1
|
Liu Y, Yao X, Wen C, Li D, Zhang J, Xi B, Cummings BS, Zhu G. Facial Amphiphile-Modified Lipids Highly Sensitize Liposomes toward Secretory Phospholipase A 2. Mol Pharm 2024; 21:5469-5481. [PMID: 39397289 DOI: 10.1021/acs.molpharmaceut.4c00271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Upregulated secretory phospholipase A2 (sPLA2) in tumors has been proposed as a stimulus to trigger drug release from liposomes for therapeutic effects. However, the current strategy for developing sPLA2-responsive liposomes merely considering substrate preference suffers from limited membrane disruptive effects induced by enzymatic hydrolysis and safety issues resulting from the overuse of sPLA2-preferred lipids. Here, a membrane-destabilizing mechanism based on enzymatic extraction and the transition of facial amphiphiles (FAs) within lipid membranes was introduced. Enzymatic degradation of FA-modified lipids, a process involving substrate extraction of lipids from membranes and cleavage of sn-2 ester bonds by sPLA2, rotation, and interface settling of detached FAs, caused tremendous efflux of payloads from liposomes, termed the SECRIS effect. In the presence of sPLA2, oxaliplatin (L-OHP) loaded liposomes containing FA-modified lipids showed enhanced drug release, comparable in vitro cytotoxicity, and excellent in vivo antitumor efficacy and reduced adverse syndromes in Colo205-bearing mice compared to conventional sPLA2-labile formulations. The discovery of the SECRIS effect creates a new pathway to engineer liposome platforms for the treatment of sPLA2-positive tumors.
Collapse
Affiliation(s)
- Yanjiao Liu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong 529020, People's Republic of China
| | - Xingang Yao
- School of Pharmacy, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Cheng Wen
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong 529020, People's Republic of China
| | - Dan Li
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong 529020, People's Republic of China
| | - Jiawen Zhang
- School of Pharmacy, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Baomin Xi
- School of Pharmacy, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Brian S Cummings
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan 48201, United States
| | - Guodong Zhu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong 529020, People's Republic of China
| |
Collapse
|
2
|
Liposome-Tethered Gold Nanoparticles Triggered by Pulsed NIR Light for Rapid Liposome Contents Release and Endosome Escape. Pharmaceutics 2022; 14:pharmaceutics14040701. [PMID: 35456535 PMCID: PMC9025641 DOI: 10.3390/pharmaceutics14040701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Remote triggering of contents release with micron spatial and sub-second temporal resolution has been a long-time goal of medical and technical applications of liposomes. Liposomes can sequester a variety of bioactive water-soluble ions, ligands and enzymes, and oligonucleotides. The bilayer that separates the liposome interior from the exterior solution provides a physical barrier to contents release and degradation. Tethering plasmon-resonant, hollow gold nanoshells to the liposomes, or growing gold nanoparticles directly on the liposome exterior, allows liposome contents to be released by nanosecond or shorter pulses of near-infrared light (NIR). Gold nanoshells or nanoparticles strongly adsorb NIR light; cells, tissues, and physiological media are transparent to NIR, allowing penetration depths of millimeters to centimeters. Nano to picosecond pulses of NIR light rapidly heat the gold nanoshells, inducing the formation of vapor nanobubbles, similar to cavitation bubbles. The collapse of the nanobubbles generates mechanical forces that rupture bilayer membranes to rapidly release liposome contents at the preferred location and time. Here, we review the syntheses, characterization, and applications of liposomes coupled to plasmon-resonant gold nanostructures for delivering a variety of biologically important contents in vitro and in vivo with sub-micron spatial control and sub-second temporal control.
Collapse
|
3
|
Frampton MB, Blais A, Raczywolski Z, Castle A, Zelisko PM. Exploring the utility of hybrid siloxane-phosphocholine (SiPC) liposomes as drug delivery vehicles. RSC Adv 2021. [DOI: 10.1039/d0ra10052h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hybrid siloxane-phosphocholines (SiPCs) are a unique class of lipids that spontaneously form unilamellar vesicles (ULVs) that are ∼100 nm in diameter upon exposure to aqueous media without the need for extrusion and can be used as delivery vehicles.
Collapse
Affiliation(s)
- Mark B. Frampton
- Department of Chemistry
- Centre for Biotechnology
- Brock University
- St. Catharines
- Canada
| | - Andrea Blais
- Department of Chemistry
- Centre for Biotechnology
- Brock University
- St. Catharines
- Canada
| | - Zachary Raczywolski
- Department of Chemistry
- Centre for Biotechnology
- Brock University
- St. Catharines
- Canada
| | - Alan Castle
- Department of Biological Sciences
- Centre for Biotechnology
- Brock University
- St. Catharines
- Canada
| | - Paul M. Zelisko
- Department of Chemistry
- Centre for Biotechnology
- Brock University
- St. Catharines
- Canada
| |
Collapse
|
4
|
Lou J, Best MD. Strategies for altering lipid self-assembly to trigger liposome cargo release. Chem Phys Lipids 2020; 232:104966. [PMID: 32888913 DOI: 10.1016/j.chemphyslip.2020.104966] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 01/21/2023]
Abstract
While liposomes have proven to be effective drug delivery nanocarriers, their therapeutic attributes could be improved through the development of clinically viable triggered release strategies in which encapsulated drug contents could be selectively released at the sites of diseased cells. As such, a significant amount of research has been reported involving the development of stimuli-responsive liposomes and a broad range of strategies have been explored for driving content release. These have included the introduction of trigger groups at either the lipid headgroup or within the acyl chains that alter lipid self-assembly properties of known lipids as well as the rational design of lipid analogs programed to undergo conformational changes induced by events such as binding interactions. This review article describes advances in the design of stimuli-responsive liposome strategies with an eye towards emerging trends in the field.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Dr, Knoxville, TN, 37996, USA
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Dr, Knoxville, TN, 37996, USA.
| |
Collapse
|
5
|
Zhang P, Pham T, Liu C, Leon Plata P, Kalkowski J, Cheng G, Bu W, Lin B, Liu Y. Impeded Molecular Reorganization by Polyethylene Glycol Conjugation Revealed by X-ray Reflectivity and Diffraction Measurements. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:7573-7581. [PMID: 32501715 DOI: 10.1021/acs.langmuir.0c01202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Polyethylene glycol (PEG) coatings have been widely applied in pharmaceutical and biomedical systems to prevent nonspecific protein absorption, increase vesicle blood circulation time, and sustain drug release. This study systematically investigated the planar interfacial organization of phospholipid monolayers containing various amounts of PEG conjugations before and after enzyme-catalyzed degradation of the lipids using X-ray reflectivity and grazing incidence X-ray diffraction techniques. Results showed that attaching PEG to the headgroup of the lipids up to 15 mol % had limited effects on molecular packing of the lipid monolayers in the condensed phase at the gas-liquid interface and negligible effects on the enzyme adsorption to the interface. After enzyme-catalyzed degradation, equimolar fatty acids and lyso PC were generated. The fatty acids together with the subphase Ca2+ self-assembled into highly organized multilayer domains at the interface. The X-ray measurements unambiguously revealed that the densely packed PEG markedly hindered microphase separation and formation of the palmitic acid-Ca2+ complexes.
Collapse
Affiliation(s)
- Pin Zhang
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Tiep Pham
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Chang Liu
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Paola Leon Plata
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Joseph Kalkowski
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Gang Cheng
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Wei Bu
- NSF's ChemMatCARS, University of Chicago, Chicago, Illinois 60637, United States
| | - Binhua Lin
- NSF's ChemMatCARS, University of Chicago, Chicago, Illinois 60637, United States
| | - Ying Liu
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
- Richard & Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
6
|
Hindley JW, Law RV, Ces O. Membrane functionalization in artificial cell engineering. SN APPLIED SCIENCES 2020. [DOI: 10.1007/s42452-020-2357-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AbstractBottom-up synthetic biology aims to construct mimics of cellular structure and behaviour known as artificial cells from a small number of molecular components. The development of this nascent field has coupled new insights in molecular biology with large translational potential for application in fields such as drug delivery and biosensing. Multiple approaches have been applied to create cell mimics, with many efforts focusing on phospholipid-based systems. This mini-review focuses on different approaches to incorporating molecular motifs as tools for lipid membrane functionalization in artificial cell construction. Such motifs range from synthetic chemical functional groups to components from extant biology that can be arranged in a ‘plug-and-play’ approach which is hard to replicate in living systems. Rationally designed artificial cells possess the promise of complex biomimetic behaviour from minimal, highly engineered chemical networks.
Collapse
|
7
|
Ghavami M, Shiraishi T, Nielsen PE. Enzyme-Triggered Release of the Antisense Octaarginine-PNA Conjugate from Phospholipase A2 Sensitive Liposomes. ACS APPLIED BIO MATERIALS 2020; 3:1018-1025. [DOI: 10.1021/acsabm.9b01022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Mahdi Ghavami
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Takehiko Shiraishi
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Peter E. Nielsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Wang J, Chai J, Liu L, Cui Z, Duan D, Shi R, Zhang Y. Dual-functional melanin-based nanoliposomes for combined chemotherapy and photothermal therapy of pancreatic cancer. RSC Adv 2019; 9:3012-3019. [PMID: 35518954 PMCID: PMC9059978 DOI: 10.1039/c8ra09420a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/26/2018] [Indexed: 11/24/2022] Open
Abstract
Pancreatic cancer, one of the most common gastrointestinal tract cancers, leads to a high mortality rate of over 80% among patients. Conventional chemotherapy with gemcitabine (GEM) is undesirable due to the lack of effective tumor accumulation. To improve the survival of pancreatic cancer patients and the therapeutic efficiency of chemotherapy, dual-functional melanin-based nanoliposomes loaded with GEM were synthesized in our study, which combined chemotherapy and photothermal therapy (PTT). Hypothermia caused by melanin under near-infrared (NIR) laser exerted detrimental damage on pancreatic cancer cells after the passive accumulation of nanoliposomes in the tumor sites. Besides, the temperature increase could enhance the release of GEM from the nanoliposomes by changing the structural integrity of the nanoliposomes. Therefore, a synergistic antitumor effect was achieved by loading the chemotherapy agent GEM and the photothermal agent melanin into the nanoliposomes. The findings in this study strongly support that melanin-based nanoliposomes could be a desirable strategy against pancreatic carcinoma. GEM-Mel-Lip converted light to heat based on melanin after entering the tumor cells, and then the phospholipid fluidity was increased under the hyperthermia generated, resulting in the release of GEM.![]()
Collapse
Affiliation(s)
- Jian Wang
- Hepatobiliary Surgery Department
- Tianjin First Center Hospital
- China
- Tianjin Clinical Research Center for Organ Transplantation
- China
| | - Jiasui Chai
- First Central Clinic of Tianjin Medical University
- China
| | - Lei Liu
- Tianjin Clinical Research Center for Organ Transplantation
- China
- Key Laboratory of Transplant Medicine
- Chinese Academy of Medical Sciences
- China
| | - Zilin Cui
- Hepatobiliary Surgery Department
- Tianjin First Center Hospital
- China
- Tianjin Clinical Research Center for Organ Transplantation
- China
| | - Dongming Duan
- Hepatobiliary Surgery Department
- Tianjin First Center Hospital
- China
- Tianjin Clinical Research Center for Organ Transplantation
- China
| | - Rui Shi
- Hepatobiliary Surgery Department
- Tianjin First Center Hospital
- China
- Tianjin Clinical Research Center for Organ Transplantation
- China
| | - Yamin Zhang
- Hepatobiliary Surgery Department
- Tianjin First Center Hospital
- China
- Tianjin Clinical Research Center for Organ Transplantation
- China
| |
Collapse
|
9
|
Behroozi F, Abdkhodaie MJ, Abandansari HS, Satarian L, Ashtiani MK, Jaafari MR, Baharvand H. Smart liposomal drug delivery for treatment of oxidative stress model in human embryonic stem cell-derived retinal pigment epithelial cells. Int J Pharm 2018; 548:62-72. [PMID: 29802900 DOI: 10.1016/j.ijpharm.2018.05.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 04/18/2018] [Accepted: 05/15/2018] [Indexed: 01/09/2023]
Abstract
Oxidative stress has been implicated in the progression of age-related macular degeneration (AMD). Treatment with antioxidants seems to delay progression of AMD. In this study, we suggested an antioxidant delivery system based on redox-sensitive liposome composed of phospholipids and a diselenide centered alkyl chain. Dynamic light scattering assessment indicated that the liposomes had an average size of 140 nm with a polydispersity index below 0.2. The percentage of encapsulation efficiency of the liposomes was calculated by high-performance liquid chromatography. The carriers were loaded with N-acetyl cysteine as a model antioxidant drug. We demonstrated responsiveness of the nanocarrier and its efficiency in drug delivery in an oxidative stress model of human embryonic stem cell-derived retinal pigment epithelial (hESC-RPE) cells. The modeled cells treated with diselenide containing liposomes loaded with 10 mM NAC, showed a better therapeutic effect with a cell metabolic activity of 90%, which was significantly higher compared to insensitive liposomes or NAC treated groups (P < 0.05). In addition, the expression of oxidative-sensitive gene markers in diselenide containing liposomes groups were improved. Our results demonstrated fabricated smart liposomes opens new opportunity for targeted treatment of retinal degeneration.
Collapse
Affiliation(s)
- Farnaz Behroozi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Hamid Sadeghi Abandansari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Leila Satarian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
10
|
Maji D, Lu J, Sarder P, Schmieder AH, Cui G, Yang X, Pan D, Lew MD, Achilefu S, Lanza GM. Cellular Trafficking of Sn-2 Phosphatidylcholine Prodrugs Studied with Fluorescence Lifetime Imaging and Super-resolution Microscopy. PRECISION NANOMEDICINE 2018; 1:128-145. [PMID: 31249994 PMCID: PMC6597004 DOI: 10.33218/prnano1(2).180724.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While the in vivo efficacy of Sn-2 phosphatidylcholine prodrugs incorporated into targeted, non-pegylated lipid-encapsulated nanoparticles was demonstrated in prior preclinical studies, the microscopic details of cell prodrug internalization and trafficking events are unknown. Classic fluorescence microscopy, fluorescence lifetime imaging microscopy, and single-molecule super-resolution microscopy were used to investigate the cellular handling of doxorubicin-prodrug and AlexaFluor™-488-prodrug. Sn-2 phosphatidylcholine prodrugs delivered by hemifusion of nanoparticle and cell phospholipid membranes functioned as phosphatidylcholine mimics, circumventing the challenges of endosome sequestration and release. Phosphatidylcholine prodrugs in the outer cell membrane leaflet translocated to the inner membrane leaflet by ATP-dependent and ATP-independent mechanisms and distributed broadly within the cytosolic membranes over the next 12 h. A portion of the phosphatidylcholine prodrug populated vesicle membranes trafficked to the perinuclear Golgi/ER region, where the drug was enzymatically liberated and activated. Native doxorubicin entered the cells, passed rapidly to the nucleus, and bound to dsDNA, whereas DOX was first enzymatically liberated from DOX-prodrug within the cytosol, particularly in the perinuclear region, before binding nuclear dsDNA. Much of DOX-prodrug was initially retained within intracellular membranes. In vitro anti-proliferation effectiveness of the two drug delivery approaches was equivalent at 48 h, suggesting that residual intracellular DOX-prodrug may constitute a slow-release drug reservoir that enhances effectiveness. We have demonstrated that Sn-2 phosphatidylcholine prodrugs function as phosphatidylcholine mimics following reported pathways of phosphatidylcholine distribution and metabolism. Drug complexed to the Sn-2 fatty acid is enzymatically liberated and reactivated over many hours, which may enhance efficacy overtime.
Collapse
Affiliation(s)
- Dolonchampa Maji
- Optical Radiology Lab, Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Jin Lu
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University of Buffalo, Buffalo, NY 14203
| | - Anne H Schmieder
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Grace Cui
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoxia Yang
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Matthew D Lew
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Samuel Achilefu
- Optical Radiology Lab, Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Gregory M Lanza
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA.,Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
11
|
Li Y, He D, Tu J, Wang R, Zu C, Chen Y, Yang W, Shi D, Webster TJ, Shen Y. The comparative effect of wrapping solid gold nanoparticles and hollow gold nanoparticles with doxorubicin-loaded thermosensitive liposomes for cancer thermo-chemotherapy. NANOSCALE 2018; 10:8628-8641. [PMID: 29697100 DOI: 10.1039/c7nr09083h] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Since conventional chemotherapy is a systemic treatment that affects the body globally and will not concentrate inside the tumor, it causes adverse side effects to patients. In this study, doxorubicin (DOX) together with solid gold nanoparticles (GNPs) or hollow gold nanoparticles (HGNPs), respectively, is loaded inside thermosensitive liposomes (GNPs&DOX-TLs and HGNPs&DOX-TLs), where the GNPs and HGNPs act as a "nanoswitch" for killing tumor cells directly by hyperthermia and triggering DOX release from TLs in the tumor quickly by near infrared laser (NIR) illumination. In addition, this study investigated the photothermal transformation ability, NIR triggered drug release behavior, and the intracellular uptake and cytotoxicity of breast tumor cells and the thermo-chemotherapy mediated by the co-delivery of GNPs&DOX-TLs and HGNPs&DOX-TLs. GNPs and HGNPs had very different light-to-heat transduction efficiencies, while the hollow HGNPs had the advantage of NIR surface plasmon tunability, resulting in the photothermal ablation of tumors with 800 nm light penetration in tissue. The prepared HGNPs&DOX-TLs exhibited a spherical shape with a diameter of 190 nm and a ξ potential of -29 mV, which were steadily dispersed for at least one month. The co-encapsulated DOX was released under hyperthermia caused by NIR-responsive HGNPs and the local drug concentration increased along with the disintegration of the liposomal membrane. This co-delivery of HGNPs&DOX-TLs produced a synergistic cytotoxicity response, thereby enhancing anticancer efficacy 8-fold and increasing the survival time compared to GNPs&DOX-TLs. This work suggested that the co-delivery of HGNPs&DOX-TLs followed by burst-release of DOX using NIR-responsive HGNPs sensitized cancer cells to the chemotherapeutic compound, which provided a novel concept for the combination strategy of chemotherapy and photothermal therapy. These results suggest that the markedly improved therapeutic efficacy and decreased systemic toxicity of the NPs presented in this study hold significant potential for future cancer treatment.
Collapse
Affiliation(s)
- Yanan Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Mumtaz Virk M, Reimhult E. Phospholipase A 2-Induced Degradation and Release from Lipid-Containing Polymersomes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:395-405. [PMID: 29231739 DOI: 10.1021/acs.langmuir.7b03893] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Hybrid vesicles, comprising blends of amphiphilic block copolymers and phospholipids, have attracted significant attention recently because of their unique combination of chemical and physical properties. We report a method to make unilamellar hybrid vesicles with diameters of 100 nm by mixing polybutadiene-block-poly(ethylene oxide) and phosphocholine lipids using a combination of solvent inversion and sonication. We show that homogeneous hybrid vesicles are formed when one component is a minor fraction. At compositions with balanced mass fractions, separate populations of similarly sized pure liposomes and hybrid vesicles are indicated. We investigate the release kinetics of calcein encapsulated in the lumen as hybrid large and giant unilamellar vesicles (LUVs and GUVs) of different compositions are exposed to phospholipase A2 (PLA2). PLA2 hydrolyzes lipids, which leads to dissolution of lipid domains and provides a trigger for the release of calcein as pores are formed. We demonstrate that depending on the polymer mole fraction, block copolymers can either protect or boost the rate of lipid degradation and thereby the release rate from nanoscale hybrid vesicles. Strong indications of lipid phase separation into nanoscale domains in LUVs are observed. Most importantly, hybrid GUV with lipids in the fluid phase release calcein slowly as lipids in the liquid-disordered phase do not phase-separate, but they show the fastest release of all blends as LUVs. This indicates phase separation on the nanoscale in contrast to on the microscale, but it also indicates retained high mobility of lipids between the nanoscale domains, which is absent for lipids in the gel phase. Our results demonstrate several ways in which nanoscale hybrid vesicles can and should be optimized for PLA2-triggered release of water-soluble compounds.
Collapse
Affiliation(s)
- Mudassar Mumtaz Virk
- Institute for Biologically Inspired Materials, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna , Muthgasse 11, 1190 Vienna, Austria
| | - Erik Reimhult
- Institute for Biologically Inspired Materials, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna , Muthgasse 11, 1190 Vienna, Austria
| |
Collapse
|
13
|
Cobo Solis AK, Correa NM, Molina PG. Determination of Benzyl-hexadecyldimethylammonium 1,4-Bis(2-ethylhexyl)sulfosuccinate Vesicle Permeability by Using Square Wave Voltammetry and an Enzymatic Reaction. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:12080-12086. [PMID: 28992699 DOI: 10.1021/acs.langmuir.7b03001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This report describes the studies performed to determine the permeability coefficient value (P) of 1-naphthyl phosphate (1-NP) through the benzyl-hexadecyldimethylammonium 1,4-bis(2-ethylhexyl)sulfosuccinate (AOT-BHD) vesicle bilayer. 1-NP was added in the external phase and must cross the bilayer of the vesicle to react with the encapsulated enzyme (alkaline phosphatase) to yield 1-naphtholate (NPh-), the product of the enzymatic hydrolysis. This product is electrochemically detected, at basic pH value, by a square wave voltammetry technique, which can be a good alternative over the spectroscopic one, to measure the vesicle solutions because scattering (due to its turbidity) does not make any influence in the electrochemical signal. The experimental data allow us to propose a mathematical model, and a value of P = (1.00 ± 0.15) × 10-9 cm s-1 was obtained. Also, a value of P = (2.0 ± 0.5) × 10-9 cm s-1 was found by using an independent technique, ultraviolet-visible spectroscopy, for comparison. It is evident that the P values obtained from both the techniques are comparable (within the experimental error of both techniques) under the same experimental conditions. This study constitutes the first report of the 1-NP permeability determination in this new vesicle. We want to highlight the importance of the introduction of a new method and the electrochemical response of the product generated through an enzymatic reaction that occurs in the inner aqueous phase of the vesicle, where the enzyme is placed.
Collapse
Affiliation(s)
- Airam K Cobo Solis
- Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto , Agencia Postal No. 3, 5800 Río Cuarto, Argentina
| | - N Mariano Correa
- Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto , Agencia Postal No. 3, 5800 Río Cuarto, Argentina
| | - Patricia G Molina
- Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto , Agencia Postal No. 3, 5800 Río Cuarto, Argentina
| |
Collapse
|
14
|
Sum R, Swaminathan M, Rastogi SK, Piloto O, Cheong I. Beta-Hemolytic Bacteria Selectively Trigger Liposome Lysis, Enabling Rapid and Accurate Pathogen Detection. ACS Sens 2017; 2:1441-1451. [PMID: 28929742 DOI: 10.1021/acssensors.7b00333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
For more than a century, blood agar plates have been the only test for beta-hemolysis. Although blood agar cultures are highly predictive for bacterial pathogens, they are too slow to yield actionable information. Here, we show that beta-hemolytic pathogens are able to lyse and release fluorophores encapsulated in sterically stabilized liposomes whereas alpha and gamma-hemolytic bacteria have no effect. By analyzing fluorescence kinetics, beta-hemolytic colonies cultured on agar could be distinguished in real time with 100% accuracy within 6 h. Additionally, end point analysis based on fluorescence intensity and machine-extracted textural features could discriminate between beta-hemolytic and cocultured control colonies with 99% accuracy. In broth cultures, beta-hemolytic bacteria were detectable in under an hour while control bacteria remained negative even the next day. This strategy, called beta-hemolysis triggered-release assay (BETA) has the potential to enable the same-day detection of beta-hemolysis with single-cell sensitivity and high accuracy.
Collapse
Affiliation(s)
- Rongji Sum
- Department
of Molecular Pathogenesis, Temasek Life Sciences Laboratory, Singapore 117604, Singapore
| | - Muthukaruppan Swaminathan
- Department
of Molecular Pathogenesis, Temasek Life Sciences Laboratory, Singapore 117604, Singapore
- Department
of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| | - Sahil Kumar Rastogi
- Department
of Molecular Pathogenesis, Temasek Life Sciences Laboratory, Singapore 117604, Singapore
| | | | - Ian Cheong
- Department
of Molecular Pathogenesis, Temasek Life Sciences Laboratory, Singapore 117604, Singapore
- Department
of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| |
Collapse
|
15
|
Solomon D, Gupta N, Mulla NS, Shukla S, Guerrero YA, Gupta V. Role of In Vitro Release Methods in Liposomal Formulation Development: Challenges and Regulatory Perspective. AAPS JOURNAL 2017; 19:1669-1681. [PMID: 28924630 DOI: 10.1208/s12248-017-0142-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/28/2017] [Indexed: 01/20/2023]
Abstract
In the past few years, measurement of drug release from pharmaceutical dosage forms has been a focus of extensive research because the release profile obtained in vitro can give an indication of the drug's performance in vivo. Currently, there are no compendial in vitro release methods designed for liposomes owing to a range of experimental challenges, which has created a major hurdle for both development and regulatory acceptance of liposome-based drug products. In this paper, we review the current techniques that are most often used to assess in vitro drug release from liposomal products; these include the membrane diffusion techniques (dialysis, reverse dialysis, fractional dialysis, and microdialysis), the sample-and-separate approach, the in situ method, the continuous flow, and the modified United States Pharmacopeia methods (USP I and USP IV). We discuss the principles behind each of the methods and the criteria that assist in choosing the most appropriate method for studying drug release from a liposomal formulation. Also, we have included information concerning the current regulatory requirements for liposomal drug products in the United States and in Europe. In light of increasing costs of preclinical and clinical trials, applying a reliable in vitro release method could serve as a proxy to expensive in vivo bioavailability studies. Graphical Abstract Appropriate in-vitro drug release test from liposomal products is important to predict the in-vivo performance.
Collapse
Affiliation(s)
- Deepak Solomon
- Research and Development, Neofluidics LLC, 6650 Lusk Blvd, Suite B102, San Diego, California, 92121, USA
| | - Nilesh Gupta
- Research and Development, Neofluidics LLC, 6650 Lusk Blvd, Suite B102, San Diego, California, 92121, USA
| | - Nihal S Mulla
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Drake University, 2507 University Ave, Des Moines, Iowa, 50311, USA
| | - Snehal Shukla
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, St. Albert Hall, B14, Queens, New York, 11439, USA
| | - Yadir A Guerrero
- Research and Development, Neofluidics LLC, 6650 Lusk Blvd, Suite B102, San Diego, California, 92121, USA
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, St. Albert Hall, B14, Queens, New York, 11439, USA.
| |
Collapse
|
16
|
Østrem RG, Parhamifar L, Pourhassan H, Clergeaud G, Nielsen OL, Kjær A, Hansen AE, Andresen TL. Secretory phospholipase A 2 responsive liposomes exhibit a potent anti-neoplastic effect in vitro, but induce unforeseen severe toxicity in vivo. J Control Release 2017; 262:212-221. [PMID: 28754610 DOI: 10.1016/j.jconrel.2017.07.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/30/2017] [Accepted: 07/24/2017] [Indexed: 01/22/2023]
Abstract
The clinical use of liposomal drug delivery vehicles is often hindered by insufficient drug release. Here we present the rational design of liposomes optimized for secretory phospholipase A2 (sPLA2) triggered drug release, and test their utility in vitro and in vivo. We hypothesized that by adjusting the level of cholesterol in anionic, unsaturated liposomes we could tune the enzyme specificity based on membrane fluidity, thus obtaining liposomes with an improved therapeutic outcome and reduced side effects. Cholesterol is generally important as a component in the membranes of liposome drug delivery systems due to its stabilizing effects in vivo. The incorporation of cholesterol in sPLA2 sensitive liposomes has not previously been possible due to reduced sPLA2 activity. However, in the present work we solved this challenge by optimizing membrane fluidity. In vitro release studies revealed enzyme specific drug release. Treatment of two different cancer cell lines with liposomal oxaliplatin revealed efficient growth inhibition compared to that of clinically used stealth liposomes. The in vivo therapeutic effect was evaluated in nude NMRI mice using the sPLA2 secreting mammary carcinoma cell line MT-3. Three days after first treatment all mice having received the novel sPLA2 sensitive liposome formulation were euthanized due to severe systemic toxicity. Thus the present study demonstrates that great caution should be implemented when utilizing sPLA2 sensitive liposomes and that the real utility can only be disclosed in vivo. The present studies have clinical implications, as sPLA2 sensitive formulations are currently undergoing clinical trials (LiPlaCis®).
Collapse
Affiliation(s)
- Ragnhild Garborg Østrem
- Technical University of Denmark, Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Produktionstorvet, 2800 Kgs. Lyngby, Denmark
| | - Ladan Parhamifar
- Technical University of Denmark, Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Produktionstorvet, 2800 Kgs. Lyngby, Denmark
| | - Houman Pourhassan
- Technical University of Denmark, Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Produktionstorvet, 2800 Kgs. Lyngby, Denmark
| | - Gael Clergeaud
- Technical University of Denmark, Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Produktionstorvet, 2800 Kgs. Lyngby, Denmark
| | - Ole Lerberg Nielsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 3, 1870 Frederiksberg C, Denmark
| | - Andreas Kjær
- Cluster for Molecular Imaging, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anders Elias Hansen
- Technical University of Denmark, Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Produktionstorvet, 2800 Kgs. Lyngby, Denmark; Cluster for Molecular Imaging, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Thomas Lars Andresen
- Technical University of Denmark, Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Produktionstorvet, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
17
|
Imamura R, Murata N, Shimanouchi T, Yamashita K, Fukuzawa M, Noda M. A Label-Free Fluorescent Array Sensor Utilizing Liposome Encapsulating Calcein for Discriminating Target Proteins by Principal Component Analysis. SENSORS (BASEL, SWITZERLAND) 2017; 17:E1630. [PMID: 28714873 PMCID: PMC5539792 DOI: 10.3390/s17071630] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 01/10/2023]
Abstract
A new fluorescent arrayed biosensor has been developed to discriminate species and concentrations of target proteins by using plural different phospholipid liposome species encapsulating fluorescent molecules, utilizing differences in permeation of the fluorescent molecules through the membrane to modulate liposome-target protein interactions. This approach proposes a basically new label-free fluorescent sensor, compared with the common technique of developed fluorescent array sensors with labeling. We have confirmed a high output intensity of fluorescence emission related to characteristics of the fluorescent molecules dependent on their concentrations when they leak from inside the liposomes through the perturbed lipid membrane. After taking an array image of the fluorescence emission from the sensor using a CMOS imager, the output intensities of the fluorescence were analyzed by a principal component analysis (PCA) statistical method. It is found from PCA plots that different protein species with several concentrations were successfully discriminated by using the different lipid membranes with high cumulative contribution ratio. We also confirmed that the accuracy of the discrimination by the array sensor with a single shot is higher than that of a single sensor with multiple shots.
Collapse
Affiliation(s)
- Ryota Imamura
- Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| | - Naoki Murata
- Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| | - Toshinori Shimanouchi
- Graduate School of Environmental and Life Science, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Kaoru Yamashita
- Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| | - Masayuki Fukuzawa
- Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| | - Minoru Noda
- Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| |
Collapse
|
18
|
Fouladi F, Steffen KJ, Mallik S. Enzyme-Responsive Liposomes for the Delivery of Anticancer Drugs. Bioconjug Chem 2017; 28:857-868. [PMID: 28201868 DOI: 10.1021/acs.bioconjchem.6b00736] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Liposomes are nanocarriers that deliver the payloads at the target site, leading to therapeutic drug concentrations at the diseased site and reduced toxic effects in healthy tissues. Several approaches have been used to enhance the ability of the nanocarrier to target the specific tissues, including ligand-targeted liposomes and stimuli-responsive liposomes. Ligand-targeted liposomes exhibit higher uptake by the target tissue due to the targeting ligand attached to the surface, while the stimuli-responsive liposomes do not release their cargo unless they expose to an endogenous or exogenous stimulant at the target site. In this review, we mainly focus on the liposomes that are responsive to pathologically increased levels of enzymes at the target site. Enzyme-responsive liposomes release their cargo upon contact with the enzyme through several destabilization mechanisms: (1) structural perturbation in the lipid bilayer, (2) removal of a shielding polymer from the surface and increased cellular uptake, (3) cleavage of a lipopeptide or lipopolymer incorporated in the bilayer, and (4) activation of a prodrug in the liposomes.
Collapse
Affiliation(s)
- Farnaz Fouladi
- Department of Pharmaceutical Sciences, North Dakota State University , Fargo, North Dakota 58108, United States
| | - Kristine J Steffen
- Department of Pharmaceutical Sciences, North Dakota State University , Fargo, North Dakota 58108, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University , Fargo, North Dakota 58108, United States
| |
Collapse
|
19
|
Mizukami S, Kashibe M, Matsumoto K, Hori Y, Kikuchi K. Enzyme-triggered compound release using functionalized antimicrobial peptide derivatives. Chem Sci 2017; 8:3047-3053. [PMID: 28451373 PMCID: PMC5380883 DOI: 10.1039/c6sc04435b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/07/2017] [Indexed: 12/03/2022] Open
Abstract
Two strategies have been proposed to develop enzyme-triggered compound release systems.
Controlled release is one of the key technologies for medical innovation, and many stimulus-responsive nanocarriers have been developed to utilize this technology. Enzyme activity is one of the most useful stimuli, because many enzymes are specifically activated in diseased tissues. However, controlled release stimulated by enzyme activity has not been frequently reported. One of the reasons for this is the lack of versatility of carriers. Most of the reported stimulus-responsive systems involve a sophisticated design and a complicated process for the synthesis of stimulus-responsive nanocarrier components. The purpose of this study was to develop versatile controlled release systems triggered by various stimuli, including enzyme activity, without modifying the nanocarrier components. We developed two controlled release systems, both of which comprised a liposome as the nanocarrier and a membrane-damaging peptide, temporin L (TL), and its derivatives as the release-controllers. One system utilized branched peptides for proteases, and the other utilized phosphopeptides for phosphatases. In our systems, the target enzymes converted the non-membrane-damaging TL derivatives into membrane-damaging peptides and released the liposome inclusion. We demonstrated the use of our antimicrobial peptide-based controlled release systems for different enzymes and showed the promise of this technology as a novel theranostic tool.
Collapse
Affiliation(s)
- Shin Mizukami
- Institute of Multidisciplinary Research for Advanced Materials , Tohoku University , 2-1-1C Katahira, Aoba-ku , Sendai , Miyagi 980-8577 , Japan .
| | - Masayoshi Kashibe
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan .
| | - Kengo Matsumoto
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan .
| | - Yuichiro Hori
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan . .,Immunology Frontier Research Center , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan
| | - Kazuya Kikuchi
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan . .,Immunology Frontier Research Center , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan
| |
Collapse
|
20
|
Fabrication of liposomal doxorubicin exhibiting ultrasensitivity against phospholipase A 2 for efficient pulmonary drug delivery to lung cancers. Int J Pharm 2017; 517:35-41. [DOI: 10.1016/j.ijpharm.2016.11.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
|
21
|
Multi-functional vesicles for cancer therapy: The ultimate magic bullet. Colloids Surf B Biointerfaces 2016; 147:161-171. [DOI: 10.1016/j.colsurfb.2016.07.060] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 01/22/2023]
|
22
|
Holtkamp H, Grabmann G, Hartinger CG. Electrophoretic separation techniques and their hyphenation to mass spectrometry in biological inorganic chemistry. Electrophoresis 2016; 37:959-72. [DOI: 10.1002/elps.201500502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Hannah Holtkamp
- School of Chemical Sciences; University of Auckland; Auckland New Zealand
| | - Gerlinde Grabmann
- School of Chemical Sciences; University of Auckland; Auckland New Zealand
- Institute of Inorganic Chemistry; University of Vienna; Vienna Austria
| | | |
Collapse
|
23
|
Sun Y, Ji Y, Yu H, Wang D, Cao M, Wang J. Near-infrared light-sensitive liposomes for controlled release. RSC Adv 2016. [DOI: 10.1039/c6ra18702a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A 6-bromo-7-hydroxy-4-hydroxymethylcoumarin containing amphiphilic lipid was synthesized and applied as a near-infrared light triggered controlled release system.
Collapse
Affiliation(s)
- Yawei Sun
- Centre for Bioengineering and Biotechnology
- China University of Petroleum (East China)
- China
| | - Yanyun Ji
- Centre for Bioengineering and Biotechnology
- China University of Petroleum (East China)
- China
| | - Haiyan Yu
- Centre for Bioengineering and Biotechnology
- China University of Petroleum (East China)
- China
| | - Dong Wang
- Centre for Bioengineering and Biotechnology
- China University of Petroleum (East China)
- China
| | - Meiwen Cao
- Centre for Bioengineering and Biotechnology
- China University of Petroleum (East China)
- China
| | - Jiqian Wang
- Centre for Bioengineering and Biotechnology
- China University of Petroleum (East China)
- China
| |
Collapse
|
24
|
Luna MA, Silber JJ, Sereno L, Correa NM, Moyano F. Determining the substrate permeability through the bilayer of large unilamellar vesicles of DOPC. A kinetic study. RSC Adv 2016. [DOI: 10.1039/c6ra12847e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In this work we determine the permeability of DOPC vesicles in the presence of different cholesterol contents, by using the enzymatic hydrolysis of N-benzoyl-l-tyrosine p-nitroanilide catalyzed by α-chymotrypsin.
Collapse
Affiliation(s)
- Maria Alejandra Luna
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - Juana J. Silber
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - Leonides Sereno
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - N. Mariano Correa
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - Fernando Moyano
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| |
Collapse
|
25
|
Aoki I, Yoneyama M, Hirose J, Minemoto Y, Koyama T, Kokuryo D, Bakalova R, Murayama S, Saga T, Aoshima S, Ishizaka Y, Kono K. Thermoactivatable polymer-grafted liposomes for low-invasive image-guided chemotherapy. Transl Res 2015; 166:660-673.e1. [PMID: 26303887 DOI: 10.1016/j.trsl.2015.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 07/02/2015] [Accepted: 07/29/2015] [Indexed: 11/28/2022]
Abstract
The objective of this study was to develop a thermotriggered, polymer-based liposomal drug carrier with an activatable magnetic resonance imaging (MRI) contrast property for monitoring the release of substances and for localized tumor therapy. The multimodal thermoactivatable polymer-grafted liposomes (MTPLs) were tested to investigate whether the accumulation of MTPLs in colon-26 grafted tumors could be visualized in vivo using MRI and optical imaging, whether MTPLs induce signal enhancement, reflecting the release of their contents, after triggering by short-term heating (42.5°C for 10 minutes) 9 hours after MTPL administration (late-phase triggering), and whether MTPLs can provide a sufficient antitumor effect. The imaging and therapeutic properties of MTPLs were tested both in vitro and in vivo (BALB/c nude mice: heated group with MTPLs (n = 5), nonheated group with MTPLs (n = 5), heated group with doxorubicin-free MTPLs (n = 5), nonheated group with manganese-free MTPLs (n = 5), and kinetics observation group (n = 3); N = 23). Through in vivo MRI and fluorescent imaging, the MTPLs were shown to have significantly accumulated in the grafted colon-26 tumors 8 hours after administration. Delayed thermotriggering (9 hours after administration) caused MR signal enhancement, reflecting the release of their contents, after a short exposure to tolerable heat. In addition, significant antitumor effects were observed after treatment. The proposed polymer-based activatable MTPLs with a "delayed thermotrigger" provide a promising technology for cancer theranostics that allows minimal adverse effects and rapid interactive therapy.
Collapse
Affiliation(s)
- Ichio Aoki
- Molecular Imaging Center, National Institute of Radiological Sciences (NIRS), Chiba, Japan.
| | - Misao Yoneyama
- Molecular Imaging Center, National Institute of Radiological Sciences (NIRS), Chiba, Japan
| | - Jun Hirose
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Sakai, Japan
| | - Yuzuru Minemoto
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takayoshi Koyama
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Daisuke Kokuryo
- Molecular Imaging Center, National Institute of Radiological Sciences (NIRS), Chiba, Japan
| | - Rumiana Bakalova
- Molecular Imaging Center, National Institute of Radiological Sciences (NIRS), Chiba, Japan
| | - Shuhei Murayama
- Molecular Imaging Center, National Institute of Radiological Sciences (NIRS), Chiba, Japan
| | - Tsuneo Saga
- Molecular Imaging Center, National Institute of Radiological Sciences (NIRS), Chiba, Japan
| | - Sadahito Aoshima
- Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Yukihito Ishizaka
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kenji Kono
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Sakai, Japan
| |
Collapse
|
26
|
Dual-targeting nanocarrier system based on thermosensitive liposomes and gold nanorods for cancer thermo-chemotherapy. J Control Release 2015; 215:91-100. [DOI: 10.1016/j.jconrel.2015.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 07/02/2015] [Accepted: 08/02/2015] [Indexed: 01/28/2023]
|
27
|
Interaction kinetics of serum proteins with liposomes and their effect on phospholipase-induced liposomal drug release. Int J Pharm 2015; 495:827-39. [PMID: 26410758 DOI: 10.1016/j.ijpharm.2015.09.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/10/2015] [Accepted: 09/23/2015] [Indexed: 12/19/2022]
Abstract
We used surface plasmon resonance (SPR) to measure the affinity and kinetics of the interaction between serum proteins and both conventional and PEGylated liposomes. The effect of the interactions on secretory phospholipase A2 (sPLA2)-induced release of a model drug from liposomes was also assessed. SPR analysis of 12 serum proteins revealed that the mode of interaction between serum proteins and liposomes greatly varies depending on the type of protein. For example, albumin bound to liposomes at slower association/dissociation rates with higher affinity and prevented sPLA2-induced drug release from PEGylated liposomes. Conversely, fibronectin bound at faster association/dissociation rates with lower affinity and demonstrated little impact on the drug release. These results indicate that the effect of serum proteins on sPLA2 phospholipid hydrolysis varies with the mode of interaction between proteins and liposomes. Understanding how the proteins interact with liposomes and impact sPLA2 phospholipid hydrolysis should aid the rational design of therapeutic liposomal formulations.
Collapse
|
28
|
Pan D, Pham CTN, Weilbaecher KN, Tomasson MH, Wickline SA, Lanza GM. Contact-facilitated drug delivery with Sn2 lipase labile prodrugs optimize targeted lipid nanoparticle drug delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:85-106. [PMID: 26296541 PMCID: PMC4709477 DOI: 10.1002/wnan.1355] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/18/2015] [Indexed: 01/10/2023]
Abstract
Sn2 lipase labile phospholipid prodrugs in conjunction with contact-facilitated drug delivery offer an important advancement in Nanomedicine. Many drugs incorporated into nanosystems, targeted or not, are substantially lost during circulation to the target. However, favorably altering the pharmacokinetics and volume of distribution of systemic drug delivery can offer greater efficacy with lower toxicity, leading to new prolonged-release nanoexcipients. However, the concept of achieving Paul Erhlich's inspired vision of a 'magic bullet' to treat disease has been largely unrealized due to unstable nanomedicines, nanosystems achieving low drug delivery to target cells, poor intracellular bioavailability of endocytosed nanoparticle payloads, and the substantial biological barriers of extravascular particle penetration into pathological sites. As shown here, Sn2 phospholipid prodrugs in conjunction with contact-facilitated drug delivery prevent premature drug diffusional loss during circulation and increase target cell bioavailability. The Sn2 phospholipid prodrug approach applies equally well for vascular constrained lipid-encapsulated particles and micelles the size of proteins that penetrate through naturally fenestrated endothelium in the bone marrow or thin-walled venules of an inflamed microcirculation. At one time Nanomedicine was considered a 'Grail Quest' by its loyal opposition and even many in the field adsorbing the pains of a long-learning curve about human biology and particles. However, Nanomedicine with innovations like Sn2 phospholipid prodrugs has finally made 'made the turn' toward meaningful translational success.
Collapse
Affiliation(s)
- Dipanjan Pan
- Departments of Bioengineering, Materials Science and Engineering, Beckman Institute, University of Illinois, Urbana-Champaign, IL, USA
| | - Christine T N Pham
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine N Weilbaecher
- Division of Oncology, Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Michael H Tomasson
- Division of Oncology, Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Samuel A Wickline
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory M Lanza
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
29
|
Hansen AH, Mouritsen OG, Arouri A. Enzymatic action of phospholipase A2 on liposomal drug delivery systems. Int J Pharm 2015; 491:49-57. [DOI: 10.1016/j.ijpharm.2015.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022]
|
30
|
Nguyen T, Tekrony A, Yaehne K, Cramb DT. Designing a better theranostic nanocarrier for cancer applications. Nanomedicine (Lond) 2015; 9:2371-86. [PMID: 25413855 DOI: 10.2217/nnm.14.110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanocarriers show incredible potential in theranostic applications as they offer diagnostic capabilities along with the ability to encapsulate and protect drugs from degradation, be functionalized with targeting moieties and be designed with controlled release mechanisms. Most clinically approved nanocarrier drugs are liposomal formulations. As such, considerable research has been directed towards designing liposomal carriers that can release their payloads via exogenous or endogenous triggers. For triggered release to effectively increase drug bioavailability, nanocarriers must first accumulate at the tumor site via the enhanced retention and permeability effect. It has been demonstrated in the chicken embryo chorioallantoic membrane and murine xenografted models that nanoparticle surface charge and geometry, with respect to vascular endothelium fenestration size, drive this accumulation in angiogenic tissue.
Collapse
Affiliation(s)
- Trinh Nguyen
- University of Calgary, Department of Chemistry, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | | | | | | |
Collapse
|
31
|
Pan D, Kim B, Hu G, Gupta DS, Senpan A, Yang X, Schmieder A, Swain C, Wickline SA, Tomasson MH, Lanza GM. A strategy for combating melanoma with oncogenic c-Myc inhibitors and targeted nanotherapy. Nanomedicine (Lond) 2015; 10:241-51. [PMID: 25600969 PMCID: PMC4665613 DOI: 10.2217/nnm.14.101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS The activity of the transcription factor c-Myc is dependent upon heterodimerization with Max to control target gene transcription. Small-molecule inhibitors of c-Myc-Max have exhibited low potency and poor water solubility and are therefore unsuitable for in vivo application. We hypothesized that a nanomedicine approach incorporating a cryptic c-Myc inhibitor prodrug could be delivered and enzymatically released in order to effectively inhibit melanoma. MATERIALS & METHODS An Sn-2 lipase-labile Myc inhibitor prodrug was synthesized and included in two αvβ3-targeted nanoparticle platforms (20 and 200 nm). The inherent antiproliferate potency was compared with the lipid-free compound using human and mouse melanoma cell lines. RESULTS & CONCLUSION These data demonstrate for the first time a successful nanodelivery of c-Myc inhibitors and their potential use to prevent melanoma.
Collapse
Affiliation(s)
- Dipanjan Pan
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Biomedical Research Center, Office 3304, 3rd Floor, Mills Breast Cancer Institute & Carle Foundation Hospital, 502 N. Busey, Urbana, IL 61801, USA
| | - Benjamin Kim
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Grace Hu
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Deepti Sood Gupta
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Angana Senpan
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Xiaoxia Yang
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Anne Schmieder
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Corban Swain
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Samuel A Wickline
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Michael H Tomasson
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Gregory M Lanza
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| |
Collapse
|
32
|
d'Arcy R, Tirelli N. Fishing for fire: strategies for biological targeting and criteria for material design in anti-inflammatory therapies. POLYM ADVAN TECHNOL 2014. [DOI: 10.1002/pat.3264] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Richard d'Arcy
- School of Medicine/Institute of Inflammation and Repair; University of Manchester; Manchester M13 9PT UK
| | - Nicola Tirelli
- School of Medicine/Institute of Inflammation and Repair; University of Manchester; Manchester M13 9PT UK
- School of Materials; University of Manchester; Manchester M13 9PT UK
| |
Collapse
|
33
|
Yan X, An X. Multifunctional polydiacetylene-liposome with controlled release and fluorescence tracing. RSC Adv 2014. [DOI: 10.1039/c4ra02414a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A multifunctional polydiacetylene-liposome (PDA-liposome) was prepared by self-assembly, which can be characterized with controlled drug release and fluorescence tracing.
Collapse
Affiliation(s)
- Xiaojuan Yan
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai, China
| | - Xueqin An
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai, China
| |
Collapse
|
34
|
Kang ML, Im GI. Drug delivery systems for intra-articular treatment of osteoarthritis. Expert Opin Drug Deliv 2013; 11:269-82. [PMID: 24308404 DOI: 10.1517/17425247.2014.867325] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Intra-articular (IA) drug delivery is very useful in the treatment of osteoarthritis (OA), the most common chronic joint affliction. However, the therapeutic effect of IA administration depends mostly on the efficacy of drug delivery. AREAS COVERED The present article reviews the current status of IA therapy for OA treatment as well as its rationale. Outlines of drug delivery parameters such as release profile, retention time, distribution, size and transport that influence the drug's biological performance in the joints are summarized. New delivery systems, currently under investigation, including liposome, nanoparticle, microparticle and hydrogel formulations are introduced. Functionalized drug delivery systems by targeting and thermoresponsiveness that are being investigated for OA treatment via IA therapy are also addressed. EXPERT OPINION Several delivery systems, including liposome, microparticles, nanoparticles and hydrogels, have been investigated for the sustained drug delivery to the joints. These can be advanced by the use of functionalized drug delivery systems that can lead targeting to specific regions and thermoresponsiveness for prolonged drug release in the joints. Further advances will bring forth new biocompatible and biodegradable materials as a drug carrier or new combination regimens. Future innovations in this field should be directed toward the development of adapted delivery systems that can induce tissue regeneration in OA patients.
Collapse
Affiliation(s)
- Mi Lan Kang
- Dongguk University Ilsan Hospital, Department of Orthopedics , Goyang 410-773 , Korea +82 31 961 7315 ; +82 31 961 7314 ;
| | | |
Collapse
|
35
|
Mellal D, Zumbuehl A. Exit-strategies - smart ways to release phospholipid vesicle cargo. J Mater Chem B 2013; 2:247-252. [PMID: 32261503 DOI: 10.1039/c3tb21086c] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
This highlight describes recent trends in fundamental phospholipid research towards possible future drug delivery technology. In particular it focuses on synthetic phospholipids and their vesicular constructs and describes selected "smart" ways to release cargo from liposomes. Various chemical and physical release triggers are discussed such as temperature changes, application of ultrasound, enzyme degradation, changes in pH, redox reactions, photochemical reactions, as well as the effects of shear stress on vesicles.
Collapse
Affiliation(s)
- Denia Mellal
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland.
| | | |
Collapse
|
36
|
Arouri A, Hansen AH, Rasmussen TE, Mouritsen OG. Lipases, liposomes and lipid-prodrugs. Curr Opin Colloid Interface Sci 2013. [DOI: 10.1016/j.cocis.2013.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
37
|
Liu D, He C, Wang AZ, Lin W. Application of liposomal technologies for delivery of platinum analogs in oncology. Int J Nanomedicine 2013; 8:3309-19. [PMID: 24023517 PMCID: PMC3767488 DOI: 10.2147/ijn.s38354] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Platinum-based chemotherapy, such as cisplatin, oxaliplatin, and carboplatin, is one of the most widely utilized classes of cancer therapeutics. While highly effective, the clinical applications of platinum-based drugs are limited by their toxicity profiles as well as suboptimal pharmacokinetic properties. Therefore, one of the key research areas in oncology has been to develop novel platinum analog drugs and engineer new platinum drug formulations to improve the therapeutic ratio further. Such efforts have led to the development of platinum analogs including nedaplatin, heptaplatin, and lobaplatin. Moreover, reformulating platinum drugs using liposomes has resulted in the development of L-NDPP (Aroplatin™), SPI-77, Lipoplatin™, Lipoxal™, and LiPlaCis®. Liposomes possess several attractive biological activities, including biocompatibility, high drug loading, and improved pharmacokinetics, that are well suited for platinum drug delivery. In this review, we discuss the various platinum drugs and their delivery using liposome-based drug delivery vehicles. We compare and contrast the different liposome platforms as well as speculate on the future of platinum drug delivery research.
Collapse
Affiliation(s)
- Demin Liu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
38
|
Oude Blenke E, Mastrobattista E, Schiffelers RM. Strategies for triggered drug release from tumor targeted liposomes. Expert Opin Drug Deliv 2013; 10:1399-410. [DOI: 10.1517/17425247.2013.805742] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Adv Drug Deliv Rev 2013; 65:36-48. [PMID: 23036225 DOI: 10.1016/j.addr.2012.09.037] [Citation(s) in RCA: 2947] [Impact Index Per Article: 267.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/10/2012] [Accepted: 09/20/2012] [Indexed: 02/06/2023]
Abstract
The first closed bilayer phospholipid systems, called liposomes, were described in 1965 and soon were proposed as drug delivery systems. The pioneering work of countless liposome researchers over almost 5 decades led to the development of important technical advances such as remote drug loading, extrusion for homogeneous size, long-circulating (PEGylated) liposomes, triggered release liposomes, liposomes containing nucleic acid polymers, ligand-targeted liposomes and liposomes containing combinations of drugs. These advances have led to numerous clinical trials in such diverse areas as the delivery of anti-cancer, anti-fungal and antibiotic drugs, the delivery of gene medicines, and the delivery of anesthetics and anti-inflammatory drugs. A number of liposomes (lipidic nanoparticles) are on the market, and many more are in the pipeline. Lipidic nanoparticles are the first nanomedicine delivery system to make the transition from concept to clinical application, and they are now an established technology platform with considerable clinical acceptance. We can look forward to many more clinical products in the future.
Collapse
|
40
|
Membrane-perturbing effect of fatty acids and lysolipids. Prog Lipid Res 2013; 52:130-40. [DOI: 10.1016/j.plipres.2012.09.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 08/20/2012] [Accepted: 09/13/2012] [Indexed: 12/13/2022]
|
41
|
Schaefer JJ, Ma C, Harris JM. Confocal Raman microscopy probing of temperature-controlled release from individual, optically-trapped phospholipid vesicles. Anal Chem 2012; 84:9505-12. [PMID: 23043532 DOI: 10.1021/ac302346n] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Control of permeability of phospholipid vesicle (liposome) membranes is critical to their applications in analytical sensing, in fundamental studies of chemistry in small volumes, and in encapsulation and release of payloads for site-directed drug delivery. Applications of liposome formulations in drug delivery often take advantage of the enhanced permeability of phospholipid membranes at their gel-to-fluid phase transition, where the release of encapsulated molecules can be initiated by an increase in temperature. Despite numerous successful liposome formulations for encapsulation and release methods to study the kinetics, this process has been limited to investigations of bulk vesicle dispersions, which provide little or no information about the vesicle membrane structure and its relationship to the kinetics of trans-membrane transport. In this work, confocal Raman microscopy is adapted to study temperature-dependent release of a model compound, 3-nitrobenzene sulfonate (3-NBS), from individual optically trapped phospholipid vesicles, while simultaneously monitoring structural changes in the vesicle membrane reported by vibrational modes of phospholipid acyl chains and the local environment of the encapsulated compound. The confocal geometry allows efficient excitation and collection of Raman scattering from a single vesicle, while optical trapping allows more than hour-long observations of the same vesicle. With window factor analysis to resolve component spectra, temperature-controlled release of 3-NBS through vesicle membranes composed of pure 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) was measured and compared to transport through a lysolipid-containing membrane specifically formulated for efficient drug delivery.
Collapse
Affiliation(s)
- Jonathan J Schaefer
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | | | | |
Collapse
|
42
|
Pan D, Sanyal N, Schmieder AH, Senpan A, Kim B, Yang X, Hu G, Allen JS, Gross RW, Wickline SA, Lanza GM. Antiangiogenic nanotherapy with lipase-labile Sn-2 fumagillin prodrug. Nanomedicine (Lond) 2012; 7:1507-19. [PMID: 22709347 PMCID: PMC3498609 DOI: 10.2217/nnm.12.27] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The chemical instability of antiangiogenic fumagillin, combined with its poor retention during intravascular transit, requires an innovative solution for clinical translation. We hypothesized that an Sn-2 lipase-labile fumagillin prodrug, in combination with a contact-facilitated drug delivery mechanism, could be used to address these problems. METHODS α(v)β(3)-targeted and nontargeted nanoparticles with and without fumagillin in the prodrug or native forms were evaluated in vitro and in vivo in the Matrigel™ (BD Biosciences, CA, USA) plug model of angiogenesis in mice. RESULTS In vitro experiments demonstrated that the new fumagillin prodrug decreased viability at least as efficacious as the parent compound, on an equimolar basis. In the Matrigel mouse angiogenesis model, α(v)β(3)-fumagillin prodrug decreased angiogenesis as measured by MRI (3T), while the neovasculature was unaffected with the control nanoparticles. CONCLUSION The present approach resolved the previously intractable problems of drug instability and premature release in transit to target sites.
Collapse
Affiliation(s)
- Dipanjan Pan
- Division of Cardiology, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Mendoza MF, Hollabaugh NM, Hettiarachchi SU, McCarley RL. Human NAD(P)H:quinone oxidoreductase type I (hNQO1) activation of quinone propionic acid trigger groups. Biochemistry 2012; 51:8014-26. [PMID: 22989153 DOI: 10.1021/bi300760u] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
NAD(P)H:quinone oxidoreductase type I (NQO1) is a target enzyme for triggered delivery of drugs at inflamed tissue and tumor sites, particularly those that challenge traditional therapies. Prodrugs, macromolecules, and molecular assemblies possessing trigger groups that can be cleaved by environmental stimuli are vehicles with the potential to yield active drug only at prescribed sites. Furthermore, quinone propionic acids (QPAs) covalently attached to prodrugs or liposome surfaces can be removed by application of a reductive trigger stimulus, such as that from NQO1; their rates of reductive activation should be tunable via QPA structure. We explored in detail the recombinant human NAD(P)H:quinone oxidoreductase type I (rhNQO1)-catalyzed NADH reduction of a family of substituted QPAs and obtained high precision kinetic parameters. It is found that small changes in QPA structure-in particular, single atom and function group substitutions on the quinone ring at R(1)-lead to significant impacts on the Michaelis constant (K(m)), maximum velocity (V(max)), catalytic constant (k(cat)), and catalytic efficiency (k(cat)/K(m)). Molecular docking simulations demonstrate that alterations in QPA structure result in large changes in QPA alignment and placement with respect to the flavin isoalloxazine ring in the active site of rhNQO1; a qualitative relationship exists between the kinetic parameters and the depth of QPA penetration into the rhNQO1 active site. From a quantitative perspective, a very good correlation is observed between log(k(cat)/K(m)) and the molecular-docking-derived distance between the flavin hydride donor site and quinone hydride acceptor site in the QPAs, an observation that is in agreement with developing theories. The comprehensive kinetic and molecular modeling knowledge obtained for the interaction of recombinant human NQO1 with the quinone propionic acid analogues provides insight into the design and implementation of the QPA trigger groups for drug delivery applications.
Collapse
Affiliation(s)
- Maria F Mendoza
- Department of Chemistry, Louisiana State University, 232 Choppin Hall, Baton Rouge, Louisiana 70803-1804, USA
| | | | | | | |
Collapse
|
44
|
Zhou HF, Yan H, Senpan A, Wickline SA, Pan D, Lanza GM, Pham CTN. Suppression of inflammation in a mouse model of rheumatoid arthritis using targeted lipase-labile fumagillin prodrug nanoparticles. Biomaterials 2012; 33:8632-40. [PMID: 22922023 DOI: 10.1016/j.biomaterials.2012.08.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 08/01/2012] [Indexed: 12/22/2022]
Abstract
Nanoparticle-based therapeutics are emerging technologies that have the potential to greatly impact the treatment of many human diseases. However, drug instability and premature release from the nanoparticles during circulation currently preclude clinical translation. Herein, we use a lipase-labile (Sn 2) fumagillin prodrug platform coupled with a unique lipid surface-to-surface targeted delivery mechanism, termed contact-facilitated drug delivery, to counter the premature drug release and overcome the inherent photo-instability of fumagillin, an established anti-angiogenic agent. We show that α(v)β(3)-integrin targeted fumagillin prodrug nanoparticles, administered at 0.3 mg of fumagillin prodrug/kg of body weight suppress the clinical disease indices of KRN serum-mediated arthritis in a dose-dependent manner when compared to treatment with the control nanoparticles with no drug. This study demonstrates the effectiveness of this lipase-labile prodrug nanocarrier in a relevant preclinical model that approximates human rheumatoid arthritis. The lipase-labile prodrug paradigm offers a translatable approach that is broadly applicable to many targeted nanosystems and increases the translational potential of this platform for many diseases.
Collapse
Affiliation(s)
- Hui-Fang Zhou
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8045, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Joshi N, Kaviratna A, Banerjee R. Multi trigger responsive, surface active lipid nanovesicle aerosols for improved efficacy of paclitaxel in lung cancer. Integr Biol (Camb) 2012; 5:239-48. [DOI: 10.1039/c2ib20122d] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Nitin Joshi
- WRCBB, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India. Fax: +91-22 2572 3480; Tel: +91-22 2576 7868
| | - Anubhav Kaviratna
- WRCBB, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India. Fax: +91-22 2572 3480; Tel: +91-22 2576 7868
| | - Rinti Banerjee
- WRCBB, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India. Fax: +91-22 2572 3480; Tel: +91-22 2576 7868
| |
Collapse
|
46
|
Pedersen PJ, Viart HMF, Melander F, Andresen TL, Madsen R, Clausen MH. Synthesis of tocopheryl succinate phospholipid conjugates and monitoring of phospholipase A2 activity. Bioorg Med Chem 2012; 20:3972-8. [DOI: 10.1016/j.bmc.2012.05.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Revised: 04/30/2012] [Accepted: 05/11/2012] [Indexed: 01/20/2023]
|
47
|
Rosholm KR, Arouri A, Hansen PL, González-Pérez A, Mouritsen OG. Characterization of fluorinated catansomes: a promising vector in drug-delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2012; 28:2773-2781. [PMID: 22149538 DOI: 10.1021/la2039834] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Catansomes, which are vesicles prepared from mixtures of oppositely charged surfactants, have been suggested as effective alternatives to phospholipid vesicles, i.e., liposomes, in applications such as drug-delivery. This is mainly due to their enhanced chemical and physical stability as well as to their relatively easy preparation, which is an advantage for large-scale productions. In this study we have investigated catansomes prepared from a perfluorinated anionic surfactant (sodium perfluorooctanoate) premixed with a hydrogenated cationic surfactant (dodecyltrimethylammonium bromide or 1-dodecylpyridinium chloride). The aim was to gain insights into the physicochemical properties of these systems, such as size, stability, surface charge, and membrane morphology, which are essential for their use in drug-delivery applications. The catansomes were mostly unilamellar and 100-200 nm in size, and were stable for more than five months at room temperature. After loading the catansomes with the fluorescent marker calcein, they were found to exhibit an appreciable encapsulation efficiency and a low calcein leakage over time. The addition of fatty acids to calcein-loaded catansomes considerably promoted the release of calcein, and the rate and efficiency of calcein release were found to be proportional to the fatty acid concentration and chain length. Our results prove the feasibility of utilizing catansomes as drug-delivery vehicles as well as provide a means to efficiently release the encapsulated load.
Collapse
Affiliation(s)
- Kadla R Rosholm
- BioNano & NanoMedicine, Center of Nano-Science, University of Copenhagen, Universitetsparken 5, DK-2100 København Ø, Denmark
| | | | | | | | | |
Collapse
|
48
|
Jia Y, Joly H, Omri A. Characterization of the interaction between liposomal formulations and Pseudomonas aeruginosa. J Liposome Res 2012; 20:134-46. [PMID: 19831502 DOI: 10.3109/08982100903218892] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The interactions between three liposomal formulations and Pseudomonas aeruginosa cells were evaluated by a lipid mixing assay and electron paramagnetic resonance (EPR) spectroscopy. The effect of the bacteria on the liposomal phase characteristics, the release of the liposomes' content, and the uptake rate of gentamicin by bacteria were monitored as a function of time, using EPR spectroscopy. The [16-DSA uptake](Total) from DPPC (1,2-dipalmitoyl-sn-glycero-3-phosphocholine) liposomes reached 93 +/- 12% over a 3-hour assay period, of which 9% crossed the bacterial inner membrane. A small amount of 16-DSA uptake from DPPC/Chol (cholesterol) vesicles was found throughout the 3-hour period of time. Although DPPC/DMPG (dimyristoylphosphatidylglycerol) vesicles showed a smaller value of [16-DSA uptake](Total) with respect to that of DPPC vesicles, they appeared to be effective in disrupting the bacterial membrane, resulting in a greater accumulation of 16-DSA inside the inner membrane. Exposure to bacteria caused the DPPC/Chol, DPPC, and DPPC/DMPG formulations to release 4.6 +/- 1.5, 17.6 +/- 1.2, and 34 +/- 3.7% of their content, respectively. Time-dependent fluid regions were developed within the vesicles when mixed with bacteria, and their growth over time depended on liposomal formulations. Incubation of gentamicin with bacteria for 3 hours resulted in 87 +/- 3% of the drug crossing the bacterial inner membrane. In conclusion, interaction between the liposome drug carriers and the bacterial cells result in vesicle fusion, disruption of the bacterial membrane, release of the liposomal content in the close vicinity of the bacteria cells, and the subsequent intracellular uptake of the released liposomal content.
Collapse
Affiliation(s)
- Yimei Jia
- Laurentian University, Sudbury, Ontario, Canada
| | | | | |
Collapse
|
49
|
Nguyen TTTN, Østergaard J, Stürup S, Gammelgaard B. Investigation of a liposomal oxaliplatin drug formulation by capillary electrophoresis hyphenated to inductively coupled plasma mass spectrometry (CE-ICP-MS). Anal Bioanal Chem 2012; 402:2131-9. [DOI: 10.1007/s00216-011-5651-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 12/09/2011] [Accepted: 12/10/2011] [Indexed: 11/25/2022]
|
50
|
Jespersen H, Andersen JH, Ditzel HJ, Mouritsen OG. Lipids, curvature stress, and the action of lipid prodrugs: Free fatty acids and lysolipid enhancement of drug transport across liposomal membranes. Biochimie 2012; 94:2-10. [DOI: 10.1016/j.biochi.2011.07.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Accepted: 07/25/2011] [Indexed: 12/31/2022]
|