1
|
Zhang M, Xiang C, Niu R, He X, Luo W, Liu W, Gu R. Liposomes as versatile agents for the management of traumatic and nontraumatic central nervous system disorders: drug stability, targeting efficiency, and safety. Neural Regen Res 2025; 20:1883-1899. [PMID: 39254548 DOI: 10.4103/nrr.nrr-d-24-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/28/2024] [Indexed: 09/11/2024] Open
Abstract
Various nanoparticle-based drug delivery systems for the treatment of neurological disorders have been widely studied. However, their inability to cross the blood-brain barrier hampers the clinical translation of these therapeutic strategies. Liposomes are nanoparticles composed of lipid bilayers, which can effectively encapsulate drugs and improve drug delivery across the blood-brain barrier and into brain tissue through their targeting and permeability. Therefore, they can potentially treat traumatic and nontraumatic central nervous system diseases. In this review, we outlined the common properties and preparation methods of liposomes, including thin-film hydration, reverse-phase evaporation, solvent injection techniques, detergent removal methods, and microfluidics techniques. Afterwards, we comprehensively discussed the current applications of liposomes in central nervous system diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, traumatic brain injury, spinal cord injury, and brain tumors. Most studies related to liposomes are still in the laboratory stage and have not yet entered clinical trials. Additionally, their application as drug delivery systems in clinical practice faces challenges such as drug stability, targeting efficiency, and safety. Therefore, we proposed development strategies related to liposomes to further promote their development in neurological disease research.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Cheng Y, Hay CD, Mahuttanatan SM, Hindley JW, Ces O, Elani Y. Microfluidic technologies for lipid vesicle generation. LAB ON A CHIP 2024; 24:4679-4716. [PMID: 39323383 PMCID: PMC11425070 DOI: 10.1039/d4lc00380b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024]
Abstract
Encapsulating biological and non-biological materials in lipid vesicles presents significant potential in both industrial and academic settings. When smaller than 100 nm, lipid vesicles and lipid nanoparticles are ideal vehicles for drug delivery, facilitating the delivery of payloads, improving pharmacokinetics, and reducing the off-target effects of therapeutics. When larger than 1 μm, vesicles are useful as model membranes for biophysical studies, as synthetic cell chassis, as bio-inspired supramolecular devices, and as the basis of protocells to explore the origin of life. As applications of lipid vesicles gain prominence in the fields of nanomedicine, biotechnology, and synthetic biology, there is a demand for advanced technologies for their controlled construction, with microfluidic methods at the forefront of these developments. Compared to conventional bulk methods, emerging microfluidic methods offer advantages such as precise size control, increased production throughput, high encapsulation efficiency, user-defined membrane properties (i.e., lipid composition, vesicular architecture, compartmentalisation, membrane asymmetry, etc.), and potential integration with lab-on-chip manipulation and analysis modules. We provide a review of microfluidic lipid vesicle generation technologies, focusing on recent advances and state-of-the-art techniques. Principal technologies are described, and key research milestones are highlighted. The advantages and limitations of each approach are evaluated, and challenges and opportunities for microfluidic engineering of lipid vesicles to underpin a new generation of therapeutics, vaccines, sensors, and bio-inspired technologies are presented.
Collapse
Affiliation(s)
- Yu Cheng
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Callum D Hay
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Suchaya M Mahuttanatan
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - James W Hindley
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Oscar Ces
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Yuval Elani
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK.
- Department of Chemical Engineering, Imperial College London, London, UK
| |
Collapse
|
3
|
Rahman MM, Wang J, Wang G, Su Z, Li Y, Chen Y, Meng J, Yao Y, Wang L, Wilkens S, Tan J, Luo J, Zhang T, Zhu C, Cho SH, Wang L, Lee LP, Wan Y. Chimeric nanobody-decorated liposomes by self-assembly. NATURE NANOTECHNOLOGY 2024; 19:818-824. [PMID: 38374413 DOI: 10.1038/s41565-024-01620-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024]
Abstract
Liposomes as drug vehicles have advantages, such as payload protection, tunable carrying capacity and improved biodistribution. However, due to the dysfunction of targeting moieties and payload loss during preparation, immunoliposomes have yet to be favoured in commercial manufacturing. Here we report a chemical modification-free biophysical approach for producing immunoliposomes in one step through the self-assembly of a chimeric nanobody (cNB) into liposome bilayers. cNB consists of a nanobody against human epidermal growth factor receptor 2 (HER2), a flexible peptide linker and a hydrophobic single transmembrane domain. We determined that 64% of therapeutic compounds can be encapsulated into 100-nm liposomes, and up to 2,500 cNBs can be anchored on liposomal membranes without steric hindrance under facile conditions. Subsequently, we demonstrate that drug-loaded immunoliposomes increase cytotoxicity on HER2-overexpressing cancer cell lines by 10- to 20-fold, inhibit the growth of xenograft tumours by 3.4-fold and improve survival by more than twofold.
Collapse
Affiliation(s)
- Md Mofizur Rahman
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
- Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Jing Wang
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Oncology and Hematology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, China
| | - Guosheng Wang
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhipeng Su
- Nanjing Regenecore Biotech Co. Ltd., Nanjing, China
| | - Yizeng Li
- Biophysics and Mathematical Biology Lab, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
| | - Yundi Chen
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
| | - Jinguo Meng
- Nanjing Regenecore Biotech Co. Ltd., Nanjing, China
| | - Yao Yao
- Nanjing Regenecore Biotech Co. Ltd., Nanjing, China
| | - Lefei Wang
- Nanjing Regenecore Biotech Co. Ltd., Nanjing, China
| | - Stephan Wilkens
- Department of Biochemistry and Molecular Biology, Upstate Medical University, Syracuse, NY, USA
| | - Jifu Tan
- Department of Mechanical Engineering, Northern Illinois University, Dekalb, IL, USA
| | - Juntao Luo
- Department of Pharmacology, Upstate Medical University, Syracuse, NY, USA
| | - Tao Zhang
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Johnson City, NY, USA
| | - Chuandong Zhu
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
- Department of Radiotherapy, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sung Hyun Cho
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Lixue Wang
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA.
- Department of Radiotherapy, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Luke P Lee
- Harvard Medical School, Harvard University; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, USA.
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea.
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea.
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA.
| |
Collapse
|
4
|
Akar S, Fardindoost S, Hoorfar M. High throughput microfluidics-based synthesis of PEGylated liposomes for precise size control and efficient drug encapsulation. Colloids Surf B Biointerfaces 2024; 238:113926. [PMID: 38677154 DOI: 10.1016/j.colsurfb.2024.113926] [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: 01/24/2024] [Revised: 04/08/2024] [Accepted: 04/20/2024] [Indexed: 04/29/2024]
Abstract
The low scalability and reproducibility of existing synthesis methods have hindered the translation of liposome nanoparticles as carriers for targeted drug delivery from conventional laboratory techniques to mass production. To this end, in this study, we present a high-throughput microfluidics-based approach for the synthesis of PEGylated liposomes with a primary focus on achieving precise size control and efficient encapsulation of hydrophobic drug molecules. In this platform, liposomes were self-assembled through a controllable mixing of lipids (EYPC, cholesterol, and DSPE-PEG 2000) dissolved in ethanol and an aqueous solution. The key parameters, including the chip design, total flow rate, flow rate ratio, lipid concentrations, as well as variations in buffer (HEPES and NaCl) and solvent composition (commercial and reagent-grade ethanol) were explored in detail. Through comprehensive parametric studies, we gained valuable insights into the influence of these variables on the size distribution of liposomes and succeeded in producing highly reproducible liposomes ranging from approximately 60 nm (corresponding to small unilamellar vesicles) to 150 nm (representing large unilamellar vesicles), all while maintaining a polydispersity index (PDI) of less than 0.2. To assess the encapsulation efficiency of hydrophobic drug molecules, Nile red (NR) was employed as a surrogate. We meticulously examined the impact of NR concentration on the drug encapsulation process, resulting in up to 74% drug encapsulation efficiency within the PEGylated liposomes. This research offers crucial advances in liposome synthesis and drug delivery, providing a high-throughput, controllable method for PEGylated liposomes with potential in pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- Shima Akar
- School of Engineering and Computer Science, University of Victoria, Victoria, BC, Canada
| | - Somayeh Fardindoost
- School of Engineering and Computer Science, University of Victoria, Victoria, BC, Canada
| | - Mina Hoorfar
- School of Engineering and Computer Science, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
5
|
Xu H, Wang Z, Wei W, Li T, Duan X. Microfluidic confined acoustic streaming vortex for liposome synthesis. LAB ON A CHIP 2024; 24:2802-2810. [PMID: 38693825 DOI: 10.1039/d4lc00184b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Liposomes have garnered significant attention owing to their favorable characteristics as promising carriers. Microfluidic based hydrodynamic flow focusing, or micro-mixing approaches enable precise control of liposome size during their synthesis due to the comparable size scale. However, current microfluidic approaches still have issues such as high flow rate dependency, complex chip structures, and ease of clogging. Herein, we present a novel microfluidic platform for size-tunable liposome synthesis based on an ultra-high-frequency acoustic resonator. By designing the shape and orientation of the acoustic resonator in the three-phase laminar flow, it combined the features of both hydrodynamic flow focusing and rapid micro-mixing. The distribution of lipid precursor solution in laminar flow and the mixing conditions could be regulated by the confined acoustic streaming vortex. We successfully synthesize liposomes with adjustable sizes and narrow size distributions. Notably, this platform regulates the product size by adjusting only the input power, which is less dependent on the flow rate. Furthermore, the vortex-like fluid flow generated along the device edge effectively prevents precipitation due to excessive lipid concentration or contact with the wall.
Collapse
Affiliation(s)
- Huihui Xu
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| | - Zhaoxun Wang
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| | - Wei Wei
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| | - Tiechuan Li
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| | - Xuexin Duan
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
6
|
Mohammadi M, Ahmed Qadir S, Mahmood Faraj A, Hamid Shareef O, Mahmoodi H, Mahmoudi F, Moradi S. Navigating the future: Microfluidics charting new routes in drug delivery. Int J Pharm 2024:124142. [PMID: 38648941 DOI: 10.1016/j.ijpharm.2024.124142] [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: 10/12/2023] [Revised: 03/30/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Microfluidics has emerged as a transformative force in the field of drug delivery, offering innovative avenues to produce a diverse range of nano drug delivery systems. Thanks to its precise manipulation of small fluid volumes and its exceptional command over the physicochemical characteristics of nanoparticles, this technology is notably able to enhance the pharmacokinetics of drugs. It has initiated a revolutionary phase in the domain of drug delivery, presenting a multitude of compelling advantages when it comes to developing nanocarriers tailored for the delivery of poorly soluble medications. These advantages represent a substantial departure from conventional drug delivery methodologies, marking a paradigm shift in pharmaceutical research and development. Furthermore, microfluidic platformsmay be strategically devised to facilitate targeted drug delivery with the objective of enhancing the localized bioavailability of pharmaceutical substances. In this paper, we have comprehensively investigated a range of significant microfluidic techniques used in the production of nanoscale drug delivery systems. This comprehensive review can serve as a valuable reference and offer insightful guidance for the development and optimization of numerous microfluidics-fabricated nanocarriers.
Collapse
Affiliation(s)
- Mohammad Mohammadi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Syamand Ahmed Qadir
- Department of Medical Laboratory Techniques, Halabja Technical Institute, Research Center, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Aryan Mahmood Faraj
- Department of Medical Laboratory Sciences, Halabja Technical College of Applied Sciences, Sulaimani Polytechnic University, Halabja, Iraq
| | - Osama Hamid Shareef
- Department of Medical Laboratory Techniques, Halabja Technical Institute, Research Center, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Hassan Mahmoodi
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mahmoudi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
7
|
Chai C, Park J. Food liposomes: Structures, components, preparations, and applications. Food Chem 2024; 432:137228. [PMID: 37633138 DOI: 10.1016/j.foodchem.2023.137228] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 08/28/2023]
Abstract
This review explores liposomes, focusing on their structure, components, the characteristics influencing their stability and applicability in foods, and preparation methods. The role of phospholipids and liposome modulators in preparing liposomes of desired structure and size is emphasized. The potential of liposomes to enhance food value through liposomal encapsulation and delivery of functional substances is reviewed. Conventional and advanced liposome preparation methods are reviewed, underscoring their impact on the marketability of liposomes. The review highlights the need for research into lecithin properties and modulators that enhance liposome stability. The need to develop cost-effective and rapid liposome preparation methods is identified as a key factor in improving the marketability of food liposomes and promoting their use in foods.
Collapse
Affiliation(s)
- Changhoon Chai
- Department of Applied Animal Science, Kangwon National University, Chuncheon-si 24341, Republic of Korea.
| | - Jinhyung Park
- Department of Applied Animal Science, Kangwon National University, Chuncheon-si 24341, Republic of Korea
| |
Collapse
|
8
|
Lin WZS, Bostic WKV, Malmstadt N. 3D-printed microfluidic device for high-throughput production of lipid nanoparticles incorporating SARS-CoV-2 spike protein mRNA. LAB ON A CHIP 2024; 24:162-170. [PMID: 38165143 PMCID: PMC10853008 DOI: 10.1039/d3lc00520h] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Lipid nanoparticles (LNPs) are drug carriers for protecting nucleic acids for cellular delivery. The first mRNA vaccines authorized by the United States Food and Drug Administration are the mRNA-1273 (Moderna) and BNT162b (BioNTech/Pfizer) vaccines against coronavirus disease 2019 (COVID-19). We designed a 3D printed Omnidirectional Sheath-flow Enabled Microfluidics (OSEM) device for producing mRNA-loaded LNPs that closely resemble the Moderna vaccine: we used the same lipid formulations to encapsulate mRNA encoding SARS-CoV-2 spike protein. The OSEM device is made of durable methacrylate-based materials that can support flow rates in the mL min-1 range and was fabricated by stereolithography (SLA), incorporating readily adaptable interfaces using commercial fluidic connectors. Two key features of the OSEM device are: 1) a 4-way hydrodynamic flow focusing region and 2) a staggered herringbone mixer (SHM). Superior to conventional planar fluid junctions, the 4-way sheath flow channel generates an evenly focused, circular center flow that facilitates the formation of LNPs with low polydispersity. Downstream, fluid mixing in the SHM is intensified by incorporating a zig-zag fluidic pathway to deliver high mRNA encapsulation efficiency. We characterized the mRNA-loaded LNPs produced in the OSEM device and showed that the enhanced 3D microfluidic structures enable a 5-fold higher throughput production rate (60 mL min-1) of LNPs compared to commercial multi-thousand-dollar micromixers. The device produced LNPs of diameter less than 90 nm, with low polydispersity (2-8%) and high mRNA encapsulation efficiency (>90%). The 3D-printed device provides a cost-effective and easily prepared solution for high-throughput LNP production.
Collapse
Affiliation(s)
- Wan-Zhen Sophie Lin
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, USA.
| | - William Kristian Vu Bostic
- USC Biomedical Engineering Department, University of Southern California, Los Angeles, California 90089, USA
| | - Noah Malmstadt
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, USA.
- USC Biomedical Engineering Department, University of Southern California, Los Angeles, California 90089, USA
- Department of Chemistry, University of Southern California, Los Angeles, California 90089, USA
- USC Norris Comprehensive Cancer Center, Los Angeles, California 90033, USA
| |
Collapse
|
9
|
Zhang H, Yang J, Sun R, Han S, Yang Z, Teng L. Microfluidics for nano-drug delivery systems: From fundamentals to industrialization. Acta Pharm Sin B 2023; 13:3277-3299. [PMID: 37655333 PMCID: PMC10466004 DOI: 10.1016/j.apsb.2023.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/10/2022] [Accepted: 12/15/2022] [Indexed: 01/27/2023] Open
Abstract
In recent years, owing to the miniaturization of the fluidic environment, microfluidic technology offers unique opportunities for the implementation of nano drug delivery systems (NDDSs) production processes. Compared with traditional methods, microfluidics improves the controllability and uniformity of NDDSs. The fast mixing and laminar flow properties achieved in the microchannels can tune the physicochemical properties of NDDSs, including particle size, distribution and morphology, resulting in narrow particle size distribution and high drug-loading capacity. The success of lipid nanoparticles encapsulated mRNA vaccines against coronavirus disease 2019 by microfluidics also confirmed its feasibility for scaling up the preparation of NDDSs via parallelization or numbering-up. In this review, we provide a comprehensive summary of microfluidics-based NDDSs, including the fundamentals of microfluidics, microfluidic synthesis of NDDSs, and their industrialization. The challenges of microfluidics-based NDDSs in the current status and the prospects for future development are also discussed. We believe that this review will provide good guidance for microfluidics-based NDDSs.
Collapse
Affiliation(s)
- Huan Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jie Yang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Rongze Sun
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Songren Han
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
10
|
Composite Norland Optical Adhesive (NOA)/silicon flow focusing devices for colloidal particle manipulation and synthesis. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
11
|
Zheng H, Tao H, Wan J, Lee KY, Zheng Z, Leung SSY. Preparation of Drug-Loaded Liposomes with Multi-Inlet Vortex Mixers. Pharmaceutics 2022; 14:pharmaceutics14061223. [PMID: 35745796 PMCID: PMC9227628 DOI: 10.3390/pharmaceutics14061223] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
The multi-inlet vortex mixer (MIVM) has emerged as a novel bottom-up technology for solid nanoparticle preparation. However, its performance in liposome preparation remains unknown. Here, two key process parameters (aqueous/organic flow rate ratio (FRR) and total flow rate (TFR)) of MIVM were investigated for liposome preparation. For this study, two model drugs (lysozyme and erythromycin) were chosen for liposome encapsulation as the representative hydrophilic and hydrophobic drugs, respectively. In addition, two modified MIVMs, one with herringbone-patterned straight inlets and one with zigzag inlets, were designed to further improve the mixing efficiency, aiming to achieve better drug encapsulation. Data showed that FRR played an important role in liposome size control, and a size of <200 nm was achieved by FRR higher than 3:1. Moreover, increasing TFR (from 1 to 100 mL/min) could further decrease the size at a given FRR. However, similar regularities in controlling the encapsulation efficiency (EE%) were only noted in erythromycin-loaded liposomes. Modified MIVMs improved the EE% of lysozyme-loaded liposomes by 2~3 times at TFR = 40 mL/min and FRR = 3:1, which was consistent with computational fluid dynamics simulations. In summary, the good performance of MIVM in the control of particle size and EE% makes it a promising tool for liposome preparation, especially for hydrophobic drug loading, at flexible production scales.
Collapse
Affiliation(s)
- Huangliang Zheng
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong; (H.Z.); (K.Y.L.)
| | - Hai Tao
- Center for Turbulence Control, Harbin Institute of Technology, Shenzhen 518055, China; (H.T.); (J.W.)
| | - Jinzhao Wan
- Center for Turbulence Control, Harbin Institute of Technology, Shenzhen 518055, China; (H.T.); (J.W.)
| | - Kei Yan Lee
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong; (H.Z.); (K.Y.L.)
| | - Zhanying Zheng
- Center for Turbulence Control, Harbin Institute of Technology, Shenzhen 518055, China; (H.T.); (J.W.)
- Correspondence: (Z.Z.); (S.S.Y.L.)
| | - Sharon Shui Yee Leung
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong; (H.Z.); (K.Y.L.)
- Correspondence: (Z.Z.); (S.S.Y.L.)
| |
Collapse
|
12
|
Recent Development of Drug Delivery Systems through Microfluidics: From Synthesis to Evaluation. Pharmaceutics 2022; 14:pharmaceutics14020434. [PMID: 35214166 PMCID: PMC8880124 DOI: 10.3390/pharmaceutics14020434] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 01/04/2023] Open
Abstract
Conventional drug administration usually faces the problems of degradation and rapid excretion when crossing many biological barriers, leading to only a small amount of drugs arriving at pathological sites. Therapeutic drugs delivered by drug delivery systems to the target sites in a controlled manner greatly enhance drug efficacy, bioavailability, and pharmacokinetics with minimal side effects. Due to the distinct advantages of microfluidic techniques, microfluidic setups provide a powerful tool for controlled synthesis of drug delivery systems, precisely controlled drug release, and real-time observation of drug delivery to the desired location at the desired rate. In this review, we present an overview of recent advances in the preparation of nano drug delivery systems and carrier-free drug delivery microfluidic systems, as well as the construction of in vitro models on-a-chip for drug efficiency evaluation of drug delivery systems. We firstly introduce the synthesis of nano drug delivery systems, including liposomes, polymers, and inorganic compounds, followed by detailed descriptions of the carrier-free drug delivery system, including micro-reservoir and microneedle drug delivery systems. Finally, we discuss in vitro models developed on microfluidic devices for the evaluation of drug delivery systems, such as the blood–brain barrier model, vascular model, small intestine model, and so on. The opportunities and challenges of the applications of microfluidic platforms in drug delivery systems, as well as their clinical applications, are also discussed.
Collapse
|
13
|
Behravan N, Zahedipour F, Jaafari MR, Johnston TP, Sahebkar A. Lipid-based nanoparticulate delivery systems for HER2-positive breast cancer immunotherapy. Life Sci 2022; 291:120294. [PMID: 34998838 DOI: 10.1016/j.lfs.2021.120294] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Lipid-based nanoparticulate delivery platforms such as liposomes help overcome cell and tissue barriers and allow prolonged therapeutic plasma drug concentrations, simultaneous targeting of tumor tissue, and increased bioavailability of numerous drugs used for treatment of cancer. The human epidermal growth factor receptor, HER2, is an important player in the pathogenesis of breast cancer and is considered a potential cancer biomarker for the design of immunotherapeutics. HER2-positive breast cancer is found in up to 30% of breast cancer patients. Currently, a variety of lipid nanoparticulate systems are being evaluated in preclinical settings and in clinical trials for targeting HER2-positive breast cancer. Advances in functionalized anti-HER2 lipid nanoparticulates have demonstrated promise and may lead to the development of new nano-immunotherapy protocols against HER2 positive breast cancer. Here we present a review of the most up-to-date literature, including our own research, on the use of lipid nanoparticulate carriers in immunotherapy of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Nima Behravan
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- 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
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Lin WZS, Evenson WE, Bostic WKV, Roberts RW, Malmstadt N. Compatibility of Popular Three-Dimensional Printed Microfluidics Materials with In Vitro Enzymatic Reactions. ACS APPLIED BIO MATERIALS 2022; 5:818-824. [PMID: 35138792 PMCID: PMC10371121 DOI: 10.1021/acsabm.1c01180] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
3D printed microfluidics offer several advantages over conventional planar microfabrication techniques including fabrication of 3D microstructures, rapid prototyping, and inertness. While 3D printed materials have been studied for their biocompatibility in cell and tissue culture applications, their compatibility for in vitro biochemistry and molecular biology has not been systematically investigated. Here, we evaluate the compatibility of several common enzymatic reactions in the context of 3D-printed microfluidics: (1) polymerase chain reaction (PCR), (2) T7 in vitro transcription, (3) mammalian in vitro translation, and (4) reverse transcription. Surprisingly, all the materials tested significantly inhibit one or more of these in vitro enzymatic reactions. Inclusion of BSA mitigates only some of these inhibitory effects. Overall, inhibition appears to be due to a combination of the surface properties of the resins as well as soluble components (leachate) originating in the matrix.
Collapse
Affiliation(s)
- Wan-Zhen Sophie Lin
- Mork Family Department of Chemical Engineering and Materials Science, 925 Bloom Walk, HED 216, Los Angeles, California 90089, United States
| | - William E Evenson
- Department of Chemistry, University of Southern California, 3620 McClintock Ave, SGM 418, Los Angeles, California 90089, United States
| | - W Kristian Vu Bostic
- USC Biomedical Engineering Department, 1042 Downey Way, Denney Research Center (DRB) 140, Los Angeles, California 90089, United States
| | - Richard W Roberts
- Mork Family Department of Chemical Engineering and Materials Science, 925 Bloom Walk, HED 216, Los Angeles, California 90089, United States.,Department of Chemistry, University of Southern California, 3620 McClintock Ave, SGM 418, Los Angeles, California 90089, United States.,USC Biomedical Engineering Department, 1042 Downey Way, Denney Research Center (DRB) 140, Los Angeles, California 90089, United States.,USC Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, California 90033, United States.,Department of Molecular and Computational Biology, University of Southern California, 1050 Child Way, RRI 201, Los Angeles, California 90089, United States
| | - Noah Malmstadt
- Mork Family Department of Chemical Engineering and Materials Science, 925 Bloom Walk, HED 216, Los Angeles, California 90089, United States.,Department of Chemistry, University of Southern California, 3620 McClintock Ave, SGM 418, Los Angeles, California 90089, United States.,USC Biomedical Engineering Department, 1042 Downey Way, Denney Research Center (DRB) 140, Los Angeles, California 90089, United States.,USC Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, California 90033, United States
| |
Collapse
|
15
|
da Costa OMMM, Firmino PCOS, Strapasson GB, de la Torre LG, Malfatti-Gasperini AA, Júnior SA. One-step Production of Sterically Stabilized Anionic Nanoliposome Using Microfluidic Device. J Oleo Sci 2022; 71:515-522. [DOI: 10.5650/jos.ess21309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
16
|
Microfluidics for Multiphase Mixing and Liposomal Encapsulation of Nanobioconjugates: Passive vs. Acoustic Systems. FLUIDS 2021. [DOI: 10.3390/fluids6090309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One of the main routes to ensure that biomolecules or bioactive agents remain active as they are incorporated into products with applications in different industries is by their encapsulation. Liposomes are attractive platforms for encapsulation due to their ease of synthesis and manipulation and the potential to fuse with cell membranes when they are intended for drug delivery applications. We propose encapsulating our recently developed cell-penetrating nanobioconjugates based on magnetite interfaced with translocating proteins and peptides with the purpose of potentiating their cell internalization capabilities even further. To prepare the encapsulates (also known as magnetoliposomes (MLPs)), we introduced a low-cost microfluidic device equipped with a serpentine microchannel to favor the interaction between the liposomes and the nanobioconjugates. The encapsulation performance of the device, operated either passively or in the presence of ultrasound, was evaluated both in silico and experimentally. The in silico analysis was implemented through multiphysics simulations with the software COMSOL Multiphysics 5.5® (COMSOL Inc., Stockholm, Sweden) via both a Eulerian model and a transport of diluted species model. The encapsulation efficiency was determined experimentally, aided by spectrofluorimetry. Encapsulation efficiencies obtained experimentally and in silico approached 80% for the highest flow rate ratios (FRRs). Compared with the passive mixer, the in silico results of the device under acoustic waves led to higher discrepancies with respect to those obtained experimentally. This was attributed to the complexity of the process in such a situation. The obtained MLPs demonstrated successful encapsulation of the nanobioconjugates by both methods with a 36% reduction in size for the ones obtained in the presence of ultrasound. These findings suggest that the proposed serpentine micromixers are well suited to produce MLPs very efficiently and with homogeneous key physichochemical properties.
Collapse
|
17
|
Liposomes: Advancements and innovation in the manufacturing process. Adv Drug Deliv Rev 2020; 154-155:102-122. [PMID: 32650041 DOI: 10.1016/j.addr.2020.07.002] [Citation(s) in RCA: 279] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/13/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
Liposomes are well recognised as effective drug delivery systems, with a range of products approved, including follow on generic products. Current manufacturing processes used to produce liposomes are generally complex multi-batch processes. Furthermore, liposome preparation processes adopted in the laboratory setting do not offer easy translation to large scale production, which may delay the development and adoption of new liposomal systems. To promote advancement and innovation in liposome manufacturing processes, this review considers the range of manufacturing processes available for liposomes, from laboratory scale and scale up, through to large-scale manufacture and evaluates their advantages and limitations. The regulatory considerations associated with the manufacture of liposomes is also discussed. New innovations that support leaner scalable technologies for liposome fabrication are outlined including self-assembling liposome systems and microfluidic production. The critical process attributes that impact on the liposome product attributes are outlined to support potential wider adoption of these innovations.
Collapse
|
18
|
Ni M, Tresset G, Iliescu C, Hauser CAE. Ultrashort Peptide Theranostic Nanoparticles by Microfluidic-Assisted Rapid Solvent Exchange. IEEE Trans Nanobioscience 2020; 19:627-632. [PMID: 32746332 DOI: 10.1109/tnb.2020.3007103] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ultrashort peptides (USPs), composed of three to seven amino acids, can self-assemble into nanofibers in pure water. Here, using hydrodynamic focusing and a solvent exchange method on a microfluidic setup, we convert these nanofibers into globular nanoparticles with excellent dimensional control and polydispersity. Thanks to USP nanocarriers' structure, different drugs can be loaded. We used Curcumin as a model drug to evaluate the performance of USP nanocarriers as a novel drug delivery vehicle. These nanoparticles can efficiently cross the cell membrane and possess nonlinear optical properties. Therefore, we envisage USP nanoparticles as promising future theranostic nanocarriers.
Collapse
|
19
|
Zartner L, Muthwill MS, Dinu IA, Schoenenberger CA, Palivan CG. The rise of bio-inspired polymer compartments responding to pathology-related signals. J Mater Chem B 2020; 8:6252-6270. [PMID: 32452509 DOI: 10.1039/d0tb00475h] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Self-organized nano- and microscale polymer compartments such as polymersomes, giant unilamellar vesicles (GUVs), polyion complex vesicles (PICsomes) and layer-by-layer (LbL) capsules have increasing potential in many sensing applications. Besides modifying the physicochemical properties of the corresponding polymer building blocks, the versatility of these compartments can be markedly expanded by biomolecules that endow the nanomaterials with specific molecular and cellular functions. In this review, we focus on polymer-based compartments that preserve their structure, and highlight the key role they play in the field of medical diagnostics: first, the self-assembling abilities that result in preferred architectures are presented for a broad range of polymers. In the following, we describe different strategies for sensing disease-related signals (pH-change, reductive conditions, and presence of ions or biomolecules) by polymer compartments that exhibit stimuli-responsiveness. In particular, we distinguish between the stimulus-sensitivity contributed by the polymer itself or by additional compounds embedded in the compartments in different sensing systems. We then address necessary properties of sensing polymeric compartments, such as the enhancement of their stability and biocompatibility, or the targeting ability, that open up new perspectives for diagnostic applications.
Collapse
Affiliation(s)
- Luisa Zartner
- Chemistry Department, University of Basel, Mattenstr. 24a, BPR1096, Basel, Switzerland.
| | | | | | | | | |
Collapse
|
20
|
Webb C, Forbes N, Roces CB, Anderluzzi G, Lou G, Abraham S, Ingalls L, Marshall K, Leaver TJ, Watts JA, Aylott JW, Perrie Y. Using microfluidics for scalable manufacturing of nanomedicines from bench to GMP: A case study using protein-loaded liposomes. Int J Pharm 2020; 582:119266. [DOI: 10.1016/j.ijpharm.2020.119266] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
|