantage in the parenteral world of reducing the number of required injections, thus increasing effectiveness as well as patient compliance. To date, a plethora of excipients has been proposed to formulate depot systems, and among those, lipids stand out due to their unique biocompatibility properties and safety profile. Looking at the several long-acting drug delivery systems based on lipids designed so far, a legitimate question may arise: How far away are we from an ideal depot formulation? Here, we review sustained release lipid-based platforms developed in the last 5 years, namely oil-based solutions, liposomal systems, in situ forming systems, solid particles, and implants, and we critically discuss the requirements for an ideal depot formulation with respect to the used excipients, biocompatibility, and the challenges presented by the manufacturing process. Finally, we delve into lights and shadows originating from the current setups of in vitro release assays developed with the aim of assessing the translational potential of depot injectables.
Collapse
Affiliation(s)
| | - Paola Luciani
- Pharmaceutical Technology Research Group, Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland;
| |
Collapse
|
30
|
Camilo CJJ, Leite DOD, Silva ARA, Menezes IRA, Coutinho HDM, Costa JGM. Lipid vesicles: applications, principal components and methods used in their formulations: A review. ACTA BIOLÓGICA COLOMBIANA 2020. [DOI: 10.15446/abc.v25n2.74830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Liposomes and niosomes are currently the most studied lipid vesicles in the nanomedicine field. The system formed by a phospholipid bilayer in aqueous medium allows these vesicles to carry both hydrophilic and lipophilic compounds, providing an increase in solubility of drugs lready used in conventional therapy. The focus on the development of these vesicles should be directed to determining the ideal composition, with low toxicity, biocompatibility and which remains stable for long periods. These characteristics are related to the components used for formulation and the substances that will be encapsulated. Another important point relates to the methods used during formulation, which are important in determining the type of vesicle formed, whether these be large or small, unilamellar or multilamellar. Because of the deliberate actions applied in the development of these vesicles, this review sought to gather updated information regarding the different methods used, including their main components while considering the behavior of each of them when used in different formulations. Also, data showing the importance of formulations in the medical field evidencing studies performed with liposome and niosome vesicles as promising in this area, and others, were included. The approach allows a better understanding of the participation of components in formulations such as cholesterol and non-ionic surfactants, as well as the basis for choosing the ideal components and methods for future research in the development of these vesicles.
Collapse
|
31
|
An in vitro digestion study of encapsulated lactoferrin in rapeseed phospholipid-based liposomes. Food Chem 2020; 321:126717. [PMID: 32259734 DOI: 10.1016/j.foodchem.2020.126717] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/23/2022]
Abstract
Effectiveness of liposomes elaborated with rapeseed phospholipid (RP) extracted from a residue of oil processing, stigmasterol (ST) and/or hydrogenated phosphatidylcholine (HPC) for the encapsulation lactoferrin (LF) was studied; lipid membrane of liposomes was characterized (bilayer size, chain conformational order, lateral packing, lipid phase, and morphology) and the protection offered to the encapsulated LF during in vitro digestion was determined. Liposomes composed of RP+STLC(low concentration) showed spherical and irregular vesicles without perforations. Lamellar structure was organized in a liquid-ordered phase with a potential orthorhombic packing. Stability and size of the liposomes were more affected by gastric digestion than intestinal digestion; 67-80% of the initially encapsulated LF remained intact after gastric digestion whereas the percentage was reduced to 16-35% after intestinal digestion. Our results shows that liposomes elaborated with RP, properly combined with other lipids, can be a useful oral delivery system of molecules sensitive to digestive enzymes.
Collapse
|
32
|
He Y, Qin L, Huang Y, Ma C. Advances of Nano-Structured Extended-Release Local Anesthetics. NANOSCALE RESEARCH LETTERS 2020; 15:13. [PMID: 31950284 PMCID: PMC6965527 DOI: 10.1186/s11671-019-3241-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/26/2019] [Indexed: 05/08/2023]
Abstract
Extended-release local anesthetics (LAs) have drawn increasing attention with their promising role in improving analgesia and reducing adverse events of LAs. Nano-structured carriers such as liposomes and polymersomes optimally meet the demands of/for extended-release, and have been utilized in drug delivery over decades and showed satisfactory results with extended-release. Based on mature technology of liposomes, EXPAREL, the first approved liposomal LA loaded with bupivacaine, has seen its success in an extended-release form. At the same time, polymersomes has advances over liposomes with complementary profiles, which inspires the emergence of hybrid carriers. This article summarized the recent research successes on nano-structured extended-release LAs, of which liposomal and polymeric are mainstream systems. Furthermore, with continual optimization, drug delivery systems carry properties beyond simple transportation, such as specificity and responsiveness. In the near future, we may achieve targeted delivery and controlled-release properties to satisfy various analgesic requirements.
Collapse
Affiliation(s)
- Yumiao He
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Linan Qin
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Yuguang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China.
| | - Chao Ma
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China.
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
33
|
Klein ME, Mauch S, Rieckmann M, Martínez DG, Hause G, Noutsias M, Hofmann U, Lucas H, Meister A, Ramos G, Loppnow H, Mäder K. Phosphatidylserine (PS) and phosphatidylglycerol (PG) nanodispersions as potential anti-inflammatory therapeutics: Comparison of in vitro activity and impact of pegylation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 23:102096. [DOI: 10.1016/j.nano.2019.102096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 07/01/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023]
|
34
|
Synergy of Phospholipid-Drug Formulations Significantly Deactivates Profibrogenic Human Hepatic Stellate Cells. Pharmaceutics 2019; 11:pharmaceutics11120676. [PMID: 31842373 PMCID: PMC6969915 DOI: 10.3390/pharmaceutics11120676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023] Open
Abstract
The pivotal role of hepatic stellate cells (HSCs) in orchestrating the bidirectional process of progression and regression of liver fibrosis makes them an ideal target for exploring new antifibrotic therapies. Essential phospholipids (EPLs), with their polyenylphosphatidylcholine (PPC) fraction, either alone or combined with other hepatoprotective substances such as silymarin, are recommended in hepatic impairment, but a scientific rationale for their use is still lacking. Herein, we compared the ability of EPLs to restore quiescent-like features in HSCs with that of dilinoleoylphosphatidylcholine (DLPC), PPC fraction’s main component. Specifically, we screened at the cellular level the antifibrotic effects of PPC formulations in the presence and absence of silymarin, by using LX-2 cells (pro-fibrogenic HSCs) and by assessing the main biochemical hallmarks of the activated and deactivated states of this cell line. We also proved the formulations’ direct effect on the motional order of cell membranes of adherent cells. LX-2 cells, examined for lipid droplets as a quiescence marker, showed that PPCs led to a more prominent deactivation than DLPC. This result was confirmed by a reduction of collagen and α-SMA expression, and by a profound alteration in the cell membrane fluidity. PPC–silymarin formulations deactivated HSCs with a significant synergistic effect. The remarkable bioactivity of PPCs in deactivating fibrogenic HSCs paves the way for the rational design of new therapeutics aimed at managing hepatic fibrosis.
Collapse
|
35
|
Zhang Y, Pu C, Tang W, Wang S, Sun Q. Gallic acid liposomes decorated with lactoferrin: Characterization, in vitro digestion and antibacterial activity. Food Chem 2019; 293:315-322. [DOI: 10.1016/j.foodchem.2019.04.116] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/28/2019] [Accepted: 04/30/2019] [Indexed: 01/05/2023]
|
36
|
Adjuvant Strategies for More Effective Tuberculosis Vaccine Immunity. Microorganisms 2019; 7:microorganisms7080255. [PMID: 31409028 PMCID: PMC6724148 DOI: 10.3390/microorganisms7080255] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/03/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis infection is responsible for the most deaths by a single infectious agent worldwide, with 1.6 million deaths in 2017 alone. The World Health Organization, through its "End TB" strategy, aims to reduce TB deaths by 95% by 2035. In order to reach this goal, a more effective vaccine than the Bacillus Calmette-Guerin (BCG) vaccine currently in use is needed. Subunit TB vaccines are ideal candidates, because they can be used as booster vaccinations for individuals who have already received BCG and would also be safer for use in immunocompromised individuals in whom BCG is contraindicated. However, subunit TB vaccines will almost certainly require formulation with a potent adjuvant. As the correlates of vaccine protection against TB are currently unclear, there are a variety of adjuvants currently being used in TB vaccines in preclinical and clinical development. This review describes the various adjuvants in use in TB vaccines, their effectiveness, and their proposed mechanisms of action. Notably, adjuvants with less inflammatory and reactogenic profiles that can be administered safely via mucosal routes, may have the biggest impact on future directions in TB vaccine design.
Collapse
|
37
|
|
38
|
Breitsamer M, Winter G. Vesicular phospholipid gels as drug delivery systems for small molecular weight drugs, peptides and proteins: State of the art review. Int J Pharm 2018; 557:1-8. [PMID: 30572079 DOI: 10.1016/j.ijpharm.2018.12.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 10/27/2022]
Abstract
Lipid-based drug delivery has been investigated for a long time when it comes to liposomes and solid-lipid implants or solid-lipid nanoparticles. The promising, characteristic properties of these systems have led to the development of newer lipid-based drug delivery systems for the sustained release of drugs like liposomes for sustained delivery of substances, DepoFoam™ technology, phospholipid-based phase separation gels and vesicular phospholipid gels. Vesicular phospholipid gels (VPGs) are highly concentrated, viscous dispersions of high amounts of phospholipids in aqueous drug solution. The easy, solvent-free manufacturing process, high biocompatibility and various applications, as depot formulation for the sustained delivery of drugs and as a storage form of small unilamellar vesicles make VPGs highly attractive as drug carriers. Over the last years, the solvent free preparation process has advanced from high pressure homogenization to dual centrifugation (DC). Thereby a very simple one step process has been established for the preparation of VPGs. The semisolid VPG was first described in 1997 by Brandl et al. Since then, many formulations have been developed, encapsulating small molecular weight drugs like 5-FU (2003), cetrorelix (2005), cytarabine (2012) and exenatide (2015). In 2010, the first pharmaceutical protein, erythropoietin, was encapsulated in VPGs and sustained release of the substance was shown in vitro. In 2015, G-CSF was encapsulated in VPGs and tested in vivo for rotator cuff repair in a rat model and for PEGylated IFN-β-1b sustained release from vesicular phospholipid gels was demonstrated in vitro. Further, a very elegant administration technique for VPGs via needle-free injection was established. However this promising drug delivery system does still leave space for improvement and optimization. This review summarizes information about lipid-based depot systems in general and focuses on the historical development of VPGs. It emphasizes the advantages and drawbacks of VPGs as drug delivery device. Additionally, novel preparation methods and applications of VPGs will be discussed. A focus will be set on delivery of pharmaceutical proteins and peptides.
Collapse
Affiliation(s)
- Michaela Breitsamer
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Munich, Germany.
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|