1
|
Soto-Arriaza M, Cena Ahumada E, Bonardd S, Melendez J. Calcein release from DPPC liposomes by phospholipase A2 activity: Effect of cholesterol and amphipathic copolymers. J Liposome Res 2024; 34:617-629. [PMID: 38850012 DOI: 10.1080/08982104.2024.2361610] [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: 08/02/2023] [Revised: 05/09/2024] [Accepted: 05/25/2024] [Indexed: 06/09/2024]
Abstract
In this study, we evaluated the impact of incorporating diblock and triblock amphiphilic copolymers, as well as cholesterol into DPPC liposomes on the release of a model molecule, calcein, mediated by exogenous phospholipase A2 activity. Our findings show that calcein release slows down in the presence of copolymers at low concentration, while at high concentration, the calcein release profile resembles that of the DPPC control. Additionally, calcein release mediated by exogenous PLA2 decreases as the amount of solubilized cholesterol increases, with a maximum between 18 mol% and 20 mol%. At concentrations higher than 24 mol%, no calcein release was observed. Studies conducted on HEK-293 and HeLa cells revealed that DPPC liposomes reduced viability by only 5% and 12%, respectively, after 3 hours of incubation, while DPPC liposome in presence of 33 mol% of Cholesterol reduced viability by approximately 11% and 23%, respectively, during the same incubation period. For formulations containing copolymers at low and high concentrations, cell viability decreased by approximately 20% and 40%, respectively, after 3 hours of incubation. Based on these preliminary results, we can conclude that the presence of amphiphilic copolymers at low concentration can be used in the design of new DPPC liposomes, and together with cholesterol, they can modulate liposome stabilization. The new formulations showed low cytotoxicity in HEK-293 cells, and it was observed that calcein release depended entirely on PLA2 activity and the presence of calcium ions.
Collapse
Affiliation(s)
- Marco Soto-Arriaza
- Escuela de Química y Farmacia, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Eduardo Cena Ahumada
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Sebastián Bonardd
- Centro de Física de Materiales (CSIC, UPV/EHU)-Materials Physics Center (MPC), Donostia-San Sebastían, Spain
- Department of Polymers and Advanced Materials: Physics, Chemistry and Technology, University of the Basque Country UPV/EHU, Donostia-San Sebastian, Spain
| | | |
Collapse
|
2
|
Inoue KI, Yamamoto T, Hatori Y, Hiraide T, Ye S. Hydrolysis of phospholipid monolayers by phospholipase A2 revealed by heterodyne-detected sum frequency generation (HD-SFG) spectroscopy. J Chem Phys 2024; 161:154704. [PMID: 39404221 DOI: 10.1063/5.0231282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Phospholipase A2 (PLA2) catalyzes the hydrolysis of the sn-2 acyl ester linkage in phospholipid, producing lysophospholipid and fatty acid in the presence of Ca2+. The hydrolysis mediated by PLA2 has attracted much interest in various fields, such as pharmacy and biotechnology. It is recognized that PLA2 cannot hydrolyze phospholipid monolayers at high surface coverage. However, the origin of different PLA2 activities is not fully understood yet. The present study investigated the interaction between DPPC (16:0 PC) monolayer and PLA2 using heterodyne-detected sum frequency generation spectroscopy, which is interface-specific spectroscopy and highly sensitive to molecular symmetry based on a second-order nonlinear optical process. It was revealed that PLA2 adsorbs to the DPPC monolayer on the aqueous solution surface only when the surface coverage is low. The adsorption at the low surface coverage significantly changes the interfacial structures of PLA2 and the hydration, which are stabilized by the presence of Ca2+. Therefore, the restriction of the hydrolysis of phospholipid monolayers at high surface coverage can be rationalized by the inhabitation of the PLA2 adsorption. The present study deepens our molecular-level understanding of the hydrolysis of phospholipids by PLA2.
Collapse
Affiliation(s)
- Ken-Ichi Inoue
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
| | - Takashi Yamamoto
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
| | - Yosuke Hatori
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
| | - Takeru Hiraide
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
| | - Shen Ye
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
| |
Collapse
|
3
|
Wei Y, Lv J, Zhu S, Wang S, Su J, Xu C. Enzyme-responsive liposomes for controlled drug release. Drug Discov Today 2024; 29:104014. [PMID: 38705509 DOI: 10.1016/j.drudis.2024.104014] [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: 12/16/2023] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
Compared to other nanovectors, liposomes exhibit unique advantages, such as good biosafety and high drug-loading capacity. However, slow drug release from conventional liposomes makes most payloads unavailable, restricting the therapeutic efficacy. Therefore, in the last ∼20 years, enzyme-responsive liposomes have been extensively investigated, which liberate drugs under the stimulation of enzymes overexpressed at disease sites. In this review, we elaborate on the research progress on enzyme-responsive liposomes. The involved enzymes mainly include phospholipases, particularly phospholipase A2, matrix metalloproteinases, cathepsins, and esterases. These enzymes can cleave ester bonds or specific peptide sequences incorporated in the liposomes for controlled drug release by disrupting the primary structure of liposomes, detaching protective polyethylene glycol shells, or activating liposome-associated prodrugs. Despite decades of efforts, there are still a lack marketed products of enzyme-responsive liposomes. Therefore, more efforts should be made to improve the safety and effectiveness of enzyme-responsive liposomes and address the issues associated with production scale-up.
Collapse
Affiliation(s)
- Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Shiyu Zhu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Shanghai 200433, China.
| |
Collapse
|
4
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
5
|
Koroleva M. Multicompartment colloid systems with lipid and polymer membranes for biomedical applications. Phys Chem Chem Phys 2023; 25:21836-21859. [PMID: 37565484 DOI: 10.1039/d3cp01984e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Multicompartment structures have the potential for biomedical applications because they can act as multifunctional systems and provide simultaneous delivery of drugs and diagnostics agents of different types. Moreover, some of them mimic biological cells to some extent with organelles as separate sub-compartments. This article analyses multicompartment colloidal structures with smaller sub-units covered with lipid or polymer membranes that provide additional protection for the encapsulated substances. Vesosomes with small vesicles encapsulated in the inner pools of larger liposomes are the most studied systems to date. Dendrimer molecules are enclosed by a lipid bilayer shell in dendrosomes. Capsosomes, polymersomes-in-polymer capsules, and cubosomes-in-polymer capsules are composed of sub-compartments encapsulated within closed multilayer polymer membranes. Janus or Cerberus emulsions contain droplets composed of two or three phases: immiscible oils in O/W emulsions and aqueous polymer or salt solutions that are separated into two or three phases and form connected droplets in W/O emulsions. In more cases, the external surface of engulfed droplets in Janus or Cerberus emulsions is covered with a lipid or polymer monolayer. eLiposomes with emulsion droplets encapsulated into a bilayer shell have been given little attention so far, but they have very great prospects. In addition to nanoemulsion droplets, solid lipid nanoparticles, nanostructured lipid carriers and inorganic nanoparticles can be loaded into eLiposomes. Molecular engineering of the external membrane allows the creation of ligand-targeted and stimuli-responsive multifunctional systems. As a result, the efficacy of drug delivery can be significantly enhanced.
Collapse
Affiliation(s)
- Marina Koroleva
- Mendeleev University of Chemical Technology, Miusskaya sq. 9, Moscow 125047.
| |
Collapse
|
6
|
Zhou W, Jia Y, Liu Y, Chen Y, Zhao P. Tumor Microenvironment-Based Stimuli-Responsive Nanoparticles for Controlled Release of Drugs in Cancer Therapy. Pharmaceutics 2022; 14:2346. [PMID: 36365164 PMCID: PMC9694300 DOI: 10.3390/pharmaceutics14112346] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/22/2022] [Accepted: 10/28/2022] [Indexed: 07/22/2023] Open
Abstract
With the development of nanomedicine technology, stimuli-responsive nanocarriers play an increasingly important role in antitumor therapy. Compared with the normal physiological environment, the tumor microenvironment (TME) possesses several unique properties, including acidity, high glutathione (GSH) concentration, hypoxia, over-expressed enzymes and excessive reactive oxygen species (ROS), which are closely related to the occurrence and development of tumors. However, on the other hand, these properties could also be harnessed for smart drug delivery systems to release drugs specifically in tumor tissues. Stimuli-responsive nanoparticles (srNPs) can maintain stability at physiological conditions, while they could be triggered rapidly to release drugs by specific stimuli to prolong blood circulation and enhance cancer cellular uptake, thus achieving excellent therapeutic performance and improved biosafety. This review focuses on the design of srNPs based on several stimuli in the TME for the delivery of antitumor drugs. In addition, the challenges and prospects for the development of srNPs are discussed, which can possibly inspire researchers to develop srNPs for clinical applications in the future.
Collapse
Affiliation(s)
- Weixin Zhou
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yujie Jia
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200065, China
| | - Yani Liu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Chen
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pengxuan Zhao
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
7
|
Zou D, Yang P, Liu J, Dai F, Xiao Y, Zhao A, Huang N. Exosome-Loaded Pro-efferocytic Vascular Stent with Lp-PLA 2-Triggered Release for Preventing In-Stent Restenosis. ACS NANO 2022; 16:14925-14941. [PMID: 36066255 DOI: 10.1021/acsnano.2c05847] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The efferocytosis defect is regarded as a pivotal event of atherosclerosis. The failure to clear apoptotic cells in atherosclerotic plaques under vascular stents causes a failure to resolve the inflammation underneath. However, efferocytosis repair is still confined to nonstenting therapeutics. Here, we identified a pro-efferocytotic agent and accordingly developed a bioresponsive pro-efferocytotic vascular stent aimed for poststenting healing. Exosomes derived from mesenchymal stem cells were found to be able to regulate efferocytosis via SLC2a1, STAT3/RAC1, and CD300a pathways and modulate foam cell formation processes through a CD36-mediated pathway. Pro-efferocytotic exosomes were encapsulated into liposome-based multivesicular chambers and grafted onto vascular stents. The multivesicular vesicles were able to release exosomes under the Lp-PLA2 environment. Compared to bare metal stents, exosome-stents in the presence of Lp-PLA2 enhanced the ratio of apoptotic cell clearance and reduced the neointimal thickness in the mal-efferocytotic rat model. Overall, we identified a pro-efferocytic agent─exosomes that are able to regulate target cells via multiple signaling pathways and are good candidates to serve complex pathological environments, and this bioresponsive pro-efferocytotic vascular stent is an attractive approach for prevention of poststenting complications.
Collapse
Affiliation(s)
- Dan Zou
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Ping Yang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Jianan Liu
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Fanfan Dai
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Yangyang Xiao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Ansha Zhao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Nan Huang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| |
Collapse
|
8
|
Salata GC, Malagó ID, Lopes LB. A Lipid-Based In Situ-Forming Hexagonal Phase for Prolonged Retention and Drug Release in the Breast Tissue. AAPS PharmSciTech 2022; 23:260. [PMID: 36123553 DOI: 10.1208/s12249-022-02411-9] [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: 06/13/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022] Open
Abstract
In this study, the addition of monoolein to phosphatidylcholine (PC), tricaprylin, and propylene glycol (PG) mixtures was studied to produce fluid precursor formulations (FIPs) that could transform into hexagonal phase (resistant to aqueous dilution) in vitro and in vivo. The overall goal was to obtain FIPs that could incorporate chemopreventive drugs for subcutaneous administration in the mammary tissue to inhibit the development and/or recurrence of breast cancer. Increasing PG content reduced FIP viscosity up to ~ 2.5-fold, while increases in PC (over monoolein) increased the formation of emulsified systems. The hexagonal phase was observed at 20% of water and higher, with the minimum amount of water necessary for this formation increasing with PG content. The selected FIP formed a depot in vivo after ~ 24 h of administration; its structure was compatible with the hexagonal phase and it remained in the mammary tissue for at least 30 days, prolonging the permanence of a fluorescent probe. In vitro, the release of the synthetic retinoid fenretinide was slow, with ~ 9% of the drug released in 72 h. Consistent with this slow release, fenretinide IC50 in breast cancer cells was ~ 100-fold higher in the selected FIP compared to its solution. The FIP reduced cell migration and presented higher cytotoxicity towards tumor compared to non-tumor cells. Given the limited number of options for pharmacological prevention of breast cancer development and recurrences, this formulation could potentially find applicability to reduce the frequency of administration and improve local concentrations of chemopreventive drugs.
Collapse
Affiliation(s)
- Giovanna C Salata
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, 1524 Av. Prof. Lineu Prestes, São Paulo-SP, 05508-000, Brazil
| | - Isabella D Malagó
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, 1524 Av. Prof. Lineu Prestes, São Paulo-SP, 05508-000, Brazil
| | - Luciana B Lopes
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, 1524 Av. Prof. Lineu Prestes, São Paulo-SP, 05508-000, Brazil.
| |
Collapse
|
9
|
Dutta G, Manickam S, Sugumaran A. Stimuli-Responsive Hybrid Metal Nanocomposite - A Promising Technology for Effective Anticancer Therapy. Int J Pharm 2022; 624:121966. [PMID: 35764265 DOI: 10.1016/j.ijpharm.2022.121966] [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: 05/04/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022]
Abstract
Cancer is one of the most challenging, life-threatening illnesses to cure, with over 10 million new cases diagnosed each year globally. Improved diagnostic cum treatment with common side-effects are warranting for successful therapy. Nanomaterials are recognized to improve early diagnosis, imaging, and treatment. Recently, multifunctional nanocomposites attracted considerable interest due to their low-cost production, and ideal thermal and chemical stability, and will be beneficial in future diagnostics and customized treatment capacity. Stimuli-Responsive Hybrid Metal Nanocomposites (SRHMNs) based nanocomposite materials pose the on/off delivery of bioactive compounds such as medications, genes, RNA, and DNA to specific tissue or organs and reduce toxicity. They simultaneously serve as sophisticated imaging and diagnostic tools when certain stimuli (e.g., temperature, pH, redox, ultrasound, or enzymes) activate the nanocomposite, resulting in the imaging-guided transport of the payload at defined sites. This review in detail addresses the recent advancements in the design and mechanism of internal breakdown processes of the functional moiety from stimuli-responsive systems in response to a range of stimuli coupled with metal nanoparticles. Also, it provides a thorough understanding of SRHMNs, enabling non-invasive interventional therapy by resolving several difficulties in cancer theranostics.
Collapse
Affiliation(s)
- Gouranga Dutta
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Jalan Tungku Link Gadong, BE1410, Brunei Darussalam
| | - Abimanyu Sugumaran
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India.
| |
Collapse
|
10
|
Shi D, Feng C, Xie J, Zhang X, Dai H, Yan L. Recent Progress of Nanomedicine on Secreted Phospholipase A2 as a Potential Therapeutic Target. J Mater Chem B 2022; 10:7349-7360. [DOI: 10.1039/d2tb00608a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Overexpressed secretory phospholipase A2 (sPLA2) is found in many inflammatory diseases and various types of cancer. sPLA2 can catalyze the hydrolysis of phospholipid sn-2 ester bond to lysophosphatidylcholine and free...
Collapse
|
11
|
Salata GC, Malagó ID, Carvalho Dartora VFM, Marçal Pessoa AF, Fantini MCDA, Costa SKP, Machado-Neto JA, Lopes LB. Microemulsion for Prolonged Release of Fenretinide in the Mammary Tissue and Prevention of Breast Cancer Development. Mol Pharm 2021; 18:3401-3417. [PMID: 34482696 DOI: 10.1021/acs.molpharmaceut.1c00319] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The need of pharmacological strategies to preclude breast cancer development motivated us to develop a non-aqueous microemulsion (ME) capable of forming a depot after administration in the mammary tissue and uptake of interstitial fluids for prolonged release of the retinoid fenretinide. The selected ME was composed of phosphatidylcholine/tricaprylin/propylene glycol (45:5:50, w/w/w) and presented a droplet diameter of 175.3 ± 8.9 nm. Upon water uptake, the ME transformed successively into a lamellar phase, gel, and a lamellar phase-containing emulsion in vitro as the water content increased and released 30% of fenretinide in vitro after 9 days. Consistent with the slow release, the ME formed a depot in cell cultures and increased fenretinide IC50 values by 68.3- and 13.2-fold in MCF-7 and T-47D cells compared to a solution, respectively. At non-cytotoxic concentrations, the ME reduced T-47D cell migration by 75.9% and spheroid growth, resulting in ∼30% smaller structures. The depot formed in vivo prolonged a fluorochrome release for 30 days without producing any sings of local irritation. In a preclinical model of chemically induced carcinogenesis, ME administration every 3 weeks for 3 months significantly reduced (4.7-fold) the incidence of breast tumors and increased type II collagen expression, which might contribute to limit spreading. These promising results support the potential ME applicability as a preventive therapy of breast cancer.
Collapse
Affiliation(s)
- Giovanna Cassone Salata
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, São Paulo 05508-000, Brazil
| | - Isabella D Malagó
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, São Paulo 05508-000, Brazil
| | - Vanessa F M Carvalho Dartora
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, São Paulo 05508-000, Brazil
| | - Ana Flávia Marçal Pessoa
- Departamento de Cirurgia, LIM26, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo, 455, São Paulo, São Paulo 01246903, Brazil
| | - Márcia Carvalho de Abreu Fantini
- Departamento de Física Aplicada, Instituto de Física, Universidade de São Paulo, Rua do Matão, 1371, São Paulo, São Paulo 05508-090, Brazil
| | - Soraia K P Costa
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, São Paulo 05508-000, Brazil
| | - João Agostinho Machado-Neto
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, São Paulo 05508-000, Brazil
| | - Luciana B Lopes
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
12
|
Wang G, Zannikou M, Lofchy L, Li Y, Gaikwad H, Balyasnikova IV, Simberg D. Liposomal Extravasation and Accumulation in Tumors as Studied by Fluorescence Microscopy and Imaging Depend on the Fluorescent Label. ACS NANO 2021; 15:11880-11890. [PMID: 34197075 PMCID: PMC8789216 DOI: 10.1021/acsnano.1c02982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Tumor trafficking of liposomes is routinely monitored via fluorescence microscopy and imaging. To investigate whether an accumulation of liposomes depends on the type of fluorescent label, we prepared PEGylated liposomes dual-labeled with indocarbocyanine lipids (ICLs: DiD or DiI) and fluorescent phospholipids (FPLs: Cy3-DSPE or Cy5-DSPE) with similar cyanine headgroups but different spectra. Using ex vivo confocal microscopy and imaging, we compared tumor extravasation and accumulation of ICLs and FPLs. After systemic injection in a syngeneic mouse model of 4T1 breast cancer, ICLs and FPLs initially colocalized in tumor blood vessels and perivascular space. At later time points, ICLs spread over a significantly larger tumor area and accumulated in tumor macrophages, whereas FPLs were mostly restricted to the vasculature with limited extravascular signal. This phenomenon was independent of liposomal composition and ICL/FPL type and was also observed in syngeneic intracranial GL261 glioma and LY2 head and neck cancer models. The dual-labeled liposomes were stable in plasma and delivered both dyes to tumors at early time points. Notably, while the level of ICLs increased over time, FPLs gradually disappeared from tumors and other organs in vivo, likely due to degradation of the phospholipid. These findings demonstrate that trafficking and stability of the label is of critical importance when assessing extravasation and accumulation of nanocarriers in tumors and other organs by fluorescence microscopy and imaging.
Collapse
Affiliation(s)
| | - Markella Zannikou
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | | | | | | | - Irina V Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | | |
Collapse
|
13
|
Mollazadeh S, Mackiewicz M, Yazdimamaghani M. Recent advances in the redox-responsive drug delivery nanoplatforms: A chemical structure and physical property perspective. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111536. [PMID: 33255089 DOI: 10.1016/j.msec.2020.111536] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Poor water solubility, off-target toxicity, and small therapeutic window are among major obstacles for the development of drug products. Redox-responsive drug delivery nanoplatforms not only overcome the delivery and pharmacokinetic pitfalls observed in conventional drug delivery, but also leverage the site-specific delivery properties. Cleavable diselenide and disulfide bonds in the presence of elevated reactive oxygen species (ROS) and glutathione concentration are among widely used stimuli-responsive bonds to design nanocarriers. This review covers a wide range of redox-responsive chemical structures and their properties for designing nanoparticles aiming controlled loading, delivery, and release of hydrophobic anticancer drugs at tumor site.
Collapse
Affiliation(s)
- Shirin Mollazadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marcin Mackiewicz
- Faculty of Chemistry, Biological and Chemical Research Center, University of Warsaw, Poland
| | - Mostafa Yazdimamaghani
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Németh Z, Pallagi E, Dobó DG, Csóka I. A Proposed Methodology for a Risk Assessment-Based Liposome Development Process. Pharmaceutics 2020; 12:E1164. [PMID: 33260443 PMCID: PMC7760874 DOI: 10.3390/pharmaceutics12121164] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 12/18/2022] Open
Abstract
The requirements of a liposomal formulation vary depending on the pharmaceutical indication, the target patient population, and the corresponding route of administration. Different preparation methods require various material attributes (MAs) (properties and characteristics of the components) and process parameters (PPs) (settings of the preparation method). The identification of the quality target product profile for a liposome-based formulation, the critical quality attributes of the liposomes, and the possible MAs and PPs that may influence the key characteristics of the vesicles facilitates pharmaceutical research. Researchers can systematise their knowledge by using the quality by design (QbD) approach. The potential factors that influence the quality of the product can be collected and studied through a risk assessment process. In this paper, the requirements of a liposome formulation prepared via the thin-film hydration preparation technique are presented; furthermore, the possible factors that have an impact on the quality of the final product and have to be considered and specified during the development of a liposomal formulation are herein identified and collected. The understanding and the application of these elements of QbD in the pharmaceutical developments help to influence the quality, the achievements, and the success of the formulated product.
Collapse
Affiliation(s)
| | | | | | - Ildikó Csóka
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös u. 6., H-6720 Szeged, Hungary; (Z.N.); (E.P.); or (D.G.D.)
| |
Collapse
|
15
|
Li D, An X, Mu Y. A liposomal hydrogel with enzyme triggered release for infected wound. Chem Phys Lipids 2019; 223:104783. [DOI: 10.1016/j.chemphyslip.2019.104783] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/19/2019] [Accepted: 06/08/2019] [Indexed: 12/11/2022]
|
16
|
Zhang P, Villanueva V, Kalkowski J, Liu C, Donovan AJ, Bu W, Schlossman ML, Lin B, Liu Y. Molecular interactions of phospholipid monolayers with a model phospholipase. SOFT MATTER 2019; 15:4068-4077. [PMID: 30958491 DOI: 10.1039/c8sm01154k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The intrinsic overexpression of secretory phospholipase A2 (sPLA2) in various pro-inflammatory diseases and cancers has the potential to be exploited as a therapeutic strategy for diagnostics and treatment. To explore this potential and advance our knowledge of the role of sPLA2 in related diseases, it is necessary to systematically investigate the molecular interaction of the enzyme with lipids. By employing a Langmuir trough integrated with X-ray reflectivity and grazing incidence X-ray diffraction techniques, this study examined the molecular packing structure of 1,2-palmitoyl-sn-glycero-3-phosphocholine (DPPC) films before and after enzyme adsorption and enzyme-catalyzed degradation. Molecular interaction of sPLA2 (from bee venom) with the DPPC monolayer exhibited Ca2+ dependence. DPPC molecules at the interface without Ca2+ retained a monolayer organization; upon adsorption of sPLA2 to the monolayer the packing became tighter. In contrast, sPLA2-catalyzed degradation of DPPC occurred in the presence of Ca2+, leading to disruption of the ordered monolayer structure of DPPC. The interfacial film became a mixture of highly ordered multilayer domains of palmitic acid (PA) and loosely packed monolayer phase of 1-palmitoyl-2-hydroxy-sn-glycero-3-phosphocholine (lysoPC) that potentially contained the remaining un-degraded DPPC. The redistribution of lipid degradation products into the third dimension, which produced multilayer PA domains, damaged the structural integrity of the original lipid layer and may explain the bursting of liposomes observed in other studies after a latency period of mixing liposomes with sPLA2. A quantitative understanding of the lipid packing and lipid-enzyme interaction provides an intuitive means of designing and optimizing lipid-related drug delivery systems.
Collapse
Affiliation(s)
- Pin Zhang
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Li Y, Du L, Wu C, Yu B, Zhang H, An F. Peptide Sequence-Dominated Enzyme-Responsive Nanoplatform for Anticancer Drug Delivery. Curr Top Med Chem 2019; 19:74-97. [PMID: 30686257 DOI: 10.2174/1568026619666190125144621] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/06/2018] [Accepted: 11/23/2018] [Indexed: 02/08/2023]
Abstract
Enzymatic dysregulation in tumor and intracellular microenvironments has made this property
a tremendously promising responsive element for efficient diagnostics, carrier targeting, and drug
release. When combined with nanotechnology, enzyme-responsive drug delivery systems (DDSs) have
achieved substantial advancements. In the first part of this tutorial review, changes in tumor and intracellular
microenvironmental factors, particularly the enzymatic index, are described. Subsequently, the
peptide sequences of various enzyme-triggered nanomaterials are summarized for their uses in various
drug delivery applications. Then, some other enzyme responsive nanostructures are discussed. Finally,
the future opportunities and challenges are discussed. In brief, this review can provide inspiration and
impetus for exploiting more promising internal enzyme stimuli-responsive nanoDDSs for targeted tumor
diagnosis and treatment.
Collapse
Affiliation(s)
- Yanan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Liping Du
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Chunsheng Wu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hui Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
18
|
Ahmed KS, Hussein SA, Ali AH, Korma SA, Lipeng Q, Jinghua C. Liposome: composition, characterisation, preparation, and recent innovation in clinical applications. J Drug Target 2018; 27:742-761. [PMID: 30239255 DOI: 10.1080/1061186x.2018.1527337] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the last decades, pharmaceutical interested researches aimed to develop novel and innovative drug delivery techniques in the medical and pharmaceutical fields. Recently, phospholipid vesicles (Liposomes) are the most known versatile assemblies in the drug delivery systems. The discovery of liposomes arises from self-forming enclosed phospholipid bilayer upon coming in contact with the aqueous solution. Liposomes are uni or multilamellar vesicles consisting of phospholipids produced naturally or synthetically, which are readily non-toxic, biodegradable, and are readily produced on a large scale. Various phospholipids, for instance, soybean, egg yolk, synthetic, and hydrogenated phosphatidylcholine consider the most popular types used in different kinds of formulations. This review summarises liposomes composition, characterisation, methods of preparation, and their applications in different medical fields including cancer therapy, vaccine, ocular delivery, wound healing, and some dermatological applications.
Collapse
Affiliation(s)
- Kamel S Ahmed
- a Department of Pharmaceutics , School of Pharmaceutical Sciences, Jiangnan University , Wuxi , PR China.,b Department of Pharmaceutics , Faculty of Pharmacy, Minia University , Minia , Egypt
| | - Saied A Hussein
- c Department of Biomedical Engineering , College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan , PR China
| | - Abdelmoneim H Ali
- d State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, School of Food Science and Technology, Jiangnan University , Wuxi , PR China
| | - Sameh A Korma
- d State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, School of Food Science and Technology, Jiangnan University , Wuxi , PR China
| | - Qiu Lipeng
- a Department of Pharmaceutics , School of Pharmaceutical Sciences, Jiangnan University , Wuxi , PR China
| | - Chen Jinghua
- a Department of Pharmaceutics , School of Pharmaceutical Sciences, Jiangnan University , Wuxi , PR China
| |
Collapse
|
19
|
Dahan A, Markovic M, Keinan S, Kurnikov I, Aponick A, Zimmermann EM, Ben-Shabat S. Computational modeling and in-vitro/in-silico correlation of phospholipid-based prodrugs for targeted drug delivery in inflammatory bowel disease. J Comput Aided Mol Des 2017; 31:1021-1028. [PMID: 29101519 DOI: 10.1007/s10822-017-0079-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/25/2017] [Indexed: 12/31/2022]
Abstract
Targeting drugs to the inflamed intestinal tissue(s) represents a major advancement in the treatment of inflammatory bowel disease (IBD). In this work we present a powerful in-silico modeling approach to guide the molecular design of novel prodrugs targeting the enzyme PLA2, which is overexpressed in the inflamed tissues of IBD patients. The prodrug consists of the drug moiety bound to the sn-2 position of phospholipid (PL) through a carbonic linker, aiming to allow PLA2 to release the free drug. The linker length dictates the affinity of the PL-drug conjugate to PLA2, and the optimal linker will enable maximal PLA2-mediated activation. Thermodynamic integration and Weighted Histogram Analysis Method (WHAM)/Umbrella Sampling method were used to compute the changes in PLA2 transition state binding free energy of the prodrug molecule (∆∆Gtr) associated with decreasing/increasing linker length. The simulations revealed that 6-carbons linker is the optimal one, whereas shorter or longer linkers resulted in decreased PLA2-mediated activation. These in-silico results were shown to be in excellent correlation with experimental in-vitro data. Overall, this modern computational approach enables optimization of the molecular design of novel prodrugs, which may allow targeting the free drug specifically to the diseased intestinal tissue of IBD patients.
Collapse
Affiliation(s)
- Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel.
| | - Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| | - Shahar Keinan
- Cloud Pharmaceuticals Inc., 6 Davis Dr., Research Triangle Park, NC, 27709, USA
| | - Igor Kurnikov
- Cloud Pharmaceuticals Inc., 6 Davis Dr., Research Triangle Park, NC, 27709, USA
| | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL, 32608, USA
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL, 32608, USA
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| |
Collapse
|
20
|
Kono Y, Jinzai H, Kotera Y, Fujita T. Influence of Physicochemical Properties and PEG Modification of Magnetic Liposomes on Their Interaction with Intestinal Epithelial Caco-2 Cells. Biol Pharm Bull 2017; 40:2166-2174. [PMID: 28966298 DOI: 10.1248/bpb.b17-00563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study aimed to investigate the effect of particle size (100, 500 nm), surface charge (cationic, neutral and anionic) and polyethylene glycol (PEG) modification of magnetic liposomes on their interaction with the human intestinal epithelial cell line, Caco-2. The cellular associated amount of all the magnetic liposomes was significantly increased by the presence of a magnetic field. The highest association and internalization into Caco-2 cells was observed with magnetic cationic liposomes. Moreover, small magnetic liposomes were more efficiently associated and taken up into the cells, than large ones. In contrast, PEG modification significantly attenuated the enhancing effect of the magnetic field on the cellular association of magnetic liposomes. We also found that magnetic cationic liposomes had the highest retention properties to Caco-2 cells. Moreover, the retention of large magnetic liposomes to the cells was much longer than that of small ones. In addition, magnetic cationic and neutral liposomes had relatively high stability in Caco-2 cells, whereas magnetic anionic liposomes rapidly degraded. These results indicate that the physicochemical properties and PEG modification of magnetic liposomes greatly influences their intestinal epithelial transport.
Collapse
Affiliation(s)
- Yusuke Kono
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University.,Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University
| | - Hitomi Jinzai
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Yota Kotera
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Takuya Fujita
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University.,Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University.,Research Center for Drug Discovery and Development, Ritsumeikan University
| |
Collapse
|
21
|
Lee Y, Thompson DH. Stimuli-responsive liposomes for drug delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1450. [PMID: 28198148 PMCID: PMC5557698 DOI: 10.1002/wnan.1450] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/25/2022]
Abstract
The ultimate goal of drug delivery is to increase the bioavailability and reduce the toxic side effects of the active pharmaceutical ingredient (API) by releasing them at a specific site of action. In the case of antitumor therapy, association of the therapeutic agent with a carrier system can minimize damage to healthy, nontarget tissues, while limit systemic release and promoting long circulation to enhance uptake at the cancerous site due to the enhanced permeation and retention effect (EPR). Stimuli-responsive systems have become a promising way to deliver and release payloads in a site-selective manner. Potential carrier systems have been derived from a wide variety of materials, including inorganic nanoparticles, lipids, and polymers that have been imbued with stimuli-sensitive properties to accomplish triggered release based on an environmental cue. The unique features in the tumor microenvironment can serve as an endogenous stimulus (pH, redox potential, or unique enzymatic activity) or the locus of an applied external stimulus (heat or light) to trigger the controlled release of API. In liposomal carrier systems triggered release is generally based on the principle of membrane destabilization from local defects within bilayer membranes to effect release of liposome-entrapped drugs. This review focuses on the literature appearing between November 2008-February 2016 that reports new developments in stimuli-sensitive liposomal drug delivery strategies using pH change, enzyme transformation, redox reactions, and photochemical mechanisms of activation. WIREs Nanomed Nanobiotechnol 2017, 9:e1450. doi: 10.1002/wnan.1450 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Y Lee
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - D H Thompson
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
22
|
Sybachin AV, Zaborova OV, Efimova AA, Ballauff M, Yaroslavov AA. Electrostatic complexes of liquid and solid liposomes with spherical polycationic brushes. POLYMER SCIENCE SERIES C 2017. [DOI: 10.1134/s181123821701012x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
23
|
Arouri A, Lauritsen KE, Nielsen HL, Mouritsen OG. Effect of fatty acids on the permeability barrier of model and biological membranes. Chem Phys Lipids 2016; 200:139-146. [PMID: 27725161 DOI: 10.1016/j.chemphyslip.2016.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/26/2016] [Accepted: 10/04/2016] [Indexed: 10/20/2022]
Abstract
Because of the amphipathicity and conical molecular shape of fatty acids, they can efficiently incorporate into lipid membranes and disturb membrane integrity, chain packing, and lateral pressure profile. These phenomena affect both model membranes as well as biological membranes. We investigated the feasibility of exploiting fatty acids as permeability enhancers in drug delivery systems for enhancing drug release from liposomal carriers and drug uptake by target cells. Saturated fatty acids, with acyl chain length from C8 to C20, were tested using model drug delivery liposomes of 1,2- dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and the breast cancer MCF-7 cell line as a model cell. A calcein release assay demonstrated reduction in the membrane permeability barrier of the DPPC liposomes, proportionally to the length of the fatty acid. Differential scanning calorimetry (DSC) and dynamic light scattering (DLS) experiments revealed that C12 to C20 fatty acids can stabilize DPPC liposomal bilayers and induce the formation of large structures, probably due to liposome aggregation and bilayer morphological changes. On the other hand, the short fatty acids C8 and C10 tend to destabilize the bilayers and only moderately cause the formation of large structures. The effect of fatty acids on DPPC liposomes was not completely transferrable to the MCF-7 cell line. Using cytotoxicity assays, the cells were found to be relatively insensitive to the fatty acids at apoptotic sub-millimolar concentrations. Increasing the fatty acid concentration to few millimolar substantially reduced the viability of the cells, most likely via the induction of necrosis and cell lysis. A bioluminescence living-cell-based luciferase assay showed that saturated fatty acids in sub-cytotoxic concentrations cannot reduce the permeability barrier of cell membranes. Our results confirm that the membrane perturbing effect of fatty acids on model membranes cannot simply be carried over to biological membranes of live cells.
Collapse
Affiliation(s)
- Ahmad Arouri
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark; The Lundbeck Foundation Nanomedicine Research Center for Cancer Stem Cell Targeting Therapeutics (NanoCAN), University of Southern Denmark, Odense, Denmark.
| | - Kira E Lauritsen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Henriette L Nielsen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Ole G Mouritsen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark; The Lundbeck Foundation Nanomedicine Research Center for Cancer Stem Cell Targeting Therapeutics (NanoCAN), University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
24
|
Zhdanov VP, Agnarsson B, Höök F. Kinetics of enzyme-mediated hydrolysis of lipid vesicles. Chem Phys Lett 2016. [DOI: 10.1016/j.cplett.2016.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
25
|
Hong CY, Han CT, Chao L. Nonspecific Binding Domains in Lipid Membranes Induced by Phospholipase A2. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:6991-6999. [PMID: 27218880 DOI: 10.1021/acs.langmuir.5b03915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Phospholipase A2 (PLA2) is a peripheral membrane protein that can hydrolyze phospholipids to produce lysolipids and fatty acids. It has been found to play crucial roles in various cellular processes and is thought as a potential candidate for triggering drug release from liposomes for medical treatment. Here, we directly observed that PLA2 hydrolysis reaction can induce the formation of PLA2-binding domains at lipid bilayer interface and found that the formation was significantly influenced by the fluidity of the lipid bilayer. We prepared supported lipid bilayers (SLBs) with various molar ratios of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) to adjust the reactivity and fluidity of the lipid bilayers. A significant amount of the PLA2-induced domains was observed in mixtures of DPPC and DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine) but not in either pure DPPC or pure DOPC bilayer, which might be the reason that previous studies rarely observed these domains in lipid bilayer systems. The fluorescently labeled PLA2 experiment showed that newly formed domains acted as binding templates for PLA2. The AFM result showed that the induced domain has stepwise plateau structure, suggesting that PLA2 hydrolysis products may align as bilayers and accumulate layer by layer on the support, and the hydrophobic acyl chains at the side of the layer structure may be exposed to the outside aqueous environment. The introduced hydrophobic region could have hydrophobic interactions with proteins and therefore can attract the binding of not only PLA2 but also other types of proteins such as proteoglycans and streptavidin. The results suggest that the formation of PLA2-induced domains may convert part of a zwitterionic nonsticky lipid membrane to a site where biomolecules can nonspecifically bind.
Collapse
Affiliation(s)
- Chia Yee Hong
- Department of Chemical Engineering, National Taiwan University , Taipei, Taiwan 10617
| | - Chung-Ta Han
- Department of Chemical Engineering, National Taiwan University , Taipei, Taiwan 10617
| | - Ling Chao
- Department of Chemical Engineering, National Taiwan University , Taipei, Taiwan 10617
| |
Collapse
|
26
|
Hansen AH, Lomholt MA, Hansen PL, Mouritsen OG, Arouri A. Optimization and modeling of the remote loading of luciferin into liposomes. Int J Pharm 2016; 508:128-34. [PMID: 27163524 DOI: 10.1016/j.ijpharm.2016.04.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/15/2016] [Accepted: 04/19/2016] [Indexed: 12/29/2022]
Abstract
We carried out a mechanistic study to characterize and optimize the remote loading of luciferin into preformed liposomes of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine/1,2-dipalmitoyl-sn-glycero-3-phosphoglycerol (DPPC/DPPG) 7:3 mixtures. The influence of the loading agent (acetate, propionate, butyrate), the metal counterion (Na(+), K(+), Ca(+2), Mg(+2)), and the initial extra-liposomal amount of luciferin (nL(add)) on the luciferin Loading Efficiency (LE%) and luciferin-to-lipid weight ratio, i.e., Loading Capacity (LC), in the final formulation was determined. In addition, the effect of the loading process on the colloidal stability and phase behavior of the liposomes was monitored. Based on our experimental results, a theoretical model was developed to describe the course of luciferin remote loading. It was found that the highest luciferin loading was obtained with magnesium acetate. The use of longer aliphatic carboxylates or inorganic proton donors pronouncedly reduced luciferin loading, whereas the effect of the counterion was modest. The remote-loading process barely affected the colloidal stability and drug retention of the liposomes, albeit with moderate luciferin-induced membrane perturbations. The correlation between luciferin loading, expressed as LE% and LC, and nL(add) was established, and under our conditions the maximum LC was attained using an nL(add) of around 2.6μmol. Higher amounts of luciferin tend to pronouncedly perturb the liposome stability and luciferin retention. Our theoretical model furnishes a fair quantitative description of the correlation between nL(add) and luciferin loading, and a membrane permeability coefficient for uncharged luciferin of 1×10(-8)cm/s could be determined. We believe that our study will prove very useful to optimize the remote-loading strategies of moderately polar carboxylic acid drugs in general.
Collapse
Affiliation(s)
- Anders Højgaard Hansen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark(1); The Lundbeck Foundation Nanomedicine Research Center for Cancer Stem Cell Targeting Therapeutics (NanoCAN), University of Southern Denmark, Odense, Denmark(2).
| | - Michael A Lomholt
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark(1).
| | - Per Lyngs Hansen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark(1).
| | - Ole G Mouritsen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark(1); The Lundbeck Foundation Nanomedicine Research Center for Cancer Stem Cell Targeting Therapeutics (NanoCAN), University of Southern Denmark, Odense, Denmark(2).
| | - Ahmad Arouri
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark(1); The Lundbeck Foundation Nanomedicine Research Center for Cancer Stem Cell Targeting Therapeutics (NanoCAN), University of Southern Denmark, Odense, Denmark(2).
| |
Collapse
|